Buscar en
Open Respiratory Archives
Toda la web
Inicio Open Respiratory Archives GEMA 5.3. Spanish Guideline on the Management of Asthma
Journal Information
Vol. 5. Issue 4.
(October - December 2023)
Share
Share
Download PDF
More article options
Visits
4134
Vol. 5. Issue 4.
(October - December 2023)
Special article
Full text access
GEMA 5.3. Spanish Guideline on the Management of Asthma
GEMA 5.3. Guía Española para el Manejo del Asma
Visits
4134
Vicente Plaza Morala,
Corresponding author
vplaza@santpau.cat

Corresponding author.
, Isam Alobidb, Cesáreo Álvarez Rodríguezc, Marina Blanco Apariciod, Jorge Ferreirae, Gabriel Garcíaf, Antonio Gómez-Outesg, Noé Garín Escriváh, Fernando Gómez Ruizi, Antonio Hidalgo Requenaj, Javier Korta Muruak, Jesús Molina Parísl, Francisco Javier Pellegrini Belinchónm, Javier Plaza Zamoran, Manuel Praena Crespoo, Santiago Quirce Gancedop, José Sanz Ortegaq, José Gregorio Soto Camposr
a Neumología, Hospital de la Santa Creu i Sant Pau, Barcelona, España
b Otorrinolaringología, Hospital Clinic de Barcelona, España
c Medicina de Urgencias, Hospital de Verín, Orense, España
d Neumología, Complejo Hospitalario Universitario, A Coruña, España
e Hospital de São Sebastião – CHEDV, Santa Maria da Feira, Portugal
f Neumonología, Hospital Rossi La Plata, Argentina
g Farmacología clínica, Agencia Española de Medicamentos y Productos Sanitarios (AEMPS), Madrid, España
h Farmacia Hospitalaria, Hospital de la Santa Creu i Sant Pau, Barcelona, España
i Medicina de familia, Centro de Salud de Bargas, Toledo, España
j Medicina de familia, Centro de Salud Lucena I, Lucena, Córdoba, España
k Neumología Pediátrica, Hospital Universitario Donostia, Donostia-San, Sebastián, España
l Medicina de familia, semFYC, Centro de Salud Francia, Fuenlabrada, Dirección Asistencial Oeste, Madrid, España
m Pediatría, Centro de Salud de Pizarrales, Salamanca, España
n Farmacia comunitaria, Farmacia Dr, Javier Plaza Zamora, Mazarrón, Murcia, España
o Centro de Salud La Candelaria, Sevilla, España
p Alergología, Hospital Universitario La Paz, Madrid, España
q Alergología Pediátrica, Hospital Católico Universitario Casa de Salud, Valencia, España
r Neumología, Hospital Universitario de Jerez, Jerez de la Frontera, España
Ver más
Podcast
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (24)
Show moreShow less
Tables (70)
Table 1. Classification of the quality of evidence.
Table 2. Prevalence of asthma in adults and adolescents.
Table 3. Factors associated with the development of asthma.
Table 4. Triggers of asthma symptoms and exacerbations.
Table 5. Cells and structural elements of the airways involved in asthma.
Table 6. Inflammatory cells involved in asthma.
Table 7. Relevant molecules involved in the asthma inflammatory process.
Table 8. Mechanisms of airway obstruction in asthma.
Table 9. Mechanisms of bronchial hyperresponsiveness.
Table 10. Differential diagnosis of childhood asthma.
Table 11. F Traditional phenotypes in wheezing children from the Tucson study based on their long-term time.
Table 12. Asthma Predictive Index.
Table 13. Key questions for the diagnostic suspicion of asthma.118
Table 14. Differential diagnosis of asthma in adults.
Table 15. Reversibility and daily variability criteria recommended for the diagnosis of asthma.
Table 16. Standard battery of aeroallergens used in intraepidermal skin tests or prick test.*
Table 17. The classification of asthma severity when it is well-controlled with treatment (stratified by steps).
Table 18. Classification of asthma control in adults.
Table 19. Main risk factors for exacerbations.
Table 20. Asthma Control Questionnaire in Children (CAN).210
Table 21. Pediatric Asthma Control Test (ACT) questionnaire validated in Spanish.212,213
Table 22. Risk factors for asthma exacerbations in children.214,215
Table 23. Asthma treatment goals.
Table 24. Characteristics of inhaled β2-adrenergic agonists.
Table 25. Approximate potency of ICS (based on results of efficacy/safety studies).
Table 26. Aerodynamic properties provided by inhalers (based in part on Giner 2016).216
Table 27. In Information and basic skills that should be learned by a patient with asthma.
Table 28. Asthma action plan.
Table 29. Educational tasks to be implemented at each visit.
Table 30. Risk factors for life-threatening asthma exacerbation.
Table 31. Assessment of severity of asthma exacerbation.
Table 32. Drugs and doses commonly used for treating asthma exacerbations.
Table 33. Criteria for hospital admission and ICU admission.
Table 34. Criteria for specialized evaluation of patients within a month after an asthma exacerbation episode.531
Table 35. Key aspects of the education of a child with asthma.
Table 36. Components of a personalized action plan.
Table 37. Written action plan to maintain asthma control.
Table 38. Action plan for treating an asthma exacerbation at home.
Table 39. Comparable doses of inhaled glucocorticoids commonly used in pediatric age (μg/day).
Table 40. Pulmonary Score for clinical assessment of asthma exacerbation in children.*
Table 41. Overall evaluation of the severity of asthma exacerbation in children by integrating the Pulmonary Score and the arterial oxygen saturation.
Table 42. Phenotypes of rhinitis.
Table 43. Classification of allergic rhinits.
Table 44. Interrelationship between rhinitis and asthma: risk factors for asthma.
Table 45. Criteria for the indication of biologics in the treatment of nasal poliposis proposed by the POLINA consensus.694
Table 46. Severe uncontrolled asthma: definition and control.
Table 47. Differential diagnosis in asthma: diseases mimicking asthma and their corresponding diagnostic tests.
Table 48. Common comorbidities and aggravating factors of asthma with their corresponding diagnostic tests and treatment.
Table 49. Severe asthma phenotypes.
Table 50. Biologics approved for the treatment of SUA and their characteristics.
Table 51. Protocol of monitoring side effects of systemic glucocorticoids.
Table 52. Diseases mimicking severe asthma in children.
Table 53. Causative agents of occupational asthma.994,995
Table 54. Diagnostic tests in occupational asthma.
Table 55. Classification of some NSAIDs based on their capacity of inhibition of cyclooxygenase isoforms.1055
Table 56. Possible pharmacological interactions between drugs used for the treatment of COVID-19 and those used for asthma (based on the proposals from the Neumo SEFH Group 2020).1106
Table 57. Diagnostic criteria ISHAM 2013, updated 2016.
Table 58. Classification of ABPA based on radiological findings.
Table 59. Diagnostic criteria of EGPA according to the American College of Rheumatology 1990.1146
Table 60. Classification criteria of EGPA according to the American College of Rheumatology/European Alliance of Associations for Rheumatology 2022.1147
Table 61. Actions aimed to improve continuity of care in asthma.
Table 62. Healthcare quality indicators for asthma proposed by the multidisciplinary expert group (Asmaforum II).
Show moreShow less
Additional material (3)
Abstract

The Spanish Guideline on the Management of Asthma, better known by its acronym in Spanish GEMA, has been available for more than 20 years. Twenty-one scientific societies or related groups both from Spain and internationally have participated in the preparation and development of the updated edition of GEMA, which in fact has been currently positioned as the reference guide on asthma in the Spanish language worldwide.

Its objective is to prevent and improve the clinical situation of people with asthma by increasing the knowledge of healthcare professionals involved in their care. Its purpose is to convert scientific evidence into simple and easy-to-follow practical recommendations. Therefore, it is not a monograph that brings together all the scientific knowledge about the disease, but rather a brief document with the essentials, designed to be applied quickly in routine clinical practice. The guidelines are necessarily multidisciplinary, developed to be useful and an indispensable tool for physicians of different specialties, as well as nurses and pharmacists.

Probably the most outstanding aspects of the guide are the recommendations to: establish the diagnosis of asthma using a sequential algorithm based on objective diagnostic tests; the follow-up of patients, preferably based on the strategy of achieving and maintaining control of the disease; treatment according to the level of severity of asthma, using six steps from least to greatest need of pharmaceutical drugs, and the treatment algorithm for the indication of biologics in patients with severe uncontrolled asthma based on phenotypes. And now, in addition to that, there is a novelty for easy use and follow-up through a computer application based on the chatbot-type conversational artificial intelligence (ia-GEMA).

Keywords:
Asthma
practical guidelines
diagnosis
treatment
Resumen

La Guía Española para el Manejo del Asma, mejor conocida por su acrónimo en español, GEMA, está a nuestra disposición desde hace más de veinte años. Veintiuna sociedades científicas o grupos relacionados, tanto de España como de otros países, han participado en la preparación y desarrollo de la edición actualizada de GEMA que, de hecho, se ha posicionado en la actualidad a nivel mundial como la guía de referencia sobre asma en lengua española.

Su objetivo es prevenir y mejorar la situación clínica de las personas con asma, aumentando el conocimiento de los profesionales sanitarios involucrados en su cuidado. Su propósito es convertir la evidencia científica en recomendaciones prácticas sencillas y fáciles de seguir. Por lo tanto, no se trata de una monografía que reúna todo el conocimiento científico sobre la enfermedad, sino más bien de un documento conciso con lo esencial, diseñado para ser aplicado rápidamente en la práctica clínica de rutina. Las recomendaciones son necesariamente multidisciplinares, están desarrolladas para ser útiles y una herramienta indispensable para médicos de diferentes especialidades, así como para profesionales de enfermería y farmacia.

Seguramente, los aspectos más destacados de la guía son las recomendaciones para: establecer el diagnóstico del asma utilizando un algoritmo secuencial basado en pruebas diagnósticas objetivas; el seguimiento de los pacientes, preferentemente basado en la estrategia de lograr y mantener el control de la enfermedad; el tratamiento según el nivel de gravedad del asma utilizando seis escalones, desde la menor hasta la mayor necesidad de medicamentos, y el algoritmo de tratamiento basado en fenotipos para la indicación de biológicos en pacientes con asma grave no controlada. A esto se suma ahora una novedad para su fácil uso y seguimiento a través de una aplicación informática basada en la inteligencia artificial conversacional de tipo chatbot (ia-GEMA).

Full Text
Foreword

Once again, I have the privilege and satisfaction of presenting the 2023 update of the Spanish Guideline for the Management of Asthma (GEMA), GEMA 5.3. Furthermore, this is a special edition as we celebrate the 20th anniversary of the acronym “GEMA.” Although we consider that the guideline was born in 1997 with the SEPAR-semFYC consensus on asthma (GEMA 1.0), it is worth noting that it was the first consensus conducted by SEPAR with another society. GEMA is now an international multidisciplinary guideline in which 17 scientific societies from Spain, Latin America (ALAT), and Portugal (SPP) participate. Twenty years later, it is a source of great pride for us to mention that GEMA is the world's reference guideline in Spanish for this disease. It is a global guide for a globalized world.

As in previous editions, in this one we have followed the same procedure to carry out the usual annual update, which basically consisted of incorporating new and relevant bibliographic references published in 2022. For this purpose, “our” four experts in reviewing the scientific literature, Drs. Astrid Crespo (Pulmonology), Miguel Ángel Lobo (Family Medicine), Álvaro Gimeno (Pediatrics), and Manuel Rial (Allergology), reviewed the articles published during that period in the main indexed journals of international literature. This review selected the 40 most appropriate citations for the update, which were used to edit the proGEMA5.3 text. This bibliographic selection, although not exclusively, was primarily used by the members of the GEMA Executive Committee to discuss and decide on the novelties for this new edition.

The main changes, edited in blue for easy identification, can be grouped into the following six conceptual areas:

Remission in asthma. A fashion subject of this year could not have been overlooked in the guide. However, all of this is provisional because, along with the Asthma Forum, SEPAR is sponsoring a consensus on the topic that involves over 120 asthma specialists, and its outcome will be binding for the future GEMA 5.4.

Diagnosis of asthma in adults and children. The publication of the recent ERS/ATS consensus on spirometry required the addition as a complementary criterion of bronchodilation in adults of the new ≥ 10% of the predicted FEV1 or FVC value, besides the classic criterion of ≥ 12% and 200mL of FEV1 after inhalation of the bronchodilator.

Treatment of asthma. Theophyllines are removed from the therapeutic armamentarium. The consequences of excessive use of SABA (SABINA studies) are described. Triple therapy in a single inhaler of fluticasone/vilanterol/umeclidinium, is incorporated into the treatment of severe asthma, which has been approved by regulatory agencies in Hispanic America but not by European agencies.

Treatment of severe uncontrolled asthma. A new definition is introduced, which includes the use of a third drug (LAMA) at high doses of ICS/LABA to establish it. New algorithms (with a new design) for the treatment of severe uncontrolled asthma are provided, including a specific one for corticosteroid-dependent asthma. Tezepelumab is included in the therapeutic algorithm, and benralizumab in the treatment of T2 asthma with blood eosinophils between 150-300/μL.

New recommended questionnaires. The Asthma Impairment and Risk Questionnaire (AIRQ) to determine the current level of control and future risk, a weighted scoring system is used, taking into account FEV1 (forced expiratory volume in 1 second), exacerbations, oral corticosteroid use, and asthma symptoms. This scoring system helps assess the response to biological drugs in asthma (FEOS). The Sino-Nasal Outcome Test 22 (SNOT-22) is used to assess the impact and quality of life caused by rhinosinusitis.

Finally, it is important to highlight some of the most relevant contributions from the recent POLINA consensus (chronic rhinosinusitis and nasal polyposis), such as the proposed stepped treatment approach based on severity and control, as well as the criteria for prescribing biological drugs.

However, without diminishing the importance of these changes, which are undoubtedly very relevant, perhaps the most notable innovation in this edition is the incorporation of a new tool called iaGEMA. It is a computer application that includes artificial intelligence software with which one can interact (chatbot-like). The application is addressed to the healthcare professional and will be capable of providing GEMA recommendations in response to real clinical questions regarding the care of asthma of patients. We believe that this will be the first guide that incorporates this technology, and we anticipate that the future of guideline implementation will involve technologies like the one offered by this new iaGEMA application.

Finally, on behalf of the members of the GEMA Executive Committee, the true “engine” behind the guide, it is a great personal satisfaction to have been able, once again, to fulfill the commitment of updating the guide. I would like to express my gratitude to all of them, as well as to the four expert reviewers and the staff at Luzan5, for their hard work and invaluable support, which have been crucial for successfully achieving the present GEMA 5.3 edition.

Dr. Vicente Plaza Moral

On behalf of the Executive Committee of GEMA 5.3

Objective

The main objective of the present guideline is to improve the control and quality of life of patients with asthma by enhancing the technical expertise of healthcare professionals in charge of them, particularly in aspects related to prevention and diagnosis-therapeutic evaluation of the disease.

GEMA, however, is a platform that brings together a series of complementary actions, all designed to reach the aforementioned objective, among which this document acquires a special relevance: an evidence-based clinical practice guideline. Further documents will complete the GEMA “family” (e.g., pocket-size GEMA, GEMA for patients, GEMA for educators, etc.).

Specifically, the current document (clinical practice guideline) as well as the whole strategy conforming the GEMA 5.3 platform, is addressed to professions in the setting of Family and Community Medicine; Primary Care Pediatrics; Pneumology, Allergology, Pediatric Pneumology and Allergology; Otorhinolaryngology; Pharmacology; Hospital and Primary Care Pharmacy; General and Specialized in Respiratory Diseases Nursing, educators, teachers, patients, and patients’ relatives and caregivers.

Method

Searching for evidence. Based on the previous (complete) edition of GEMA,1 published in 2015, and following the recommendations for Updating Clinical Practice Guidelines in the National Health System,2 the members of the Executive Committee undertook a systematic search of the literature to select and evaluate articles on asthma published from 2015 to 2020 (Pro-GEMA Project). After reviewing high impact factor journals of Pneumology, Allergology, Pediatrics, Primary Care, Internal Medicine and Otorhinolaryngology, which were also classified within the two first quartiles of their specialty field, a total of 120 documents were selected (abstracts available at http://www.progema-gemasma.com/foco.html) that were considered of interest for updating this guideline. All these documents were provided to the authors for evaluation. Furthermore, authors were encouraged to perform their own literature searches for specific topics. To this purpose, the procedure normally established to develop clinical practice guidelines was followed.3 Also, the reference lists of the main international practice guidelines4,5 were reviewed in order to identify the most relevant systematic reviews and clinical trials. These guidelines were searched in specialized databases (National Guideline Clearinghouse, National Library of Guidelines) and the TRIP medical literature meta-search engine database. Databases from the Centre for Reviews and Dissemination (DARE y HTA database) and The Cochrane Library were reviewed in order to identifying systematic reviews and evaluations of additional technologies. The search was completed with an update of the systematic reviews from the date of search and relevant studies included in the main electronic databases of original studies (MEDLINE, CENTRAL and EMBASE).

Classification of evidence. To assess the quality of evidence, an alphabetic classification was used (table 1) that classifies the information into four categories (A, B, C, D) reflecting the grade of confidence in the results obtained in the available studies. Category A would correspond to a high quality evidence and D to a very low quality. For category A. confidence in the results is high and the potential modification of available findings by further studies is unlikely. In contrast, for lower categories, C or D, the confidence level will be low or very low, and there is a high probability that further studies will modify the results, or even the direction of the effect. However, it must be remember that this system is very useful to categorize the evidence regarding therapeutic efficacy of drugs or other therapeutic actions, but the effect of other interventions may be underestimated. This can explain why evidence from studies aimed at determining the appropriateness of some diagnostic procedures has often been assigned a level of evidence C.

Table 1.

Classification of the quality of evidence.

Categories of evidence
SR of RCTs with or without MA; and RCTs with low risk of bias. Evidence based on a substantial number of well-designed studies with consistent results. 
SR of RCTs with or without MA; and RCTs with moderate risk of bias. Evidence obtained from a limited number of studies and/or inconsistent results. 
Evidence obtained from non-randomized, observational or uncontrolled studies. 
Clinical experience or scientific literature that cannot be included in category C. 

SR: systematic reviews; RCTs: randomized controlled trials; MA: meta-analysis.

Taking into account the recent emergence of new approaches used to classify the quality of evidence based on aspects other than the study design,6,7 some of the characteristics of the GRADE framework were used,8 although the GRADE system was not applied in full.

Classification of recommendations. To classify the relevance and consistency of clinical recommendations, the same method used in the previous editions of GEMA was followed, in which recommendations were categorized in two levels: robust recommendations (R1), that is, those to be associated with more benefits than risks according to the opinion of the group of authors, and weak recommendations (R2), that is, those in which some uncertainty exists as to whether its application might entail more benefits than risks. To carry out this distribution in R1 o R2, the quality of information was weighed (based on the above-mentioned classification), along with the balance between risks and benefits of interventions, the costs (according to the available specialized literature), and the patients’ values and preferences (through the participation of FENAER members).

The categorization of the recommendation level was established by consensus, first of the authors (see below for the working method used) and finally by the agreement of reviewers (through the Delphi method), whose opinions were binding for the final version of all recommendations.

Drafting text and building consensus of recommendations. The writing process was based on a pyramidal consensus system going from a multidisciplinary thematic mini-consensus by chapter to a large final consensus among all authors and reviewers. Based on the document of the previous edition and the new references on asthma published between 2015 and 2020, a group of authors and coordinators made up by experts from the participating scientific societies drew up the new chapter sections they were assigned (including the classification of evidence and recommendations). The authors submitted their texts to each chapter coordinators who were members of the GEMA Executive Committee. After unifying and reviewing the texts, the chapter coordinator submitted the draft to the authors of each chapter in order to reach the first partial consensus. After implementation of changes, all chapters were brought together in one single document which, in turn, was sent to all authors and coordinators for telematics discussion (and for face-to-face group discussion, when necessary) and approval. The resulting document was submitted to experts in the methodology of clinical practice guidelines from the INPECS (Institute for Clinical and Healthcare Excellence), who made a critical review of the methodology and writing of both the text and the recommendations. Finally, after these modifications and improvements, recommendations were revised and agreed on (through the Delphi method) by a group of experts in asthma from the participating societies. Recommendations not achieving a certain consensus level were removed from the final document.

Method followed for bibliographic updating of GEMA 5.3. Four asthma experts, Drs. Astrid Crespo (Pulmonology), Miguel Ángel Lobo (Family Medicine), Álvaro Gimeno (Pediatrics), and Manuel Rial (Allergology), reviewed the articles published on the disease since the previous GEMA update (GEMA 5.2). They focused on journals with high impact factors, many of which are ranked in the first quartiles of the specialties of Pulmonology, Allergology, Pediatrics, Family Medicine, and Internal Medicine. As in previous instances, this selection of articles was predominantly (although not exclusively) used by the members of the GEMA Executive Committee to discuss and decide on the majority of novelties to be included in the new GEMA 5.3.

Editorial independence

The GEMA5.0 project was funded by pharmaceutical companies listed on the back cover of the document. The viewpoints of these funding bodies did not influence the content of the guide.

The authors of this guide declare that in the past two years, they have received honoraria for their participation in meetings, congresses, or research projects organized by the following pharmaceutical companies: ALK, AstraZeneca, Bial, Boehringer-Ingelheim, Chiesi, Esteve, GlaxoSmithKline, Leti, Menarini, MSD, Mundipharma, Novartis, Orion, Pfizer, Sanofi, Teva, and Zambón.

1Introduction1.1Definition

Asthma is a syndrome that includes various clinical phenotypes that share similar clinical manifestations, probably of different etiologies. Classically, it is defined as a chronic inflammatory disease of the respiratory tract involving various cells and mediators of inflammation. It is partially influenced by genetic factors and is characterized by bronchial hyperresponsiveness and a variable degree of airflow obstruction that is totally or partially reversible by either the action of drugs or spontaneously.9 As a chronic disease included in the current different strategies for the care of patients with chronic conditions, the objective of asthma management is to achieve and maintain control of the disease and prevention of future risks, particularly exacerbations, which can be life-threatening and generate a burden for the society.10

1.2Prevalence

Asthma prevalence is highly variable worldwide, ranging from 2% in Tartu (Estonia) to 11.9% in Melbourne (Australia). Similarly, the prevalence of wheezing (over the last 12 months) varies from 4.1% in Mumbai (India) to 32% in Dublin (Ireland).11,12

According to the 2015 Global Burden of Disease study, the prevalence of asthma has increased worldwide by 12.6% from 1990 to 2015. On the contrary, the age-standardized mortality rate has decreased by almost 59% in the same period.13 This increase in prevalence mainly affects middle-aged individuals and women and can be explained by a rise in allergic asthma, with stabilization of non-allergic asthma.14

The European Respiratory Health Study in Spain reported prevalence rates of 4.7% in Albacete, 3.5% in Barcelona, 1.1% in Galdakao, 1% in Huelva, and 1.7% in Oviedo.15 Other recent studies report highly variable prevalences based on different variables, such as age (adolescents), ranging from 10.6%16 to 13.4%;17 the method used (self-reported by the patient), 13.5%;18 or the study setting (work environment), 2.5%.19

In Spain, a study carried out in Navarre showed a prevalence of 10.6% in adolescents.16

In another study conducted in rural areas of Navarre, a prevalence of asthma of 13.4% was found among adolescents. The prevalence was slightly higher in females (13.7% compared to 10.9% in males), with rhinitis, wheezing (especially associated with physical activity), and dry cough as related symptoms.17

A study carried out in Argentina showed a prevalence of asthma in adults (between 20 and 44 years of age) of 6.4%20 (table 2).

Table 2.

Prevalence of asthma in adults and adolescents.

Author  Setting  Year  Prevalence  Comment 
Álvarez16  Navarra  2014  10.6%  Adolescents 
Elizalde17  Navarra (rural)  2018  13.4%  Adolescents 
Vila-Rigat19  Barcelona  2014  2.5%  Working-age population (16-64 years) 
López18  Madrid  2017  6.3%/13.5%  Current asthma/accumulated asthma 
Arias20  Argentina  2018  6.4%  Adults 20-44 years 
1.3Risk factors

Risk factors for the development of asthma syndrome should be distinguished from triggers of asthma symptoms or asthma exacerbations.

In relation to factors associated with the development of asthma, those better known or with a higher degree of association are shown in table 3. Many host-related factors are perinatal, while environmental factors vary greatly and can impact on patients of different age groups.

Table 3.

Factors associated with the development of asthma.

Risk factors  Evidence  Association  Type of study  Reference 
HOST-RELATED FACTORS
Atopy  OR 3.5 (2.3-5.3)  Arbes 200721 
Early menarche  OR 1.08 (1.04-1.12)  Minelli 201822 
Obesity  RR 1.50 (1.22-1.83)  Egan 201323 
Bronchial hyperresponsiveness  OR 4.2 (1.92-9.23)  Carey 199624 
RhinitisOR 3.21 (2.21-4.71)  Guerra 200225 
OR 4.16 (3.57-4.86)  Burgess 200726 
RR 3.53 (2.11-5.91)  Shaaban 200827 
PERINATAL FACTORS
Maternal age  OR 0.85 (0.79-0.92) 1.4  Gómez 201828 
Preeclampsia  OR 4.01 (1.11-14.43)  Stokholm 201729 
PrematurityOR 2.81 (2.52-3.12) 2  Been 201430 
OR 1.37 (1.17-1.62) 3  Been 201430 
OR 4.30 (2.33-7.91)  Leps 201831 
Cesarean section  HR 1.52 (1.42-1.62)  Tollånes 200832 
Neonatal jaundice  OR 1.64 (1.36-1.98)  Ku 201233 
LactationOR 0.88 (0.82-0.95) 4  Silvers 201234 
OR 0.70 (0.60-0.81) 4  Gdalevich 200135 
Tobacco consumption during pregnancyOR 1.72 (1.11-2.67)  Strachan 199636 
OR 1.85 (1.35-2.53)  Burke 201237 
OR 2.70 (1.13-6.45)  Cunningham 199638 
OR 1.65 (1.18-2.31)  Neuman 201239 
Mother's dietOR 0.49 (0.27-0.90) 2.4  Litonjua 200640 
OR 0.54 (0.33-0.88) 5.4  Wolks 201741 
OR 0.33 (0.11-0.98) 4  Devereux 200742 
OR 0.86 (0.78-0.95) 6.4  García-Marcos 201343 
Infant's diet  RR 0.66 (0.47-0.94) 7.4  Hibbs 201844 
Pulmonary function of the neonate  OR 2.10 (1.12-3.93)  Håland 200645 
ENVIRONMENTAL FACTORS
AeroallergensOR 0.49 (0.29-0.83) 8.4  Kerkhof 200946 
OR 0.68 (0.49-0.95) 9.4  Kerkhof 200946 
Workplace allergensRR 2.2 (1.3-4.0)  Kogevinas 200747 
OR 0.55 (0.43-0.70) 10.4  Hoppin 200848 
Respiratory infections  OR 0.52 (0.29-0.92) 11.4  Illi 200149 
TobaccoRR 3.9 (1.7-8.5)  Gilliland 200650 
HR 1.43 (1.15-1.77)  Coogan 201551 
HR 1.21 (1.00-1.45) 12  Coogan 201551 
Environmental contamination  OR 1.34 (1.17-1.54)  Orellano 201852 
DRUGS         
Paracetamol  OR 1.26 (1.02-1.58)  Sordillo 201553 
Antacids  RR 1.45 (1.35-1.56)  Lai 201854 
AntibioticsOR 1.12 (0.88-1.42) 13  Marra 200655 
OR 0.6 (0.4-0.96) 4  Goksör 201356 
HR 1.23 (1.20-1.27) 14  Loewen 201857 
OR 1.75 (1.40-2.17) 15  Hoskin-Parr 201358 
Hormone replacement therapy  HR (1.54 (1.13-2.09) 16  Romieu 201059 

HR: hazard ratio; OR: odds ratio. Type of study: a meta-analysis-systematic review, b prospective epidemiological study, c retrospective epidemiological study, d clinical trial.

Comments: 1 female sex, 2 very preterm, 3 moderate preterm, 4 protective factor, 5 level of vitamin D at the beginning of pregnancy, 6 Mediterranean diet, 7 vitamin D supplement, 8 dog exposure, 9 cat exposure, 10 living on a farm, 11 non-respiratory viral infection, 12 passive smoking, 13 no association, 14 prenatal exposure, 15 postnatal exposure, 16 with estrogens only.

On the other hand, the most common triggers of asthma symptoms or exacerbations are presented in table 4. It is important to be aware of them because they can lead to serious situations and, therefore, should be avoided.

Table 4.

Triggers of asthma symptoms and exacerbations.

Environmenal factorsAtmosphericPollution  SO2 NO2OzoneCOAirborne particles 
Plants  Grass pollenTree pollenWeed pollen 
Domestic  Dust mites  Animal danderCockroaches 
Fungi and virusesAlternaria alternata Cladosporium herbarum  Penicillium Aspergillus fumigatus 
Rhinovirus and other respiratory viruses   
Systemic factorsDrugsAntibiotics  β-Non-selective systemic and topical blockers 
Acetylsalicylic acid  NSAIDs 
FoodsCow milk  Cereals 
Eggs  Fish 
Nuts  Seafood 
Foods containing sulfites  Nuts, wine, lemon juice, lime juice, grape juice, dried potatoes, vinegar, seafood, beer, etc. 
Plant panallergens such as profilins or lipid transfer protein (LTP)
OtherHymenoptera venomApis melífera (bee) 
Vespula spp, Polistes dominulus (wasp) 
Work-related factorsLOW MOLECULAR WEIGHT SUBSTANCES  INDUSTRY INVOLVES
Drugs  Pharmaceutical industry
Anhydrides  Plastic industry
Diisocyanates  Polyurethane, plastic, varnish and enamel industries
Woods  Sawmills, carpentry work, cabinetmaking
Metals  Foundries, nickel plating, silver plating, tanning, boiler cleaning industries
Other  Cosmetic industry, hairdressing, photograph developing, cooling, dyes
HIGH MOLECULAR WEIGHT SUBSTANCES  INDUSTRY INVOLVED
Substances of plant origin, powder and flours  Farmers, port workers, mills, bakeries, beer industry, soy processing, cacao, coffee and tea industries, textile industry
Foods  Food industry
Plant enzymes  Food industry, pharmaceutical industry,
Vegatable gums  Food industry, printing presses, latex industry, healthcare
Fungi and spores  Bakeries, farms, farmers
Animal enzymes  Mills, carmine manufacturing

Genetic factors are gaining increasing relevance as research progresses. Current studies indicate their involvement in the onset of asthma, the phenotypic expression of the disease, the individual response to triggers of asthma symptoms or exacerbations, and very especially in the response to new therapies in cases of severe asthma.60

Finally, it should be emphasized the growing evidence of the importance of environmental pollution, both indoors, from biomass combustion, and outdoors, from the combustion of fossil fuel-derived products.61,62 This environmental pollution acts as a contributing factor in the onset of asthma and as a trigger for asthma symptoms or exacerbations. Furthermore, it contributes to increased morbidity and mortality of asthma, as well as the incidence of other chronic respiratory diseases, cardiovascular diseases, and various types of cancer.63

1.4Pathogenesis

Inflammation affects the entire respiratory tract, including the nasal mucosa, and is present even when symptoms are episodic. However, the relationship between the severity of asthma and the intensity of inflammation has not been consistently established.64 The epithelium initiates the response to inhaled substances, secreting cytokines such as Thymic Stromal Lymphopoietin (TSLP), IL-33 y IL-25, which are crucial for activation of the type 2 innate immune system (table 5).67,68

Table 5.

Cells and structural elements of the airways involved in asthma.

Bronchial epithelium: It is damaged, with a loss of both ciliated and secretory cells. Epithelial cells are sensitive to changes in their microenvironment, express multiple inflammatory proteins and release cytokines, chemokines, and lipid mediators in response to physical changes. Their production can also be stimulated by pollutants and viral infections. The repairing process following epithelial damage may be abnormal, which enhances obstruction bronchial lesions associated with asthma.65 
Bronchial smooth muscle: Its cells show an increased proliferation (hyperplasia) and growth (hypertrophy) with the expression of proinflammatory mediators similar to those found in epithelial cells.66 
Endothelial cells: They participate in the recruitment of inflammatory cells from the blood vessels to the airways through the expression of adhesion molecules. 
Fibroblasts and myofibroblasts: After being stimulated by inflammatory mediators and growth factors, these cells produce some components of the connective tissue, such as collagen and proteoglycans that are involved in airways remodeling. 
Airway cholinergic nerves: These can be activated by neural reflexes and cause bronchoconstriction and mucus secretion. Sensorial nerves may provoke symptoms such as cough and chest tightness, and may release inflammatory neuropeptides. 

Once activated, type 2 innate lymphoid cells secrete type 2 pro-inflammatory cytokines, such as IL-4, IL-5 and IL-13, which assume the role of starting and maintaining T2 response (table 6).

Table 6.

Inflammatory cells involved in asthma.

T lymphocytes (TL): are increased in number in the airways, with an imbalance in the Th1/Th2 ratio and predominance of Th2 that release specific cytokines, including IL 4, 5, 9 and 13. The cytokines orchestrate the eosinophilic inflammation and the production of IgE by B lymphocytes. Levels of LT regulators are decreased, while LT NK are increased.69 
Mastocytes: are increased in the bronchial epithelium and infiltrate the bronchial wall smooth muscle. Their activation releases mediators with bronchoconstriction and proinflammatory activity, such as histamine, leukotrienes, and prostaglandin D2.70 They are activated by allergens, osmotic stimuli (such as those causing exercise-induced bronchoconstriction) and neuronal connections. 
Eosinophils: are increased in the airways and their number correlates with severity. They are activated and their apoptosis is inhibited. They release inflammatory enzymes that harm epithelial cells and generate mediators that amplify the inflammatory response.71 
Neutrophils: are increased in the airways of some patients with severe asthma, during exacerbations, and in smokers with asthma. Their pathophysiological role is not well defined and their increase may be due to treatment with glucocorticoids.62 
Dendritic cells: act as antigen-presenting cells that interact with lymph node regulating cells and stimulate the production of Th2 lymphocytes.72 
Macrophages: may be activated by allergens through the low-affinity IgE receptors and release mediators that boost the inflammatory response, particularly in severe asthma.73 
Pulmonary neuroendocrine cells: contribute to Th2 response and stimulate mucus-producing cells.74 

On the other hand, Dendritic cells promote the development of T-helper (Th2) lymphocytes, which secrete the previously mentioned type 2 cytokines. Recent studies have shown that not all patients develop Th2 inflammation, but there are also other molecules such as IL-17 and IF-γ that are involved in the so-called Th2-low asthma.

Molecules involved in this inflammatory process are summarised in table 7.

Table 7.

Relevant molecules involved in the asthma inflammatory process.

Chemokines. These are mainly expressed by epithelial cells and are important in the recruitment of inflammatory cells in the airways. 
Cysteinyl leukotrienes. Potent bronchoconstrictors released by mast cells and eosinophils. 
Cytokines. They drive and modify the inflammatory response in asthma, and determine its severity:75• IL-1β and TNFα: amplify the inflammatory response.• GM-GSF: prolongs eosinophil survival in the airways.Cytokines derived from the epithelium:– IL-33: promotes the pro-allergic inflammatory properties of CD4 cells and acts as a chemoattractant for Th2 cells.– IL-25: involved in eosinophilic inflammation, remodeling, and bronchial hyperreactivity (the latter being more debated).– TSLP: induces eosinophilia, increases IgE levels, hyperresponsiveness, and airway remodeling.Cytokines derived from Th2 cells: – IL-4: important to the differentiation of Th2 lymphocytes, increase of mucus secretion, and IgE synthesis.– IL-5: necessary for the differentiation and survival of eosinophils.– IL-13: important for the synthesis of IgE and mucous cells metaplasia 
Histamine. Released by mast cells, contributes to bronchoconstriction and the inflammatory response. 
Nitric oxide. A potent vasodilator predominantly produced in epithelial cells by the inducible nitric oxide synthase enzyme. 
Prostaglandin D2. A bronchoconstrictor mostly derived from mast cells; it is involved in the recruitment of Th2 lymphocytes to the airways. 

GM-GSF: Granulocyte-macrophage colony-stimulating factor; TNF: Tumor necrosis factor.

Patients with asthma may present a phenomenon, known as airway remodeling, which include: thickening of the reticular layer of the basal membrane, subepithelial fibrosis, hypertrophy and hyperplasia of the bronchial smooth muscle, vascular proliferation and dilatation, mucosal gland hyperplasia and mucus hypersecretion, all of which are associated with a progressive deterioration of pulmonary function.69 Some of these changes are related to the severity of the disease and may lead to a bronchial obstruction, which is occasionally irreversible.76

These changes may result from a repairing response to chronic inflammation or may occur independently of the inflammatory process.77

Narrowing of the airways is common end result of the pathophysiological changes and the origin of most symptoms. This limitation of airflow and the symptoms it triggers can spontaneously resolve or respond to medication (reversibility) and may even be absent for some time in a particular patient. Table 8 shows the different mechanisms that contribute to the onset of obstruction.

Table 8.

Mechanisms of airway obstruction in asthma.

Contraction of bronchial smooth muscle: It occurs in response to multiple mediators and neurotransmitters with bronchoconstrictor effects and is the most prominent mechanism of airway narrowing. Monomeric G proteins (RhoA and Rac1) are involved in the contraction and proliferation of muscle cells. It is largely reversible with bronchodilator drugs. 
Edema of the airways: It is caused by the microvascular exudation in response to inflammatory mediators. It is particularly important during acute exacerbations. 
Mucus hypersecretion: It is caused by an increase in the number of goblet cells in the epithelium and an enlargement of the submucosal glands. It can lead to mucus plugs, which are associated with the severity of asthma.78 
Structural changes in the airways: Subepithelial fibrosis due to deposition of collagen fibers and proteoglycans under the basal membrane; smooth muscle hypertrophy and hyperplasia and increased circulation within the blood vessels of the bronchial wall, with enhanced permeability. 

Various triggering agents may cause a significant airway narrowing, thus leading to an asthma exacerbation. The most severe episodes usually occur in association with viral infections of the upper respiratory tract (mainly rhinovirus and respiratory syncytial virus) or exposure to allergens.79 Also, exacerbations may be caused by non-steroidal anti-inflammatory drugs (NSAIDs) in patients with hypersensitivity to these drugs, physical exercise, cold air and certain non-specific irritants.80–82 The intensity of the response to these stimuli is related to the underlying inflammation.

Bronchial hyperresponsiveness (BHR) is an additional pathophysiological characteristic of asthma, which leads to airway narrowing in response to stimuli that are harmless to people without asthma. BHR is linked to airway inflammation and repair, and is partially or totally reversible with therapy. Mechanisms involved in BHR are shown in table 9. The degree of BHR is partially correlated with the clinical severity of asthma and the inflammation markers.84 Anti-inflammatory therapy improves asthma control and attenuates BHR, but does not completely suppress it.85

Table 9.

Mechanisms of bronchial hyperresponsiveness.

Excessive contraction of the airway smooth muscle. It may result from increased volume and/or contractility of bronchial smooth muscle cells. 
Uncoupling of airway contraction. It occurs as a result of inflammatory changes in the airway wall that may lead to its narrowing and to loss of the maximum level of contraction, which can be found in healthy airways when a bronchoconstrictor agent is inhaled. 
Thickening of the airway wall. Edema and structural changes amplify the bronchial wall narrowing due to the airway muscle contraction.76 
Sensitized sensory nerves. Their sensitivity may be enhanced by inflammation resulting in exaggerating bronchoconstriction in response to sensory stimuli.83 

Variability is another important feature of asthma. It is defined as the variation or fluctuation of both symptoms and pulmonary function over time, even during the same day, beyond physiological circadian changes.

1.5Childhood asthma

Asthma is one of the most prevalent chronic diseases in childhood. According to the International Study of Asthma and Allergies in Childhood (ISAAC), the prevalence in Spain is 10%, which is similar to the prevalence in the European Union. It is more common in coastal areas and among males in the 6-7-year age group.86–89

It is estimated that more than half of adults with asthma already had the disease during childhood.90

In the first three years of life, definition, diagnostic criteria, and even the classification of asthma are complex and controversial,91 which make difficult to determine the prevalence of asthma at these ages.92,93

This is because typical symptoms (coughing, wheezing, and difficulty breathing) are common in children under 3 years of age without asthma and also for the impossibility to assess lung function routinely.

The definitive diagnosis of asthma requires the exclusion of other diseases that can present with similar signs and symptoms (table 10).94–97 In fact, some of these conditions may be associated with asthma.98

Table 10.

Differential diagnosis of childhood asthma.

Cystic fibrosis  Airway anomalies. Tracheomalacia. Vascular ring 
Bronchiectasis  Respiratory dysfunction. Induced laryngeal obstruction 
Ciliary dyskinesia  Psychogenic cough 
Chronic lung disease of prematurity  Pulmonary tuberculosis 
Chronic aspiration. Dysphagia.  Chronic interstitial disease 
Foreign body aspiration  Congenital heart disease 
Gastroesophageal reflux  Primary or secondary tumors 

The presence of personal and family atopy is the most important risk factor for the subsequent development of asthma. Other factors include age at onset, severity and frequency of episodes, male gender, and severe bronchiolitis (RSV, rhinovirus).98–100

After the first description of phenotypes in childhood asthma reported in the study of Tucson (table 11),101 a number of prospective clinical studies (cohorts of children followed since birth)102–104 or complex biostatistical studies (cluster of populations without previous hypothesis)105 have been published, all of them trying to identify different phenotypes of childhood asthma. The clinical value of these studies is controversial.103

Table 11.

F Traditional phenotypes in wheezing children from the Tucson study based on their long-term time.

1. Early-onset transient wheezing 
• Onset within the first year of life with resolution by 3 years of age.• Negative IgE and/or skin tests, with no traits or history of atopy.• Decreased pulmonary function at birth with low values at 16 years of age.• Negative findings in bronchial hyperresponsiveness and variability of peak expiratory flow (PEF) studies at 11 years of age.• Risk factors: maternal smoking during pregnancy, male sex, prematurity, cohabitation with older brothers and/or daycare attendance. 
2. Persistent (non-atopic) wheezing 
• It usually starts before the first year and persists at 6 years of age.• Both sexes affected equally.• Negative IgE and/or skin tests, with no traits or history of atopy.• Normal pulmonary function at birth, although decreased at 6 and 11 years of age.• Bronchial hyperresponsiveness that decreases with age.• Remission normally occurs in adolescence. 
3. Late-onset (atopic) wheezing 
• The first episode occurs after the first year of age; more common in boys.• Increased IgE and/or positive skin tests, atopic traits, and family history of atopy.• Normal pulmonary function at birth followed by a decline until 6 years of age; thereafter, pulmonary function stabilizes at below levels of normal.• Bronchial hyperresponsiveness.• Persistence in adolescence. 

Based on the findings from these studies, some tools or models have been developed to predict the future risk in children with asthma but a few of these instruments have been validated. The best known instrument is the Asthma Predictive Index (table 12), which was developed from the Tucson cohort study.106

Table 12.

Asthma Predictive Index.

Previous condition 
Infants with 3 or more wheezing episodes per year during the first 3 years of life who meet one major criterion and 2 minor criteria. 
Major criteria 
– Medical diagnosis of asthma in one of the parents.– Medical diagnosis of atopic eczema (at 2-3 years of age). 
Minor criteria 
– Presence of allergic rhinitis diagnosed by a physician (at 2-3 years of age).– Wheezing not associated with colds.– Peripheral blood eosinophilia equal or higher than 4%. 
Predictive values for asthma diagnosis at any time between 6 and 13 years of age 
– Positive predictive value 77%.– Negative predictive value 68%. 

Although other indexes or modifications of the Asthma Predictive Index have been developed, this one continues to be the most useful, because of its simplicity, having been more validated, and better positive likelihood ratio.107

The diagnosis of asthma in children under 3 years of age must be probabilistic, a probability that increases in the presence of atopy. The term asthma should not be avoided when there are more than 3 episodes a year, or severe episodes, of coughing, wheezing, and difficulty breathing, with a good response to maintenance treatment with inhaled glucocorticoids and worsening of symptoms upon withdrawal of this medication.

2Diagnosis2.1Clinical features

The diagnosis of asthma should be considered in the presence of clinical suspicion based on signs and symptoms, such as wheezing (the most typical symptom),108 dyspnea or breathing difficulty, cough, and chest tightness. These are named “guide symptoms”,109,110 which are usually variable regarding intensity and the time of appearance, occurring mainly at night or in the early morning and are caused by different triggers (viral infections, allergens, tobacco smoke, exercise, emotions, etc.). Seasonal variations, along with a family and personal history of atopy are important aspects to be considered.111–114

Usually, several signs or symptoms appear together; when they occur as single manifestations, they are usually poor predictive of asthma.111,115,116 None of these symptoms and signs are specific to asthma,117 hence the need to include some objective diagnostic test, usually respiratory function tests.

The patient's clinical history should also include other aspects, such as the onset of symptoms, the presence of chronic rhinosinusitis with or without polyposis, rhinitis, dermatitis, and a family history of asthma or atopy,112 all of which increase the probability to establish a diagnosis of asthma. Table 13 shows the key questions for the identification of patients with suspected asthma.109,110

Table 13.

Key questions for the diagnostic suspicion of asthma.118

– Have you ever had “whistling” in the chest?– Have you had cough especially at night?– Have you had cough, wheezing, breathing difficulty in certain periods of the year or when in contact with animals, plants, tobacco or at the workplace?– Have you had cough, “whistling”, breathing difficulty after a moderate or intense physical exercise?  – Have you had colds lasting more than 10 days or “going down into the chest”?– Have you used inhaled medications that relieve your symptoms?– Do you have any kind of allergy? Do you have any relatives with asthma or allergy? 
Modified from García Polo 2012 and Martín Olmedo 2001.109,110

On physical examination, wheezing on auscultation of the chest is most characteristic finding, and sometimes nasal obstruction on anterior rhinoscopy, as well as dermatitis or eczema. However, a normal physical examination does not exclude a diagnosis of asthma.

If the onset of the disease presents with acute symptoms, a brief medical history and physical examination will be performed, and treatment will be initiated. Objective diagnostic tests will be conducted once the symptoms are under control.115

If asthma is suspected, a differential diagnosis with other diseases, particularly chronic obstructive pulmonary disease (COPD) should be made, as shown in table 14.

Table 14.

Differential diagnosis of asthma in adults.

  ASTHMA  COPD 
Age at onset  Any age  After 40 years of age 
Smoking  Irrelevant  Almost always 
Presence of atopy  Common  Uncommon 
Family history  Common  Not assessable 
Symptom variability  Yes  No 
Reversibility of bronchial obstruction  Significant  Usually less significant 
Response to glucocorticoids  Very good, almost always  Undetermined or variable 
  Other possible diseases  Characteristic symptoms 
Age between 15 and 40 years  – Inducible laryngeal obstruction– Hyperventilation– Inhaled foreign body– Cystic fibrosis– Bronchiectasis– Congenital heart disease– Pulmonary thromboembolism  – Dyspnea, inspiratory stridor– Fainting, paresthesia– Sudden onset of symptoms– Excessive cough and mucus– Recurrent infections– Heart murmurs– Sudden onset of dyspnea, tachypnea, chest pain 
Age older than 40 years of age  – Inducible laryngeal obstruction– Hyperventilation– Bronchiectasis– Parenchymal lung disease– Heart failure– Pulmonary thromboembolism  – Dyspnea, inspiratory stridor– Fainting, paresthesia– Recurrent infections– Exertional dyspnea, non-productive cough– Exertional dyspnea, nighttime symptoms– Sudden onset of dyspnea, tachypnea 
Modified from GINA 2019 and Plaza 2019113,117.
2.2Pulmonary function in adults

The diagnosis of asthma is established when in a patient with suspected symptoms of the disease, a pulmonary function test (preferably spirometry) objectively demonstrates an alteration compatible with asthma.119

The main functional abnormalities of asthma are airflow obstruction, reversibility, variability, and bronchial hyperresponsiveness.

Spirometry is the first-choice diagnostic test, as shown in the algorithm of the diagnostic process (Figure 1). The main parameters to be determined are forced expiratory volume in one second (FEV1) and forced vital capacity (FVC). Reference values should be adjusted to the age and ethnic group/race of each patient. Airway obstruction is defined as a FEV1/FVC ratio below the lower limit of reference values, which has been arbitrarily set at 0.7120 However, this criterion may lead to an overestimation of airway obstruction in patients of advanced age.121 For this reason, it is recommended to use international reference values that are suitable for all ages and allow expressing the results as deviations from the mean (Z-score), establishing the lower limit of normality (LLN) at -1.64.122,123

Figure 1.

Diagnostic algorithm.

**In children, a 12% increase is sufficient to consider this test as positive, even if <200ml. **In case of a negative bronchoconstriction test, a diagnosis of eosinophilic bronchitis should be considered. ***Alternatively, inhaled glucocorticoids at very high doses, 1500 – 2000μg of fluticasone propionate, 3 or 4 times a day for 2-8 weeks may be used.

(0.43MB).

A reduced FEV1 value confirms the obstruction, helps to establish its severity, and indicates a greater risk of exacerbations.124 On the other hand, many patients with asthma may show spirometric values close to the reference range or even a non-obstructive (restrictive) pattern due to air trapping.

For the bronchodilation test, the administration of 4 successive/puffs of 100μg of salbutamol, or its equivalent, using a pressurized inhaler with spacer and repeating spirometry after 15minutes is recommended. A response is considered to be positive (or significant bronchodilation) when there is a ≥ 12% and a ≥ 200ml increase in FEV1 from baseline (table 15)121 or >10% of the theoretical value of FEV1 or FVC.123 An alternative criterion for bronchodilation is an increase of the peak expiratory flow (PEF) of >20%.125 Reversibility can also be identified as an improvement in FEV108 or PEF after 2 weeks of treatment with systemic glucocorticoids (prednisone 40mg/day or equivalent) or 2-8 weeks of inhaled glucocorticoids (1500-2000mg/day of fluticasone propionate or equivalent).126 Although reversibility of bronchial obstruction is a typical characteristic of asthma, it is not present in all patients.

Table 15.

Reversibility and daily variability criteria recommended for the diagnosis of asthma.

Reversibility  Post-Bd FEV1−pre-Bd FEV1≥200 mlandPost-Bd FEV1−pre-Bd FEV1pre-Bd FEV1×100≥12% 
Daily variability  Maximum PEF−minimum PEFMaximum PEF×100Variability ≥ 20% during ≥ 3 days per week, in a 2-week recording 

FEV1: forced expiratory volume in one second; PEF: peak expiratory flow; Bd: bronchodilation

Variability, or excessive fluctuation of pulmonary function over time, is important for the diagnosis and control of asthma. The most widely recommended daily variability index is the PEF amplitude in relation to the averaged mean over at least 1-2 weeks (table 15) and recorded before the use of medication.127 A PEF variability ≥ 20% is diagnostic of asthma.128

Bronchial hyperresponsiveness is the terms used to define an excessive narrowing of the bronchial lumen in response to physical or chemical stimuli that usually only cause a small or negligible reduction of the airways.129 The identification of this exaggerated response to a bronchonstrictor by means of a non-specific challenge test may be useful in patients with clinical suspicion of asthma and normal pulmonary function. Direct agents, such as methacholine or histamine, or indirect agents, such as adenosine monophosphate, mannitol or hypertonic saline solution can be used.130 Indirect agents show a better relationship with inflammation and a higher sensitivity to the effect of glucocorticoids.131 In addition, mannitol offers the advantage of being administered via a dry power inhaler.132

The analysis of bronchial hyperresponsiveness is performed in terms of sensitivity or threshold, determining the dose or concentration that produces a 20% decrease in FEV1 compared to the post-diluent value.129,133 Recently, it has been recommended, in the case of methacholine, to use the cumulative dose of methacholine that reduces FEV1 by 20% (PD20) compared to the value obtained after diluent administration.134 This type of bronchial provocation has high sensitivity but limited specificity,135 making it more useful for excluding rather than confirming the diagnosis of asthma. Bronchial hyperresponsiveness is also present in other conditions such as allergic rhinitis, COPD, bronchiectasis, cystic fibrosis, or heart failure. The mannitol test is considered to be positive when a 15% fall in FEV1 from baseline (PD15) occurs or when there is an incremental decrease of FEV1 of ≥ 10% between two consecutive doses.129 This test is more useful to confirm the diagnosis of asthma (particularly in cases of exercise-induced bronchoconstriction) because its specificity is >95%, although its sensitivity is 60%.

Fractional exhaled nitric oxide (FENO) is a non-invasive measure of bronchial inflammation associated with the allergic-T2 phenotype (see section 7.3) and is partially related to eosinophilic inflammation. Although both FeNO and eosinophils are part of the T2 inflammatory cascade, these two biomarkers are regulated by different inflammatory pathways. The determination procedure of FENO has been standardized,136 and the recently recommended cutoff point is >40 ppb in adults who are not taking glucocorticoids.115,137 It achieves high sensitivity and specificity for the diagnosis of asthma in non-smoking patients not using inhaled glucocorticoids,138 especially when associated with reduced FEV1.139 However, a normal FENO value does not exclude the diagnosis of asthma, particularly in non-atopic individuals.140

2.3Pulmonary function in children

Although most children with asthma have FEV1 values within the reference range,140,141 respiratory function tests are essential for establishing the diagnosis of asthma.142 They contribute decisively to the diagnosis, although their normality does not exclude the diagnosis of asthma and, for this reason it should be performed periodically. However, they do not sufficiently discriminate the level of severity.143

With the appropriate method, reliable forced spirometry can be obtained in children from the age of three. Above the age of 5-6, the functional diagnosis of asthma is similar to that in adults. In children, FEV1/FVC ratio correlates better with asthma severity than FEV1.131,144 The availability of international reference values suitable for all ages,122all ages equations, allows to express the results as deviations from the mean (z-score), establishing the LLN at -1.64. In children, obstruction is defined by an FEV1/FVC ratio<LLN (lower limit of normality).

A bronchodilation test is considered positive when the increase in FEV1 as compared with baseline value is equal or higher than 12% or 9% in relation to the predicted value.145,146 The ERS/ATS proposes for the general population a change of FEV1 greater than 10% of the predicted value.123

As children can exhale all the air in 2-3seconds, an expiration lasting this amount of time may be considered valid provided its validity can be confirmed by an expert's visual inspection of the correctness of the maneuver.147 Less strict reproducibility criteria are also acceptable: 100ml or 10% of FEV1.148

The FEF25-75% value does not provide any relevant information and therefore does no contribute to clinical decision-making.149

If diagnosis is uncertain, methacholine and exercise challenge tests may be of special interest in children, since exercise challenge test is relatively easy to perform, reproducible and has a high specificity for diagnosing asthma, although its sensitivity is low.150

The algorithm shown in fig. 2 is useful to establish the diagnosis of asthma in children.

Figure 2.

Diagnostic algorithm for asthma in children.

Positive bronchodilation test (BDT): increase of FEV1 >12% as compared with base.

(0.44MB).

Between 3 and 5 years of age, it is indispensable to use the adequate methodology and appropriate reference values and do not extrapolate values of older children.151–153 Since these children may occasionally have expiration times lower than 1 second, the most useful value would be FEV0.5 or FEV0.75 rather than FEV1.154 In this age segment, the normal FEV1/FVC value would be greater than 90%.

As for the use of the bronchodilator test at this age, the cut-off point for both FEV1 and FEV0.5 or FEV0.75 remains to be determined.155,156 Other tests that may be useful in the management of preschool children with asthma include forced impulse oscillometry (IOS),157–159 the measurement of airway resistance using the interrupter technique (Rint), the tidal flow-volume curve or measurement of airway resistance by plethysmography.

Any of these techniques must be adapted to ATS/ERS guidelines on pulmonary function in preschool children.154 For children under 2 years of age, the rapid thoracoabdominal compression is the most widely used technique.

To perform reliable pulmonary function tests in children, particularly in those younger than 5-6 years of age, it is essential to have nursing staff specifically trained in these techniques as well as laboratories adapted for children.

The measurement of FENO also allows assessing the degree of bronchial inflammation in the child.160 The evaluation of FENO in young children is not relevant for predicting a diagnosis of asthma at school age.161 The diagnostic reliability of FENO in asthma is compromised by the wide confidence intervals of this measurement and the overlapping of FENO values between children without asthma and atopic dermatitis.

Cut-off points above 35 ppb have been suggested to be considered as positive,162,163 but values above 25 ppb in a child with compatible symptoms may support the diagnosis of asthma.143

Regarding its usefulness in the follow-up and adjustment of treatment, its benefits could not have been demonstrated. At follow-up, it is important to know the best value of the patient since therapeutic decisions should be based on variations regarding this optimum value.164 Treatment with inhaled glucocorticoids reduces FENO concentration, so that measurement of FENO may be a predictor of response.165 In some cases (particularly in the most severe ones), upward changes from the optimal value may be indicative of the risk of future exacerbations.166

Although potentially useful as guidance, the available evidence does not confirm its reliability to evaluate adherence to ICS treatment.

FENO can be determined in young children by the multiple breath-exhalation technique, with reference values having been established for the age between 1 and 5 years.118 In this age segment, although some studies have shown an association between high FENO levels and the risk of asthma,167,168 this correlation has not been clearly established.

In general, there is no consistent evidence to recommend the routine use of FENO in the follow-up of children with asthma, and its use should be restricted to the specialized consultation setting.169

Its use for the adjustment of treatment should be complementary to clinical and functional evaluation, and in no case should be considered as a single test.168,170

2.4Allergy evaluation

The aim of allergy testing is to determine the presence of sensitization to aeroallergens that may influence the development of the allergic asthma phenotype or to trigger exacerbations. These tests can be performed in any patient with asthma regardless of their age. The anamnesis helps to evaluate personal and family history of atopy (rhinoconjunctivitis, atopic dermatitis, food allergy) and the relationship between symptoms and allergen exposure. To make a diagnosis of allergic asthma, in addition to sensitization to inhaled allergens, it is necessary to demonstrate the clinical relevance of the results obtained171 (fig. 3).

Figure 3.

Allergy evaluation: the presence of a concordance between the clinical history and results of the allergy study is necessary to establish the diagnosis of allergic asthma.

(0.25MB).

The Intradermal puncture testing orprick test172 with standardized extracts (table 16) is the method of choice for its high sensitivity, low cost and immediately available results. It is necessary to consider the variables affecting the results (drugs, dermographism, etc.) and to have experience in the interpretation of results (false positives by cross-reactivity).173

Table 16.

Standard battery of aeroallergens used in intraepidermal skin tests or prick test.*

Mites  Dermatophagoides pteronyssinus/farinae Lepidoglyphus destructor, Blomia tropicalis 
Dander  Cat, dog 
Pollens  Grasses, Olea europaea, Cupressus spp, Platanus spp, Salsola kali, Parietaria judaica, Artemisia vulgaris 
Fungi  Alternaria alternata, Aspergillus fumigatus 
*

Other extracts can be added according to environmental exposure (such as professional allergens) or geographic prevalence.

The specific IgE against complete aeroallergens, with the same meaning than prick testing, has a lower sensitivity and a higher cost.174 The specific IgE against allergenic components allows distinguishing between primary sensitization and cross-reactivity,175 and in polysensitized patients improves the selection of the composition of specific immunotherapy with allergens.176

The specific bronchial challenge test may be useful when a discrepancy exists between the clinical history and the results of sensitization, although it is not recommended as a routine procedure and should be performed by expert professionals.

2.5Classification of severity in adults

Asthma has usually been classified according to its severity, although both the definition and assessment of severity has changed over time.113,120,177 Severity is an intrinsic property of asthma that reflects the intensity of its pathophysiological abnormalities.178

The classification of asthma according to clinical and functional parameters has been traditionally divided into four categories: intermittent, mild persistent, moderate persistent and severe persistent.113,120,177

It should be kept in mind that asthma severity involves both the intensity of the process and its response to treatment.179,180 Severity is usually evaluated while the patient is being treated and it is classified according to the need for maintenance therapy to achieve control of symptoms and exacerbations179,180 (table 17).

Table 17.

The classification of asthma severity when it is well-controlled with treatment (stratified by steps).

Severity  Intermitent  Persistent
    Mild  Moderate  Severe 
Minimal treatment requirements to maintain control  Step 1  Step 2  Step 3 or Step 4  Step 5 or Step 6 

It is not necessarily a constant characteristic of asthma, as it can vary over time (months or years), so that periodic reassessment of severity is required.

The majority of the asthmatic population suffers from intermittent or mild persistent asthma.181,182 These seemingly non-severe forms of the disease should not underestimate their inflammatory nature.183,184 The absence of symptoms in mild and intermittent asthma requires a correct clinical and functional evaluation of the patient for accurate classification and subsequent adjustment of treatment.

2.6Control and measuring methods

Asthma control is the extent to which disease manifestations can be either absent or maximally reduced by therapeutic interventions, and treatment goals are met,178,180 largely reflecting the adequacy of treatment (fig. 4).

Figure 4.

Relationship between severity and control of asthma. The level of control reflects to a large extent the appropriateness of treatment. Some patients suffer from difficult-to-control asthma (DCA).

Modified from Osborne, et al.185.

(0.11MB).

Asthma has been arbitrarily classified according to the degree of disease control in: well-controlled asthma, partially controlled asthma and poorly controlled asthma, based on the criteria shown in table 18.113 Some asthma patients may show a good control of both symptoms and pulmonary function, while simultaneously experiencing frequent exacerbations, whereas some other patients have daily symptoms and very few exacerbations.

Table 18.

Classification of asthma control in adults.

  Well controlled(all of the following)  Partially controlled (any measure in any weel)  Poorly controlled 
Daytime symptoms  None or ≤ 2 days a month  >2 days a month   
Limitation of activities  None  Any   
Nighttime symptoms/awakenings  None  any   
Need for reliever (rescue) medication (short-acting β2- adrenergic agonist)  None or ≤ 2 days a month  >2 days a month  If ≥ 3 characteristics of asthma partially controlled
Pulmonary function FEV1  ≥ 80% predicted value or z-score (-1.64)  <80% predicted value z-score (-1.64) 
PEF  ≥ 80% better personal value  <80% better personal value   
Exacerbations  None  ≥ 1/year  ≥ 1 in any week 

FEV1: forced expiratory volume in one second; PEF: peak expiratory flow.

Thus, when trying to minimize the clinical expression of asthma two major aspects should be borne in mind:180 on the one hand, the day-to-day disease manifestations (current control) and, on the other side, its future consequences (future risk), as shown in Figure 5.

Figure 5.

Domains and risk factors that determine the degree of asthma control.

*Evaluate risk factors.

(0.21MB).

Within the current control domain, control would be defined by the presence of daytime and nighttime symptoms; the frequent use of rescue medication for symptomatic relieve; maintenance of pulmonary function within or close to normal limits; the absence of limitations of daily living activities, including family, social, work or school activities, and physical exercise; and finally, the fulfillment of expectations of both patients and their families regarding the quality of care received.

As for the future risk domain, control includes: the absence of exacerbations; the lack of the need of using systemic glucocorticoids, visits to emergency departments and hospitalizations; the prevention of an excessive loss of pulmonary function and the development of a fixed airway obstruction (and an anomalous lung development in the case of children); and finally, the use of an optimal pharmacotherapy with minimum or no adverse effects.

As defined in the control of asthma, a number of procedures should be used for its evaluation.186 The essential tool for assessing asthma control is the continued follow-up medical visit. In this visit, the domains of current control and future risk of exacerbations should be evaluated, together with possible presence of fixed airflow obstruction and treatment-associated adverse effects, and finally and most importantly, the adherence to treatment, including a reminder of the self-management plan and actions to be taken in case of disease decompensation, and trying to reinforce the patient-healthcare professional relationship at each visit.

In order to facilitate and standardize the evaluation of the domain of current control of asthma, different simple questionnaires and easy to be completed by the patient have been developed. The Asthma Control Test (ACT)187,188 and the Asthma Control Questionnaire (ACQ)189,190 have been validated and culturally adapted for use in Spain. Validation of the ACT questionnaire is more detailed for its use in clinical practice with well-defined cut-off points, so that a score equal to or greater than 20 is highly consistent with well-controlled asthma, between 19 and 16 with partially controlled/not well-controlled asthma, and equal to or lower than 15 with poorly controlled asthma.187,188 The minimum clinically relevant difference is 3 points.191 Also, the Spanish version of the ACQ questionnaire has been validated, with cut-off values based on actual clinical practice192,193 with <0.5 for well-controlled asthma, between 0.5 and 0.99 for partially controlled asthma, and ≥ 1 for poorly controlled asthma. However, the reliability of both questionnaires to detect poorly controlled asthma is low,194 and for this reason they should never be used as single tools to evaluate asthma control. To determine the degree of current asthma control and the future risk, the Asthma Impairment and Risk Questionnaire (AIRQ) was developed. The AIRQ is a 10-item instrument that evaluates the presence of symptoms during the previous 2 weeks and the number of exacerbations in the last 12 months;195 the Spanish version has been recently validated.196

Factors associated with the risk of exacerbations include the presence of uncontrolled asthma symptoms and history of severe exacerbations, but other factors may increase the risk of exacerbations in the absence of uncontrolled asthma or previous severe exacerbations (table 19).

Table 19.

Main risk factors for exacerbations.

– Absence of current control: ACT <20 or ACQ >1.5.– History of exacerbations: ≥ 1 severe exacerbation in the previous year or history of almost life-threatening asthma.– No use of inhaled steroids: not prescribed, poor adherence or critical errors with the use of inhalers.– Excessive use of rescue medication: ≥ 3 inhalers per year (≥ 2 puffs/day).– Type 2 inflammation: increased peripheral blood/sputum eosinophils, increased FENO.– Pulmonary function: low baseline FEV1, reversibility with the bronchodilator.– Psychosocial problems, low socioeconomic level.– Exposures: tobacco smoke, allergens, pollution.– Comorbidities: obesity, sleep apnea-hypopnea syndrome, chronic rhinosinusitis, gastroesophageal reflux, food allergy, pregnancy. 
Adapted from GINA 2019113.

Assessment of biomarkers of type 2 inflammation may contribute to stratify the patient's risk, and taking into account that peripheral blood eosinophilia197–199 or sputum eosinophilia200 as well as increased FENO in a patient treated with inhaled glucocorticoids201 are additional factors that increase the risk of exacerbations.

In the patient with severe asthma, adjustment of treatment with inhaled glucocorticoids has been recommended, taking into account results of sputum eosinophils or FENO, since this strategy is associated with a lower risk of exacerbations, although it has no effect on symptoms or pulmonary function.202

Forced spirometry is another tool that can help in the assessment of future asthma control, since a low baseline FEV1 value, in particular <60%,203 and the presence of reversibility204 have been reported as factors that increase the risk of exacerbations.

Asthma control should be evaluated at each medical visit. Once asthma treatment is started, clinical and therapeutic management of the disease should be directed toward achieving.

and maintaining control (including symptoms, exacerbations, and pulmonary function). Therefore, the degree of control will guide the decisions on maintenance treatment and dose adjustment, according to the therapeutic steps shown in the corresponding section.

2.7Remission

With the advent of biological therapy, the concept of “remission” in asthma has been reconsidered. It could be defined as the situation in which there is no disease activity, either spontaneously or as a result of treatment. Two types have been proposed: clinical remission, defined as the absence, for at least 12 months, of symptoms and exacerbations without the use of systemic steroids, in addition to optimization and stabilization of pulmonary function; and complete remission, when the patients also have no hyperresponse and bronchial inflammation.205

In clinical practice, it is possible to achieve remission without treatment, particularly in childhood-onset asthma. A study carried out in 119 children with allergic asthma, followed for 30 years, revealed that complete remission (defined according to strict criteria) was obtained in 22% of the cases, especially in those with better baseline pulmonary function or improvement in the transition to adulthood.206 In another study carried out in 200 adults diagnosed with asthma in the last year and with a subsequent follow-up of 5 years, 16% achieved remission, although with a less strict remission criterion (absence of asthma symptoms without medication for at least 1 year).207

However, the concept of “clinical remission” with treatment has its limitations. A study of 31 well-controlled asthma patients, treated with an ICS and followed up for 1 year, showed that the risk of exacerbations persisted in almost half of them. This risk was higher in those with blood or sputum eosinophilia.200 Another study conducted in 347 patients treated with mepolizumab, with a mean follow-up of 3.5 years, reported a progressive decline in pulmonary function, even reaching levels below baseline.208

The concept of “remission,” with or without treatment, should encompass the absence of clinical manifestations, hyperresponsiveness and bronchial inflammation for a prolonged period of time. However, confirmatory evidence is required for its validation. This should verify that patients in remission maintain stabilized pulmonary function and do not suffer exacerbations. At the time of writing this new version of GEMA, a broad consensus is underway to establish a definition of this concept.

2.8Control and classification of severity in children2.8.1Clinical severity

The classification of severity is different according to the moment at which asthma is evaluated: at the onset, at the time of diagnosis or thereafter once control of the disease has been achieved. In the first case, the level of severity depends on the frequency and intensity of symptoms (number of attacks and between-attack status: mainly exercise tolerance and nighttime symptoms), the need for a rescue bronchodilator and the values of respiratory function tests. In small children in whom lung function testing is not feasible, severity is only classified according to symptomatology.

Some children with asthma present symptoms intermittently, episodically, more or less frequently, while others suffer from more persistent symptoms. The type of moderate or severe asthma is determined by the frequency and intensity of the symptoms. In any case, the classification of severity is established once treatment is started, based on the medication necessary to keep the child well-controlled.

In this way, the patient who requires step 5 or 6 treatment will have severe asthma, the one who needs step 3 or 4, a moderate asthma, the one who requires step 1 or 2, a mild asthma.

Childhood asthma varies substantially over time, even during a single year, which makes its classification difficult. Most young children experience asthma symptoms during viral infections only; they may experience, therefore, moderate or severe asthma in the winter and remain asymptomatic in spring and summer seasons. In order to typify correctly a case of asthma in children, it is necessary to specify, in addition to severity, the triggering factors in the individual patient and the degree of control of asthma.

2.8.2Control

El Asthma control is defined by the extent to which clinical manifestations have declined or disappeared with the treatment prescribed.209 It includes two components: current symptom control and future risk (future consequences of such control).113

The current control of symptoms is evaluated by the presence and frequency of symptoms, both at daytime and nighttime, the need of rescue medication and the presence of some limitation for daily life activities. The criteria established to define the degree of control vary from one guideline to another, but generally it is classified as good or poorly controlled asthma, although some guidelines also introduce the concept of partially controlled.113

To facilitate symptom control evaluation, there are available specific Spanish validated questionnaires. One of these questionnaires is the Asthma Control Questionnaire in Children (CAN) (Control de Asma en Niños), with a version for 9-14 year-old children and another version for parents (2-8 year-old children). This instrument evaluates 9 questions about clinical manifestations within the last 4 weeks and is scored between 0 (good control) and 36 (poor control). A patient is considered to be poorly controlled when scores are equal to or higher than 8210 (table 20). Also available is the Childhood Asthma Control Test (c-ACT),211 validated in Spanish212,213 for 4-11 year-old children, which includes 7 questions (4 for the child

Table 20.

Asthma Control Questionnaire in Children (CAN).210

1.- In the last 4 weeks, how often have you coughed during the day without having a cold?  4.- In the last 4 weeks, how often have you had wheezing at night?  7.- When the child exercises (plays, runs, etc.) or bursts out laughing, does he/she coughs or wheezes? 
4. More than once a day3. Once a day2. 3 to 6 times a week1. Once or twice a week0. Never  4. More than once a night3. Once a night2. 3 to 6 times a week1. Once or twice a week0. Never  4. Always3. Almost always2. Sometimes1. Almost never0. Never 
2.- In the last 4 weeks, how often have you coughed at night without having a cold?  5.- In the last 4 weeks, how often have you had breathing difficulty during the day?  8.- In the last 4 weeks, how many times has he/she had to visit the emergency department because of his/her asthma? 
4. More than once a night3. Once a night2. 3 to 6 times a week1. Once or twice a week0. Never  4. More than once a day3. Once a day2. 3 to 6 times a week1. Once or twice a week0. Never  4. More than 3 times3. 3 times2. Twice1. Once0. Never 
3.- In the last 4 weeks, how often have had wheezing/whistling sounds inyour chest during the day?  6.- In the last 4 weeks, how often have you had breathing difficulty during the night?  9.- In the last 4 weeks, how many times has the child been admitted to hospital because of her/his asthma? 
4. More than once a day3. Once a day2. 3 to 6 times a week1. Once or twice a week0. Never  4. More than once a night3. Once a day2. 3 to 6 times a week1. Once or twice a week0. Never  4. More than 3 times3. 3 times2. Twice1. Once0. Never 

and 3 for the parents/caregivers). A patient is considered to be poorly controlled when the score is lower than 20 (table 21).

Table 21.

Pediatric Asthma Control Test (ACT) questionnaire validated in Spanish.212,213

The future risk evaluates the presence of risk factors for exacerbations (table 22), to develop a fixed airflow limitation (undertreatment with ICS, prematurity,214 environmental exposure to tobacco smoke, low FEV1, severe asthma, previous hospitalizations) and for suffering treatment-related side effects (frequent courses of oral glucocorticoids, high doses of ICS).113,215

Table 22.

Risk factors for asthma exacerbations in children.214,215

– At least one exacerbation in the previous year.– Previous care in the ICU or need of intubation.– Excessive use of SABA.– Persistent and/or uncontrolled symptoms.– Lack of adherence to treatment*, inadequate inhalation technique.– Low FEV1. Positive bronchodilation test.– Exposure to allergens in case of allergy/atopy.– Exposure to tobacco smoke.– Comorbidities: obesity, allergic rhinitis, food allergy.– Important psychological or socioeconomic problems.– Other: peripheral blood or sputum eosinophilia; increase of FENO in routine control visits. 
*

The ratio between the number of control medications administered and the total number of control medications prescribed is <0.5.

In addition to the control of clinical symptoms and pulmonary function, measurement of FENO has been advocated as an approach to assess the control of inflammation. Although potentially useful in some patients, FENO measurement does not seem to add relevant benefits to the aforementioned follow-up and treatment strategies.215

4.1Recommendations

2.1. Asthma should be suspected in a patient with wheezing, dyspnea (or breathing difficulty), cough and chest tightness of variable intensity and frequency.  R2 
2.2. En In case of suspected asthma, seasonal variations and personal or family history of asthma or atopy are important aspects to be considered, although none of these or none of the signs or symptoms, especially when isolated, are specific of asthma.  R2 
2.3. The diagnosis of asthma should be based on objective measures of functional involvement. Spirometry with a bronchodilation test is the diagnostic study of choice.  R2 
2.4. The diagnosis of asthma should be considered in the presence of daily variability of peak expiratory flow (PEF) >20%, or an increased fractional exhaled nitric oxide (FENO) >40 ppb in patients who have not been treated with glucocorticoids, particularly in association with reduced FEV1R2 
2.5. Non-specific bronchial challenge test should be considered to exclude the diagnosis of asthma.  R2 
2.6. Periodic spirometry testing (at least once a year) are recommended for children with asthma requiring continuous treatment.  R2 
2.7. In children, except for specialized consultation, it is not necessary to measure FENO routinely.  R2 
2.8. Allergy studies are especially indicated when aeroallergens are suspected to be involved in the development of asthma or its exacerbations, or when other associated atopic diseases are present.  R2 
2.9. The diagnosis of allergic asthma will be based on the concordance between the patient's clinical history and the results of diagnostic studies.  R2 
2.10. The severity of asthma (in adults and children) will be established according to the minimum maintenance treatment needed to achieve control. In untreated patients, the severity of asthma should be established at the beginning of treatment, with further re-evaluation once control is attained.  R2 
2.11. The severity of asthma (in adults and children) is not necessarily a constant feature and can change over time (months or years), so that periodic re-evaluation is required.  R2 
2.12. Control of asthma (in adults and children) should be evaluated at each consultation, and treatment should be adjusted to achieve and maintain control. Control has two main components that should be identified: current control and future risk.  R2 
2.13. In the objective assessment of the degree of current control of asthma (in adults and children), it is recommended using validated questionnaires for symptoms (preferably ACT in adults, and cACT and CAN in children). In the assessment of future risk of exacerbations, recommendations include questioning on previous events, spirometry, use of inhaled glucocorticoids and reliever/rescue medication, comorbidities and, in selected cases, inflammatory biomarkers (peripheral blood or sputum eosinophils and FENO).  R2 

3Maintenance treatment3.1Objectives

The main objective of asthma management is to achieve and maintain control of the disease as quick as possible, in addition to prevent exacerbations and chronic airflow obstruction and to reduce mortality at maximum. With a properly designed treatment plan, therapeutic targets (table 23) can be achieved in a large majority of patients in terms of daily symptom control (current control domain) and prevention of both exacerbations and excessive loss of pulmonary function (future risk domain).

Table 23.

Asthma treatment goals.

In the domain of current asthma control 
• To prevent daytime, nighttime and exercise-related symptoms. 
• Use of short-acting β2-agonists no more often than twice a month. 
• To maintain a normal or near-normal pulmonary function. 
• No restrictions on daily life activities and physical exercise. 
• To fulfil the expectations of both patients and their families. 
In the domain of future risk 
• To prevent exacerbations and mortality. 
• To minimize progressive loss of pulmonary function. 
• To avoid treatment-related adverse effects. 
Avoid therapeutic inertia 

To attain these objectives a global and individualized long-term strategy must be followed based on an optimally adjusted pharmacological treatment along with supervision measures, environmental control and asthma education activities.223 Pharmacological treatment should be adjusted according to the patient's degree of control, considering the most effective therapeutic options, safety and cost of the different alternatives, and taking into account the patient's satisfaction with the degree of control achieved. Patients should be periodically evaluated to determine whether objectives are being achieved. Clinical inertia and causative factors of inertia on the part of the patient, the physician and the healthcare system should be avoided.

3.1.1Pharmacological treatment

Treatment of asthma should follow an overall plan, established by consensus of the physician and the patient (and eventually by the patient's family), in which the goals, the interventions to achieve them, and the criteria for schedule modification or adaptation according to changing disease circumstances must be made clear. Distinguishing between the current control domain and the future risk domain in the control of the disease is relevant, because it has been documented that these domains may respond differently to treatment224,225. For example, some patients may have a good daily control of asthma symptoms and yet experience exacerbations, and vice versa.

Treatment should be adjusted continuously, so that the patient remains always in a well-controlled status. This cyclic treatment adjustment means that asthma control should be objectively assessed (chapter 2.6), that the patient is being treated to achieve control, and that treatment is periodically checked to maintain asthma control (fig. 6). That is, if the patient is not well controlled, treatment must be stepped up as needed in order to regain control, always taking into account the role of non-pharmacological measures, treatment adherence and risk factors susceptible to be modified.

Figure 6.

Cyclic treatment adjustment according to periodic assessment of control of asthma.

(0.2MB).

If asthma has been controlled for at least 3 months, maintenance therapy may be gradually decreased in order to determine the minimum treatment needs that are required to maintain control226. A simple scoring system (FEOS scale) that combines data of different clinical (ACT, previous exacerbations) and functional (spirometric values) variables has been developed, to determine the risk after stepping down treatment in patients with controlled asthma227.

Drugs used to treat asthma are classified as controller or maintenance medications and reliever medications, also called “rescue” medications. Controller or maintenance medications should be administered continuously during prolonged periods of time, and include inhaled glucocorticoids (ICS) or systemic glucocorticoids, leukotriene receptor antagonists (LTRA), long-acting ß2-adrenergic agonists (LABA), tiotropium and monoclonal antibodies (omalizumab, mepolizumab, reslizumab, benralizumab, and dupilumab). Chromones and sustained-release theophylline have fallen into disuse because of their lower efficacy.

Reliever medications are used on-demand for rapid treatment or prevention of bronchoconstriction, and include inhaled short-acting ß2-adrenergic agonists (SABA) (table 24) and inhaled short-acting anticholinergics (ipratropium bromide). The use on-demand of the combinations budesonide/formoterol, beclomethasone/formoterol or beclomethasone/salbutamol can also be considered reliever medications.

Table 24.

Characteristics of inhaled β2-adrenergic agonists.

  Amount per puff (μg)Time of effect (minutes)
Drug  Pressurized inhaler  Dry power  Onset  Maximum  Duration 
Short-acting           
Salbutamol  100  100  3-5  60-90  180-360 
Terbutalina  500  3-5  60-90  180-360 
Long-acting           
Formoterol  12  4.5-9-12  3-5  60-90  660-720 
Salmeterol  25  50  20-45  120-240  660-720 
Vilanterol  22  3-5  180-240  1.440 
Indacaterol  125*  120-240  1.440 
*

Authorized dose in asthma in combination with mometasone. There are other available doses but indicated in COPD (85μg in combination with glycopyrronium; 150 and 300μg as single active principle).

The six treatment steps (fig. 7) aimed at achieving asthma control are the following:

Figure 7.

Therapeutic steps for maintenance treatment in adult asthma.

ICS: Inhaled glucocorticoid; LABA: Long-acting β2- adrenergic agonist; LTRA: Leukotriene receptor antagonist; SABA: Short-acting β-adrenergic agonist.

aAfter confirmation of adequate treatment adherence and use of the inhaler (s). bLAMA: tiotropium or glycopyrronium. cWithout maintenance treatment. dOn-demand ICS+formoterol can be used when this maintenance combination is also used.

(0.59MB).
3.1.2Steps4.1.1.1STEP 1

Different treatment options can currently be considered for this step. A correct clinical and functional assessment of the patient is required for an adequate selection of treatment.

The association budesonide/formoterol on-demand can be used.228 In a randomized study on adult asthma patients with approximately half of patients having intermittent asthma and in which an open-label design was used to reflect clinical practice conditions,229 the use of budesonide/formoterol on-demand was superior to salbutamol on-demand in the prevention of exacerbations. In a small study of patients with intermittent asthma and increased fractional exhaled nitric oxide (FENO) in which both budesonide/formoterol and formoterol on-demand were compared, the combination showed a higher reduction of FENO levels.230 The association salbutamol/beclomethasone dipropionate on-demand can also be used.231 However, these indications are not included in the technical specifications of these drugs. In addition, cost-benefit studies have not been carried out.

Inhaled SABA (salbutamol or terbutaline) exclusively on-demand can be used in patients with occasional and mild daytime symptoms (maximum twice a month) and without nighttime symptoms.232,233 The patient should be asymptomatic between episodes and maintain a normal pulmonary function, and not having suffered from exacerbations in the previous year as well as not having risk factors for exacerbations (table 24).232

Although SABA at the recommended doses does not increase the risk of severe exacerbation or death,234 its excessive use (3 or more inhalers a year) is associated with a higher risk of exacerbations, use of healthcare services and negative impact in the patients’ health, particularly when used as monotherapy.235 Up to a third of patients abuses of this medication, being a phenomenon of worldwide distribution.236–238 SABA abuse is an indicator of poor control, which should alert on the need to start or optimize maintenance treatment with ICS, along with the rest of the therapies used in asthma.235

The use of inhaled SABA on-demand more than twice a month for the treatment of symptoms (independently of its preventive use before exercise) or having been suffered from exacerbations in the last year or a FEV1 <80% indicates an inadequate control of asthma and requires the use of maintenance therapy.239–241

Inhaled SABAs administered 10-15minutes before exercise are the drugs of choice to prevent exercise-induced bronchoconstriction.242

An inhaled anticholinergic as a reliever medication is only recommended in those rare cases of intolerance to SABA agents.228

4.1.1.2STEP 2

The treatment of choice at this step is an inhaled glucocorticoid (ICS) (beclomethasone, budesonide, ciclesonide, fluticasone or mometasone) at low doses and administered daily.243–246 In general, this is the first step for most patients with persistent asthma who have not been previously treated. The usual dose ranges between 200 and 400μg/day of budesonide or equivalent. Continuous administration of ICS is the most effective maintenance treatment for persistent asthma, both for the control of daily symptoms and to reduce the risk of exacerbations240,246,247,248

Table 25 includes the approximate equipotent doses of ICS (low, medium, high) based on results of studies with efficacy/safety designs and reported in the corresponding authorized technical specifications of these compounds (available online: https://cima.aemps.es/cima/publico/home.html). However, although these results seem to be questioned by data of another pharmacodynamic study carried out in a small sample of patients,249 the evidence is insufficient to make changes250 and further complementary studies are warranted.

Table 25.

Approximate potency of ICS (based on results of efficacy/safety studies).

  Low dose (μg/day)  Medium dose (μg/day)  High dose (μg/day) 
Budesonide  200-400  401-800  801-1,600 
Beclomethasone dipropionate  200-500  501-1,000  1,001-2,000 
Extrafine beclomethasone*  100-200  201-400  >400 
Ciclesonide  80-160  161-320  321-1,280 
Fluticasone propionate  100-250  251-500  501-1,000 
Fluticasone furoate  92  184 
Mometasona furoate**       
Twisthaler®  200  400  800 
Breezhaler®††  62.5  127.5  260 
Breezhaler®††,†††  136 
*

Extrafine beclomethasone dipropionate.

**

Dosis depends on the DPI device and the dry power formulation for inhalation. Equivalence between presentaions should be considered when switching the device that contains M.

Asmanex Twisthaler® (MF as the single component).

††

Atectura/Bemrist Breezhaler® (double combination of mometasone/indacaterol, MF/IND).

†††

Enerzair/Zimbus Breezhaler® (triple combination of mometasone/indacaterol/glycopyrrium, MF/IND/GLY).

Two clinical trials showed that a strategy of using a combination of budesonide/formoterol in a single inhaler on-demand compared to continuous ICS treatment in mild persistent asthma, was not inferior in preventing exacerbations (the rate of which was similarly low); however, it was inferior in the maintenance of asthma control and in the increase of pulmonary function.251,252 In a randomized open-label study,229budesonide twice a day plus salbutamol on-demand and budesonide/formoterol on-demand were similar regarding annual exacerbation rates.

Also, a similar result with beclomethasone/salbutamol has been observed.231

Results of the aforementioned studies may provide indirect evidence of a possible indication of the combinations of low dose ICS with LABA or SABA (e.g. budesonide/formoterol, beclomethasone/formoterol or beclomethasone/salbutamol), administered exclusively on-demand, in the treatment of step 2 in patients with low treatment adherence and in which specific educational interventions have been unsuccessful. However, no studies have been specifically designed to assess this therapeutic indication.

At this level, an alternative treatment includes leukotriene receptor antagonists (LTRA) or anti-leukotrienes (montelukast and zafirlukast),253,254 although ICS are more effective for long-term treatment.253 Patients who are well controlled on ICS at low doses fail to maintain the same level of asthma control with montelukast.255

LTRA would be particularly indicated as alternative drug in patients who are unable or unwilling to receive ICS or have adverse effects with ICS, have difficulties with the inhaler technique, or suffer from concomitant allergic rhinitis.256,257

In patients who have not previously received maintenance treatment with ICS, the combination of ICS at low doses and LABA as initial treatment as compared with ICS at low doses, improves symptoms and pulmonary function but has a higher cost and it does not reduce the risk of exacerbations.258

4.1.1.3STEP 3

First-line treatment at this step is a combined inhaled treatment with ICS at low doses and a LABA (salmeterol, formoterol, vilanterol or indacaterol),259–264 which can be administered using a single device (preferred option) or separate inhalers. By using this combination a more pronounced reduction of symptoms, improvement of pulmonary function, and reduction of exacerbations and use of reliever medications is obtained as compared to increasing the dose of ICS. However, an appropriate individualized risk/benefit assessment for both strategies is required.

Treatment with LABA should always be accompanied by an ICS. LABA agents must never be used as monotherapy because of a higher risk of hospitalizations and life-threatening exacerbations.265,266 ICS/LABA combinations commercialized in Spain include: fluticasone propionate with salmeterol, budesonide with formoterol, beclomethasone dipropionate with formoterol, fluticasone propionate with formoterol, fluticasone furoate with vilanterol267 and indacaterol268 with mometasone.269

Formoterol is a rapid-onset LABA. For this reason, if budesonide/formoterol or beclomethasone/formoterol combinations are chosen, they can be used as both maintenance and reliever therapy (MART strategy). This strategy leads to reduced exacerbations and a better asthma control, despite requiring a lesser amount of ICS.242,270–277 It may be assumed that other ICS combinations (fluticasone propionate) with formoterol may be effective as MART strategy, although there is no evidence of its use as maintenance and on-demand treatment and this indication is not included their technical specifications.

In any case, MART therapy always should be administered using a single inhaler device.

A further option at this step includes increasing ICS doses up to medium doses, but this approach is less effective than adding a LABA.278–280 Alternatively, ICS at low doses associated with a LTRA may be used. This option has been found to be superior to IGS monotherapy and although it is not as effective as the IGS and LABA combination, has an excellent safety profile.281–284 However, the addition of an LTRA does not appear allowing to reduce the ICS dose.285

4.1.1.4STEP 4

The first-line treatment at this step is the combination an ICS at medium doses with a LABA.258,260,262,286,287

For patients who have had at least one exacerbation in the previous year, the combination of a ICS at low doses (budesonide or beclomethasone) and formoterol, using the MART strategy, is more effective in reducing exacerbations than the same dose of an ICS and LABA in a fixed schedule, or higher doses of ICS.277,278

Alternatively, the combination of an ICS at medium doses with a LTRA can be used, although the addition of LABA to the ICS is more effective in preventing exacerbations, control of daily symptoms and improving pulmonary function.280 In patients with uncontrolled asthma despite the aforementioned treatment, triple therapy with ICS at medium doses, LABA and LAMA (tiotropium or glycopyrronium) in a single inhaler288 or different inhalers289 may be considered. However, this option has not been compared with the standard strategy of increasing the doses of ICS in the ICS+LABA combination of proven efficacy for preventing severe exacerbations, so that adequate studies are needed to define the position of the triple therapy in this therapeutic step.290

4.1.1.5STEP 5

The next step consists of increasing the dose of ICS up to a high dose in combination with LABA.260,262,291 ICS at medium and high doses are usually administered twice daily, although a greater therapeutic efficacy can be achieved with budesonide by increasing the dosing frequency up to 4 times a day.292

Other drugs can be added for maintenance therapy, with a subgroup of patients improving with the addition of LTRA.293,294

In patients insufficiently controlled with the combination of an ICS at high doses and LABA, who show post-bronchodilator FEV1/FVC ≤ 70%,295 the addition of tiotropium (in different inhalers) or glycopyrronium (in a single inhaler [pMDI Modulite© and Breezhaler©]), provides an improvement of pulmonary function and a reduction of exacerbations.288,289,295–299 With the same indication, in Latin America and the United States, there is another triple combination of a GCI/LABA/LAMA (fluticasone f./vilanterol/umeclidinium) (approved by the FDA), after demonstrating a significant improvement in lung function;300 however, this combination is not available in Europe for the treatment of asthma (it has been rejected by the EMA), as it has not shown a significant reduction in exacerbations.300

Macrolide antibiotics, particularly azithromycin administered 3 days/week for several months, may play a role as an add-on medication in patients with severe non-eosinophilic asthma and frequent exacerbations,301,302 as well as in eosinophilic asthma303 (see chapter 7).

4.1.1.6STEP 6

For asthma patients who remain uncontrolled and with frequent exacerbations, the addition of biologic drugs should be considered after a specialized evaluation and according to the endophenotype of the patient.

In cases of uncontrolled severe allergic asthma, the anti-IgE monoclonal antibody (omalizumab) by the subcutaneous route can be added, which improves daily symptoms and decreases exacerbations,304–307 increasing the overall control of the disease (see chapter 7).

In patients with uncontrolled severe eosinophilic asthma, independently of the presence of allergy, biologic drugs targeting the interleukin-5 (IL-5) pathway can be used. Currently, anti-IL-5 monoclonal antibodies, mepolizumab and reslizumab, and the anti-IL-5 receptor a chain (IL-5Ra), benralizumab, are approved as additional treatment of eosinophilic uncontrolled severe asthma (severe refractory eosinophilic asthma)308–314 (see chapter 7).

Dupilumab is a human monoclonal antibody directed against the interleukin-4 receptor subunit a (IL-4Ra) of IL-4 that blocks the effects of IL-4 and IL-13 is approved as additional treatment in patients older than 12 years of age with uncontrolled severe asthma with increased eosinophils and/or FENO (see chapter 7).

The human monoclonal antibody directed against thymic stromal lymphopoietin (TSLP) is authorized as an add-on medication of T2 and non-T2 uncontrolled severe asthma (see additional information in chapter 7).315,316

In cases in which the administration of biologic agents has failed, the indication of endobronchial thermoplasty may be considered317 (see chapter 7).

The last therapeutic option when all other alternatives have failed is the administration of systemic glucocorticoids (always used at the lowest effective dose and for the minimum period of time possible)318,221 even though they are also associated with adverse effects, occasionally serious (see chapter 7).

3.1.3Inhalers and nebulizers

Inhaled therapy is the preferred administration route for the treatment of asthma as it acts directly on the lungs, delivers a greater amount of drug into the airways, elicits a rapid response and is associated with few or no systemic effects.222,319–323

The main disadvantage of this route is the difficulty of the inhalation technique of the different devices.216,324–326

Currently available inhalation devices include: the conventional pressurized inhaler (pMDI) and the Modulite® system, which can be used with or without a spacer, the breath actuated inhaler (BAI) k-haler® and Easy-breathe®, the soft mist inhaler (SMI) Respimat®, the dry powder inhalers (DPI) (Accuhaler®, Aerolizer®, Breezhaler®, Easyhaler®, Ellipta®, Forspiro®, Genuair®, Handihaler®, Nexthaler®, Spiromax®,

Turbuhaler®, Twisthaler® and Zonda®) and the nebulizers (jet, ultrasonic or vibrating mesh). Each of them has their own technical characteristics that should be considered when prescribed (table 26).324

Table 26.

Aerodynamic properties provided by inhalers (based in part on Giner 2016).216

  Pulmonary deposition (%)Oropharyngeal deposition (%)MADM 
  In vivo  In vitro  In vivo  In vitro  (μm) 
pMDI           
- Conventional pMDI  7.8-34  53.9-82.2  1.4-8 
- Conventional pMDI with spacer  11.2-68.3  31.2  40  2-3.2 
- pMDI autodisparo  50-60  30 
- Modulite®  31-34  33-58  1-2 
- Alvesco®  50-52  32.9 
- pMDI Aerosphere*  37.7217,218  58-61219  62  3-3.2220 
BAI SMI           
- k-haler®  44.7221  23-30 
DPI (by alphabetical order)           
- Respimat®  40-53  19.3-39 
- Accuhaler®  7.6-18  15-30  3.5 
- Aerolizer®  13-20  21.7-28  73  1.9-7.9 
- Breezhaler®  36  39  45  2.8 
- Easyhaler®  18.5-31  29  2.2-3.0222 
- Ellipta®  2-4.8 
- Genuair®  30.1  54.7 
- Handihaler®  17.8  17.3-22  71  3.9 
- Inhalador Ingelheim®  16  59 
- Nexthaler®  56  43  1.4-1.5 
- Spinhaler®  11.5  30.9 
- Turbuhaler®  14.2-38  28  53-71.6  57.3-69.3  1.7-5.4 
- Twisthaler®  36-37  2-2.2 

MADM: mean aerodynamic diameter mass; BAI: breath-actuated inhaler; DPI: dry powder inhaler; pMDI: pressurized metered-dose inhaler; SMI: soft miss inhaler. The comparison of values among devices should be considered with caution because of differences in the methods and drugs used for estimating the corresponding values, as well as differences in human studies, which were performed in diverse clinical settings (healthy and ill subjects with different diseases and degrees of severity), inspiratory flows and ages.

All inhaler devices if correctly used provide en efficient deposition of the drug in the lung.320

The use of spacers is recommended for pMDI. Spacers circumvent coordination issues, improve the distribution and the amount of drug reaching the bronchial tree, reduce the deposition of drug particles in the oropharynx, decrease cough and the possibility of oral candidiasis (that may be associated with the use of ICS), decrease systemic bioavailability and, hence, the risk of systemic effects.327–330

Healthcare professionals involved in the care of patients with asthma should know the inhalation techniques of each of the devices; knowledge, however, is still insufficient.331,332

Given that the proper use of inhalers is a crucial aspect in the treatment of patients with asthma, all healthcare professionals involved, doctors, nurses and pharmacists especially those from the community due to their accessibility, should be involved in the instruction and review of the inhalation technique.333–340

The patient should be periodically trained and controlled in the use of the prescribed inhaler device, explaining its characteristics, the appropriate technique, demonstrating how it is used, then asking the patient to perform the maneuvers (with a placebo device) and correcting the possible mistakes.323,341–343

Whenever pharmacologically possible, a single type of inhaler device should be used.344,345

After the instruction in the use of the device, the patient should be given a brochure with description of the technique and receive information on how to find demonstration videos showing the correct inhalation technique.321,322,216,342,343

It is important to take advantage of control visits, performance of pulmonary function tests and admissions to the hospital to check the patient's inhalation technique.342

Hydrofluorocarbon (HFC) propellants in current pressurized cartridge inhalers (pMDIs) contribute to global warming as greenhouse gases.346,347 New less polluting HFC propellants are being investigated. Until these are available, the use of dry powder or mist devices may be preferable in new patients >6 years or with inspiratory flow >30 L/min. Changing the inhaler, for non-clinical reasons, could pose a risk of disease deterioration and/or promote low therapeutic adherence (including poor inhalation technique with the new device). Replaced inhalers and cartridges will be deposited at the convenient point of the integrated packaging management and collection system (SIGRE) of pharmacies for correct recycling.

3.2Other treatments3.2.1Smoking and environmental control

Smokers with asthma have more severe symptoms, a poorer response to ICS treatment, even in patients with mild asthma,348 and an accelerated loss of pulmonary function,349,350 so that a step-up in treatment is often required.351 The proportion of asthmatic smokers is high and similar to that in the general population. Moreover, since longitudinal studies have found a relationship between tobacco use and asthma in both adults and adolescents,352 the primary objective in environmental control is getting the patient to stop smoking. To this purpose, smokers should receive full information of the most appropriate quitting methods.353 Exposure to both environmental contaminants and passive smoking aggravates the course of asthma and constitute a risk factor for asthma development in childhood.354 Administrative regulations banning smoking in public spaces are being having a highly positive impact.355,356 Also, passive exposure to smoke of electronic cigarettes has been related with a higher risk for exacerbations and asthma symptoms,357,358 and active exposure to severe effects of respiratory health,359 so that vaping cannot be recommended as a method to quit.

Some asthma patients, particularly those with sinonasal polyposis, may experience exacerbations when administered acetylsalicylic acid or other non-steroidal anti-inflammatory drugs (NSAID). Many of these reactions are serious or even fatal,360 so that it is necessary that patients are correctly diagnosed based on evident data in the medical history (several reactions to different NSAIDs) or by means of an oral challenge test which, in severe cases, can be replaced with bronchial or nasal inhalation challenge testing.361,362 This issue is more comprehensively explained in chapter 8.5 (acetylsalicylic acid-exacerbated respiratory disease). These patients, however, among their environmental measures, should avoid the use of analgesic or anti-inflammatory treatments with drugs of the NSAID therapeutic class.

Specific recommendations should be considered in allergic asthma, once sensitizations to different allergens had been confirmed in each patient. The most effective measures are those enabling a dramatic decrease of exposure levels, such as those applicable to many patients with occupational asthma (job change) or asthma due to animal dander (removal of animals from the patient's home) or cockroach allergy (wise use of pesticides).363–368

Isolated individual interventions, such as the use of mattress covers or acaricides have not shown to be effective, not even in reducing exposure levels.369–371

However, in a recent randomized study, the use of impermeable bed covers was effective for preventing exacerbations in children and adolescents with allergic asthma triggered by dust mites.372

The use of combined specific measures has been associated with a significant reduction in the level of allergen exposure and, in consequence, of benefits in clinical efficacy.363,373,374 In a randomized trial of 937 patients with uncontrolled moderate to severe asthma and sensitization to at least one domestic allergen, in which combined measures were applied (impermeable covers, vacuum cleaners and air purifiers in the bedroom both with HEPA filters, cockroach disinsection plans), associated with a general education program, for one year, obtained a significant reduction in symptoms and unscheduled medical visits.363

Finally, the two more recent systematic reviews of the effect of combined interventions showed favorable outcomes.367,375

3.2.2Allergen immunotherapy

Subcutaneous immunotherapy with allergen extracts is an effective treatment in well-controlled allergic asthma with low or medium treatment levels (therapeutic steps 2 to 4), provided that a clinically relevant IgE-mediated sensitization against common aeroallergens has been demonstrated and well-characterized and standardized allergen extracts are used,376,377 avoiding complex mixtures.378,379 However, many patients with mild intermittent asthma (step1) suffer from moderate or severe allergic rhinitis concomitantly, which would justify the prescription of immunotherapy.380 Subcutaneous immunotherapy should not be prescribed to patients with uncontrolled severe asthma, because its efficacy is not well documented and a high risk of serious, even fatal, adverse reactions.379,381 For this reason, subcutaneous immunotherapy should only be prescribed by specialist physicians with experience in this type of treatment and administered in centers equipped with the basic resources for the immediate treatment of a possible adverse reaction.

The search for safer and more convenient alternatives for the patient has led to investigate the efficacy of sublingual immunotherapy. Some systematic reviews conclude that oral immunotherapy with capsules or lyophilized extracts can significantly reduce clinical manifestations and the use of rescue medication in children, adolescents and adults with allergic asthma.377,382–384

Most clinical trials showing clinical efficacy were performed with well-characterized extracts and at much higher doses than those usually prescribed for subcutaneous immunotherapy. The tolerability profile of sublingual immunotherapy is optimal and fatal reactions have not been reported.377,384

Sublingual immunotherapy with an oral lyophilized mite extract when added to regular pharmacological maintenance treatment is able to reduce the number of moderate to severe exacerbations385 and to improve the control of the disease, with a very favorable safety profile. Therefore, its use is recommendable for adult patients with moderately controlled or partially controlled asthma.380

When there are several immunotherapy alternatives available, priority should be given to the use of those that are considered registered drugs with well-established efficacy, safety, and quality data.

At the moment, no comparative studies on the cost-effectiveness of immunotherapy versus conventional pharmacotherapy are yet available, and they are not likely to be performed since their complex design makes them still unfeasible.

However, immunotherapy is not only useful in controlling disease manifestations, but it also offers additional advantages over pharmacotherapy, such as the maintenance of clinical benefits for several years after treatment discontinuation,386,387 a decrease in the risk of developing asthma in patients with allergic rhinitis,387,388 or the occurrence of new sensitizations in monosensitive patients.389 Finally, immunotherapy has been found to be cost-effective in comparison with pharmacotherapy alone in patients with the coexistence of allergic rhinoconjunctivitis and asthma.390,391

3.2.3Influenza and pneumococcal vaccination

Influenza392,393 and pneumococcal394,395 vaccines have not been shown to be effective in preventing asthma exacerbations.

However, since it is a cost-effective approach, and due to the high risk of complications in patients with chronic diseases396,397 and a higher risk of therapeutic failure in children,398 annual influenza vaccination should be considered in patients with moderate and severe asthma, both in adults and children. Similarly, and given that asthma population have a high risk of invasive pneumococcal disease,399,400 different international401 and national402 consensus documents as well as the National Healthcare System403 recommend the administration of pneumococcal vaccine in patients with severe asthma.

3.3Education3.3.1Objectives

Education of asthma patients is an essential component of treatment, because reduces the risk of exacerbations, improves quality of life and decreases healthcare costs,262,404 thus becoming an indispensable part of the overall management of the disease.228,404–410 The main goal of education is to provide patients with the knowledge and skills they need to improve self-care and treatment compliance. This results in a better adherence to treatment and, in consequence, in an optimal control of the disease. In addition, education promotes patient's self-control of asthma. Self-control is the situation in which the patient monitors their symptoms and applies self-management following a plan agreed with his/her doctor. Self-control supported by a healthcare professional reduces the number of consultations and exacerbations, and improves quality of life without increasing costs.411,412

3.3.2Knowledge and skills

From a practical point of view,413 education should consider two major aspects: transmission of knowledge and acquisition of skills and competences (table 27). Regarding the information that the patient should receive about asthma, their needs, previous knowledge, beliefs,414 age, severity of asthma, and the degree of involvement necessary in his/her self-control and treatment should be considered.

Table 27.

In Information and basic skills that should be learned by a patient with asthma.

1. To know that asthma is a chronic disease requiring continuous treatment even if symptoms are absent. 
2. To know the differences between inflammation and bronchoconstriction. 
3. To be able to differentiate between inflammation “controller” drugs and obstruction “reliever” drugs. 
4. To recognize the symptoms of the disease. 
5. To use inhalers correctly. 
6. To identify triggers and avoid triggering factors as much as possible. 
7. To monitor symptoms and peak expiratory flow (PEF). 
8. To recognize the signs and symptoms of asthma worsening (loss of control). 
9. To act in case of asthma worsening in order to prevent an attack or exacerbation. 

These interventions should include:415 self-management of symptoms or PEF monitoring, written action plans, and regular assessments of asthma control, asthma treatment and abilities of the healthcare personnel.411

Interventions without written action plans are less effective415,416 Actions that are exclusively informative are ineffective.408,416 Regarding the skills to be developed, patients will be trained in taking the prescribed medication, particularly in the technique of their inhalation devices,321,322,324,325,417 in the recognition of exacerbations and how to act early, and in the avoidance of allergenic triggers.418,419

Minimal educational interventions reduced to the essentials (mini-action plan, avoidance behaviors and revision of inhalation technique) have shown efficacy if they are administered repeatedly at follow-up visits.420

3.3.3Action plan

The education program should include establishing an action plan, which consists of a set of individualized written instructions in which asthma severity, disease control and the usually prescribed treatment are taken into account. The main objective of the action plan is the early detection of asthma worsening and the rapid adoption of measures to achieve quick remission. Depending on the patient's and the physician's preferences,421–423 the level of control on which the action plan should be based can be assessed in terms of severity and frequency of asthma symptoms, as well as through daily home recording of PEF. This plan should include two basic components:424–426 the usual treatment in situation of clinical stability of the disease and actions to be implemented in case of asthma worsening (table 28). This action plan will be reviewed at every clinical visit, either scheduled or unscheduled, as well as the time of hospital admissions or visits to the emergency department.

Table 28.

Asthma action plan.

Action plans improve the patient's quality of life, but a systematic review did not find other beneficial or detrimental effects with the use of a written action plan.427

3.3.4Treatment adherence

Patient's adherence to treatment is a critical factor for achieving and maintaining disease control. It is estimated that adherence in asthma patients is lower than 50%.428–430

Low adherence is associated with increased morbimortality as well as with a greater use of healthcare resources.431,432

Three types of patients with low adherence or non-adherence have been described: erratic (due to forgetfulness to take medication), deliberated (or intentionally non-adherence where the patient decides not to take medications) and involuntary or unwitting (due to failure in understanding the disease and/or its treatment).433,434

Treatment adherence should be evaluated at each medical visit using a reasonably validated method, such as the Test of Adherence to Inhalers (TAI) and pharmacy dispending medication or the combination of both.435–437

The education program should include the assessment of the level of adherence, promoting the appropriate corrective measures in case of low adherence and adapting them to the patient's pattern of non-adherence.

Participation of the patient in the choice of the inhaler provides greater therapeutic adherence and control of the disease. Therefore, patients should be involved in the selection of the inhaler device.330,332,344,345,438–441

Non-adherence to control medication in severe asthma can be detected by the FENO suppression test.442

3.3.5Other aspects to be considered

For education to be effective, a confidence relationship between the healthcare team and the patients should be established, so that patients can raise their doubts, concerns and fears. The healthcare provider should use a simple and understandable language towards both the patients and their relatives, ensuring that all concepts have been understood and encourage the patients to put forward their doubts and queries. Also, common objectives with the patient should be established, always based on written and individualized plans.

An appropriate agreement between the patient's opinions and expectations and his/her physician is one of the factors related to asthma control.443

Patients and their families should be encouraged to raise doubts and queries regarding the information received or emerging as a result of the medical visits, allowing sufficient time to be solved on the next visit.228

Since education is a continuous process and not an isolated event, each visit should give the opportunity to review, strengthen and increase the patients’ knowledge and skills; hence, it is indispensable that education should be agreed on and accepted by the whole healthcare team.408

Table 29 describes the educational tasks that should be undertaken at each visit. Once properly trained, the nursing and pharmacy staff should actively participate in the organization and management of education programs.334,444–446

Table 29.

Educational tasks to be implemented at each visit.

  Communication  Information  Instruction 
Initial visit  Assess expectationsAgree on objectives Discuss adherence issues  Basic concepts on asthma and its treatment  Inhalation techniqueSelf-monitoring 
Second visit  Evaluate achievements regarding expectations and objectivesDiscuss adherence issues  Reinforce information provided at the initial visit. Inform about environmental avoidance measures  Reinforce inhalation techniqueHow to avoid triggersInterpretation of recordsSelf-management plan 
Revisions  Evaluate achievements regarding expectations and objectivesDiscuss adherence issues and environmental avoidance measures  Reinforce the whole information  Review and reinforce inhalation techniqueReview and reinforce self-monitoring and the self-management plan 

Individualized discharge programs assisted by trained nursing personnel prevent readmissions due to exacerbations.447

Educational interventions carried out in the Primary Care setting reduce unscheduled visits and the inappropriate use of drugs, such as antibiotics.448

In the interventions to potentiate self-care, patients’ sociocultural differences should be considered.414

Educational interventions cannot exclusively be developed in the clinical setting. Interventions of self-care in schools or by other patients with asthma provide a better control, a reduction of exacerbations and an improvement of quality of life. Also, they can positively influence on adolescents to quit smoking.449,450

The use of telemedicine improved adherence to treatment431 through inhaler monitoring devices451 or reminder alarms.452 It also improves symptoms and decreases the use of medical care.453 Teleconsultation improves asthma control and quality of life454 (see section 9.4).

The efficacy of the patient's self-control in asthma is very positive. For interventions on the patient's self-management to be effective, it is necessary to combine the active participation of the patient, with training and motivation of professionals integrated into a healthcare system that values the self-control in asthma patients.455

Educational workshops are a useful tool as a complement to individualized care, being more profitable when performed during the periods of time when patients present more symptoms.456

The community pharmacist, due to its accessibility and frequent use by the patient, can identify poorly controlled patients especially those who abuse SABA agents or have low adherence to anti-inflammatory maintenance treatment. The community pharmacist can offer health education improving adherence, asthma control and obtaining better clinical and economic outcomes. If necessary, he/she can refer the patient to medical consultation.340,457–460

4.2Recommendations

3.1. SABA agents, when administered 10-15min before the exercise, are the drugs of choice to prevent exercise-induced bronchoconstriction.  R1 
3.2. In step 1,budesonide/formoterol, beclomethasone/formoterol or beclomethasone/salbutamol on-demand can be used, although this strategy is not approved in technical specifications and the cost-effectiveness is unknown.  R2 
3.3. First-choice treatment (step 2) is an ICS at low doses used on a daily basis. LTRA can be considered as an alternative treatment.  R1 
3.4. In step 2, an alternative could be the use of ICS at low doses with LABA or SABA (e.g. budesonide/formoterol, beclomethasone/formoterol, or beclomethasone/salbutamol) on-demand in patients with low adherence to treatment in whom a specific education had previously failed. However, this strategy is not approved in the products technical specifications and the cost-effectiveness is unknown.  R2 
3.5. For moderate persistent asthma, the first-line treatment is the combination of an ICS at low doses (step 3) or medium doses (step 4) with inhaled LABA.  R1 
3.6. For moderate persistent asthma, ICS at low (step 3) or medium (step 4) doses associated with LTRA may be considered as an alternative option.  R1 
3.7. The combination of budesonide/formoterol or beclomethasone/formoterol can be used as maintenance and on-demand treatment (reliever).  R1 
3.8. In severe persistent asthma (step 5) first-line treatment is an ICS at high doses in combination with LABA.  R1 
3.9. In patients with severe persistent asthma (step 5 or 6) uncontrolled with the combination of an ICS at high doses and LABA, with post-bronchodilation FEV1/FVC ≤ 70%, the addition of tiotropium or glycopyrronium has shown to improve pulmonary function and to reduce exacerbations.  R2 
3.10. SABA, budesinode/formoterol or beclomethasone/formoterol combinations and, in selected cases, short-acting anticholinergics (ipratropium bromide), are the drugs that can be used as reliever medications (in all steps).  R1 
3.11. Inhalation is the route of choice in the management of asthma.  R1 
3.12. All healthcare professionals taking care of asthma patients should be involved in teaching the inhalation technique and control of inhaled therapy.  R1 
3.13. The patient should participate in the selection of the inhaler device.  R1 
3.14. It is recommendable the use of a single type of inhaler or at least similar inhalers.  R2 
3.15. Patients should be trained on the inhalation technique of inhaler devices and their technique should be periodically supervised.  R1 
3.16. Smoking cessation is recommended in smokers with asthma.  R1 
3.17. In allergic asthma, specific combined measures of environmental control according to sensitization of the patient.  R2 
3.18. In well-controlled allergic asthma with low or medium treatment levels (steps 1 to 4), allergen immunotherapy is recommended when clinically relevant IgE-mediated sensitization against common aeroallergens has been demonstrate, and well standardized extracts are used.  R1 
3.19. Allergen immunotherapy should be prescribed by experienced specialized physicians. All administration of subcutaneous immunotherapy and the first use of sublingual immunotherapy should be carried out in centers with available basic resources for immediate treatment of a possible adverse reaction.  R2 
3.20. When different alternatives of immunotherapy are available, the use of those based on registered medicines with well-established efficacy, safety and quality should be priorized.  R2 
3.21. Patients with asthma should follow a formal education program of their disease. Informative actions alone have not been shown to be effective.  R1 
3.22. Patients with asthma should be provided with a written action plan in order to detect early asthma worsening and to be able to implement actions for rapid remission.  R1 
3.23. It is indispensable to determine the level of adherence to treatment in each individual patient. To this purpose, the use of validated methods such as the TAI questionnaire or electronic registry of pharmacy dispensing medicines is recommended.  R2 
3.24. Self-control interventions to be effective should combine the active participation of the patient, the healthcare professional and the healthcare system.  R1 

4Assessment and treatment of asthma exacerbations4.1Introduction and life-threatening risk factors

  • Concept: an asthma exacerbation is defined by an episode of deterioration of the baseline clinical status of a patient that implies the need of administering specific treatment.

  • Synonyms: in addition to crisis, it can receive other names such as agudization, exacerbation, or asthma attack.

  • Identification: it can be clinically identified by an increase of symptoms, need of reliever medication, or worsening of pulmonary function in comparison of usual daily variation in a given patient.461

  • Onset: depending on how fast exacerbation occurs, two types are identified: rapid-onset with progression in less than 3hours, and slow-onset (usually developing in days or weeks). The identification of the type of exacerbation is important because of differences in causative factors, pathogenesis and prognosis.462,463

Rapid-onset exacerbations develop by a mechanism of bronchoconstriction, are associated with a higher initial severity and vital risk than slow-onset exacerbations, although therapeutic response is usually more rapid and favorable. Triggering factors include inhaled allergens, drugs (NSAID or ß-blockers), food (due to food allergy, particularly milk and egg in childhood, and panallergens related to lipid transfer proteins in dried fruits, fruits and vegetables; or additives and preservatives), or emotional stress.

Slow-onset exacerbations account for more than 80% of asthma attacks attended in the emergency setting, and is mainly caused by an inflammatory mechanism, so that treatment response is slower. Slow-onset exacerbations are commonly caused by upper respiratory tract infections or a poor disease control.

  • Severity: the intensity of exacerbations is variable with some attacks occasionally showing mild or symptoms that may be undetectable by the patient, while other episodes are very severe and life-threatening.

  • Vital risk: a series of factors that increase the probability of suffering from life-threatening exacerbations have been reported. These factors are related to the characteristics of the current and past exacerbation episodes, adequate control of the chronic disease, and presence of a specific comorbidity (table 30).464–466

    Table 30.

    Risk factors for life-threatening asthma exacerbation.

    A. Related to the asthma exacerbation:1. Current exacerbation of rapid-onset.2. Previous episodes requiring medical consultation or hospital admissiona) Multiple visits to the emergency department in the previous year.b) Frequent hospitalizations in the previous year.c) Previous episodes of ICU admission, intubation or mechanical ventilation. 
    B. Related to chronic asthma disease and its adequate control:1. Absence of periodic control.2. Abuse of a short-acting β2-adrenergic agonist. 
    C. Cardiovascular comorbidity. 
    D. Psychological, psychiatric and social conditions that difficult treatment adherence: alexithymia, denial attitudes, anxiety, depression, psychosis. 

    ICU: intensive care unit.

4.2Assessment of severity

Assessment of the severity of the exacerbation episode determines its treatment (fig. 8),467 and is carried out in two steps:

  • Initial or static (pretreatment) evaluation: aimed at identifying signs and symptoms and objectively measuring the degree of airflow obstruction by determining FEV1 or peak expiratory flow (PEF) and their impact on gas exchange, in order to establish the level of severity of the exacerbation (table 31).

    Table 31.

    Assessment of severity of asthma exacerbation.

      Mild attack  Moderate attack  Severe attack  Life-threatening attack 
    Dyspnea  Mild  Moderate  Intense  Agonal breathing, respiratory arrest 
    Speech  Paragraphs  Sentences  Words  Absent 
    Respiratory rate (x’)  Increased  >20  >25  Bradypnea, apnea 
    Heart rate (x’)  <100  >100  >120  Bradycardia, cardiac arrest 
    Blood pressure  Normal  Normal  Normal  Hypotension 
    Use of accessory muscles  Absent  Present  Very evident  Paradoxical thoracoabdominal movement, or absent 
    Wheezing  Present  Present  Present  Silence on auscultation 
    Level of consciousness  Normal  Normal  Normal  Decreased or coma 
    FEV1 or PEF (reference values)  >70%  <70%  <50%  Not applicable 
    SaO2  >95%  <95%  <90%  <90% 
    PaO2 mm Hg  Normal  <80 (hypoxemia)  <60 (partial respiratory failure)  <60 
    PaCO2  Normal  <40  <40  >45 (hypercapnic respiratory failure) 

    FEV1: forced expiratory volume in one second; PEF: peak expiratory’ flow; x’: per minute; SaO2: oxyhemoglobin saturation; PaO2: arterial oxygen partial pressure; PaCO2: arterial partial pressure of carbon dioxide.

  • Dynamic (post-treatment) evaluation: aimed to measure changes obtained in the degree of airflow obstruction as compared to initial values, and to assess the need of other diagnostic studies.

Figure 8.

Therapeutic management of asthma exacerbation in adults.

FEV1: forced expiratory volume in one second; PEF: peak expiratory flow; SaO2: oxyhemoglobin saturation; pMDI: pressurized metered− dose inhaler; NEB: nebulized; i.v.: intravenous route; SGC: systemic glucocorticoids; ICS: inhaled glucocorticoid; NIMV: non-invasive mechanical ventilation; IMV; invasive mechanical ventilation; min: minute; Mg: magnesium; h: hour; μg: micrograms; 1st: first.

(0.58MB).

The objective of assessment is to determine factors described in table 31. The presence of signs of a life threatening asthma attack makes it necessary to consider the possibility of admission to the ICU.

Signs and symptoms that are not indicative of life-threatening asthma have a low clinical usefulness due to a poor correlation with the degree of obstruction and the large variability in their interpretation.468,469

The objective assessment of the degree of airflow obstruction by spirometry (FEV1) or using a peak expiratory flow (PEF) meter is crucial to ascertain the initial severity and evaluate treatment response. It is preferable to use the percentage value of the previous best value of the patient in the last two years, but if this datum is unknown, the percentage value in relation to the predicted value can be used. According to the values obtained, exacerbations are classified as mild, if FEV1 or PEF are equal to or greater than 70%; moderate, if FEV1 or PEF values range between 70 and 50%; and severe, if these values are lower than 50%. Life-threatening asthma attack is usually associated with values lower than 33%. The initial therapeutic response of airflow obstruction is the main prognostic factor in the assessment of the exacerbation episode.469–472

Measurement of oxygen saturation by pulse oximetry is easy to obtain in all patients and has a complementary role. Values lower than 90-92%, with or without supplemental oxygen therapy can be associated with hypercapnia and life-threatening crisis; therefore, in these cases, arterial blood gases analysis is indicated.473

Other complementary studies at the beginning of an asthma attack, such as chest X-rays or an electrocardiogram, are indicated in case of fever or suspicion of infection (pneumonia), pain or intense dyspnea that may suggest the presence of pneumothorax or pneumomediastinum, or when therapeutic response measured by objective parameters, is not appropriate and in case of a life-threatening asthma exacerbation.474–476

4.3Treatment

The immediate objective when treating an asthma attack is to preserve the patient's life, reverting airflow obstruction and hypoxemia as soon as possible, and thereafter to set up or review the therapeutic plan to prevent further attacks. The pharmacological treatment the usually recommended doses are shown in table 32. Treatment according to severity is shown in fig. 8.

Table 32.

Drugs and doses commonly used for treating asthma exacerbations.

Therapeutic groups  Drugs  Doses 
First-choice     
βz-adrenergic agonists  Salbutamol  pMDI+spacer: 200-800μg (2-8 puffs of 100μg/puff) every 10-15 min during the first hour 
    NEB intermittent: 2.5-5 mg every 20 min during the first hour 
    NEB continuous: 10-15 mg/hour 
Anticholinergics  Ipratropium bromide  pMDI+spacer: 80-160μg (4-8 puffs of 20μg every 10-15 min 
    NEB intermittent: 0.5 mg every 20 min 
Prednisone    Oral route on discharge: 50 mg every/24 hours (5-7 days) 
    Oral route on admission: 20-40 mg every/12 hours 
  Hydrocortisone  i.v.: 100-200 mg every/6 hours 
Inhaled glucocorticoids  Fluticasone propionate  pMDI+spacer: 500μg (2 puffs of 250μg/puff) every 10-15 min 
  Budesonide  pMDI+spacer: 800μg (4 puffs of 200μg/puff) every 10-15 min 
    NEB: 0.5 mg every 20 min during the first hour 
Magnesium sulfate i.v.    i.v.: 2 g infused over 20 min (one time only) 
Alternative in case of previous failure     
βz-adrenergic agonists i.v.  Salbutamol  i.v.: 200μg in 30 min followed by 0.1-0.2μg/kg/min 
Magnesium sulfate inhaled    NEB: 145-384 mg in isotonic solution 

pMDl: pressurized inhaler; NEB: nebulized; i.v.; intravenous route.

4.3.1Mild exacerbation

In clinical practice, it is difficult to differentiate a mild exacerbation from a transient loss of asthma control, since changes observed will be close to the normal range of variation for a given patient.461

Milder attacks can be managed at home by the patient him/herself or in primary care centers, provided a correct clinical and respiratory function assessment has been carried out and treatment response can safely be achieved within the first 2hours.

Asthma patients who have been provided with written action plans, including home PEF monitoring and how to act in case of loss of control, have an excellent and readily usable tool for managing mild exacerbations.477 In order to quickly implement the adequate measures, patients should be trained in identifying the early indicators of exacerbations and be ready to act immediately according to their assigned action plan, which should include the actions to be implemented according to the response to treatment.

The treatment schedule to be followed does not depend on the setting where the patient is being cared for. The therapeutic regimen should include the administration of short-acting ß2-agonists (SABA), such as salbutamol or terbutaline, and inhaled (ICS) or oral glucocorticoids. The addition of ipratropium bromide is not necessary for mild attacks, and antibiotics should not be routinely prescribed.

Inhaled SABA are the most effective and rapidly acting bronchodilators for treating asthma exacerbations. Salbutamol at doses of 200 to 400μg (2 to 4 puffs) with spacer is used.478,479

Treatment with salbutamol at doses of 2 puffs every 3-4hours can be continued until remission of the exacerbation episode.

If a favorable outcome is observed within the first 2hours of treatment (symptom resolution, PEF over 80% predicted or personal best value) and if this clinical response is maintained for 3-4hours, no more treatments are necessary.

The lack of response requires referral of the patient to a hospital emergency department.

The use of systemic glucocorticoids accelerates resolution of exacerbations and prevents relapses.480 Except for very mild attacks, systemic glucocorticoids should always be administered as early as possible,481,482 particularly in the following cases:

  • Pulmonary obstruction cannot be reversed with inhaled SABA.

  • The patient is already taken oral glucocorticoids.

  • The patient has treated him/herself a previous loss of asthma control with other therapeutic options without success.

  • There is a history of previous exacerbations requiring oral glucocorticoids.

The daily dose of prednisone is 0.5-1mg/kg of the ideal body weight (or equivalent doses of other steroids), up to 50mg; this dose should be maintained for 5 to 7 days, and may be discontinued without down-titration in order to achieve a quick improvement and prevent early relapses.482,483

The administration of glucocorticoids by the oral, intramuscular or intravenous route provides similar biological results, but the oral route is less invasive and cheaper.482,484–486

If response to inhaled bronchodilator treatment within the first hours is satisfactory, no hospital referral is required. Patients should be instructed on the need for adequate adherence to the treatment prescribed, their maintenance treatment plan should be reviewed, and a minimal asthma education intervention should be provided.487,488

4.3.2Moderate and severe exacerbations

The first measure consists of immediate oxygen administration, with a flow providing a saturation over 90% (95% in pregnant women or in patients with concomitant heart disease).489

In severe exacerbations with greater airflow obstruction and risk of hypercapnia, the use of oxygen with controlled FiO2 to obtain saturations around 93-95% is preferable than the use of high-flow oxygen therapy with which saturations around 100% can be achieved.489,490

In patients with severe exacerbations, the use of capnography to assess the trend to hypercapnia can be considered.491

Inhaled short-acting ß2-adrenergic agonists (SABA) are the first-line bronchodilator treatment. Both the dose and the dosing intervals should be individualized according to the choice of the administration system and the therapeutic response.

There is evidence that the use of a pressurized inhaler with spacer is the most cost-effective system;492 however, cost-effectiveness is lower in patients with very severe exacerbations.

It has been shown that the administration of SABA using a nebulizer or a pMDI inhaler with spacer has a similar clinical efficacy in terms of pulmonary function, length of stay in an emergency department, and risk of hospitalization. However, the dose when using a pMDI inhaler is lower.492–496

There is some debate as to whether nebulized treatment should be administered continuous or intermittently.497,498 A practical approach may include an initial continuous nebulization therapy to stabilize the patient followed by intermittent therapy.

There is no evidence to support the use of a route other than inhalation for the administration of bronchodilator medication.499 The intravenous route, with a very slow continuous infusion, should be used when there is no response to inhalation therapy in patients under mechanical ventilation and monitored in an ICU.

Similarly, no beneficial effects have been demonstrated when adding intravenous medication to the inhaled therapy.499

The use of parenteral epinephrine (subcutaneous or intravenous) is not indicated for treating exacerbations, except when these occur in a patient with anaphylaxis. In this case, the intramuscular administration is the route of choice because higher and more quickly plasma concentrations are obtained as compared with the subcutaneous route, as well as there is a greater safety margin.500–502

When administered in aerosol form, doses higher than 2mg, equivalent to 5mg salbutamol are required as lower doses are ineffective.503

The intravenous administration of epinephrine would only be indicated in case of cardiac arrest or in hypotensive patients who do not respond to intravenous volume replacement and multiple doses of intramuscular epinephrine.504,505

The use of ipratropium bromide during the initial phase of moderate or severe exacerbations concomitantly with a SABA is associated with a greater increase in pulmonary function (estimated by FEV1 or PEF) and a decrease in hospitalizations as compared to the use of a SABA alone.506,507

Systemic glucocorticoids accelerate the resolution of asthma attacks and prevent relapses.482,506,508 They should be prescribed early, within the first hour of treatment in the emergency room, since their effect starts 4-6hours after administration. They are especially indicated if no improvement is seen after the first dose of SABA, if the patient was already receiving them or if previous exacerbation episodes requiring these systemic glucocorticoids had occurred.

The preferred administration route of glucocorticoids is the oral route, as it is as effective as the intravenous administration,509 less invasive and cheaper.484,485 The intravenous route is limited to patients with severe dyspnea preventing swallowing and patients with vomiting or under mechanical ventilation.

The daily dose is 50mg of prednisone, as a single morning dose481 for 5-7 days, with no down-titration being necessary.510,511

Early use of high doses of ICS within the first hour of treatment reduces the need for hospital admission as in the case with systemic administration of glucocorticoids.508

The use of ICS together with systemic glucocorticoids provides even a higher reduction in the number of hospital admissions.508

Theophylline drugs should not be used in exacerbation episodes because of their lower efficacy and safety as compared with salbutamol.512

Routine administration of magnesium sulfate is not indicated, although in selected patients experiencing severe obstruction (FEV1 25-30% of predicted) or persistent hypoxemia, a single dose of 2g administered by infusion reduces the need for hospitalization.513–515

A systematic review of patients with severe exacerbations treated with intravenous magnesium sulfate showed a mild improvement of pulmonary function only.516

However, a more recent systematic review showed beneficial effects of inhaled magnesium sulfate added to SABA or SABA plus ipratropium bromide, reducing hospital admissions, in addition to a mild improvement of pulmonary function.517

Heliox, a mixture of helium and oxygen, in 80/20 70/30 proportion, has no place in the routine management of exacerbations due to the lack of consistent data regarding the efficacy of this compound. However, it may be considered in patients who do not respond to the usual treatment,518,519 particularly to nebulizing SABA.520

Regarding leukotriene antagonists, no data supporting their use either orally or intravenously are available. There is no evidence supporting the use of antibiotics, except in the presence of a clearly symptomatic respiratory infection.

4.3.3Treatment failure

The use of non-invasive mechanical ventilation may be an option in severe exacerbations resistant to treatment. It allows improvement of the respiratory rate, dyspnea, and, in particular, airflow obstruction due to a direct effect of positive pressure, or indirectly contributing to a better distribution of aerosols.521

Close monitoring is necessary so as not to delay the use of invasive mechanical ventilation in patients with an imminent life-threatening compromise.

4.4Hospitalization criteria

The rate of hospital admission in asthma patients attended in the emergency setting is around 20%,522 although there is a large variability among different countries.523,524 It is well known that adherence to guidelines is associated with a lower risk of hospitalization.523 A systematic review522 identified the degree of pulmonary function impairment as the most important risk factor for in-patient care.

The decision to hospitalize a patient should be made within the first three hours after the start of treatment of the exacerbation episode, given that decision-making is rarely modified by longer periods of monitorization.525

However, assessment of the patient's clinical condition and pulmonary function within the first hour after admission to the emergency room already allows to predict the need for in-patient care.526,527

Criteria for admission to the hospital or to the ICU are summarized in table 33.

Table 33.

Criteria for hospital admission and ICU admission.

Criteria for hospital admission  Criteria for ICU admission 
Remain symptomatic after treatment  Respiratory arrest 
O2 requirement to maitain SaO2 >92%  Decrease in the level of consciousness 
– PEF or FEV1 <50-60% after treatment.529– PEF or FEV1=50-70% on arrival. A minimum observation period of 12 hours is advisable.– There is no functional parameter that defines when a patient should be discharged, although PEF <75% and variability higher than 25% are associated with a higher rate of re-admissions530  Progressive functional deterioration despite treatment 
Previous life-threatening exacerbation with history of intubation and ventilation, hospital admission or visit to the emergency department due to recent asthma  SaO2 <90% despite supplementary O2PaCO2 >45 mm Hg=alarming sign of muscle exhaustion 
Failure of treatment with oral glucocorticoids in the outpatient setting  Hypercapnia, need of ventilatory support or pneumothorax 
Impossibility to ensure necessary care measures at home   
Respiratory (pneumonia, pneumothorax, pneumomediastinum) or non-respiratory comorbidities   

ICU: intensive care unit; SaO2, arterial oxygen saturation; PEF, peak expiratory flow; FEV1, forced expiratory volume in one second; PaCO2, arterial partial pressure of carbon dioxide.

Modified from Piñera-Salmerón et al., 2020528
4.5Hospital discharge criteria

There are no functional parameters that allow a patient to be discharged with complete safety, so that the decision is usually the result of the doctor's clinical observation of the patient's condition and data of arterial oxygen saturation.531

Patients may be discharged from hospital if they are capable of following their prescribed treatment at home, are paucisymptomatic or there is a reduced need for reliever medication.530

However, it is highly recommended to have an objective pulmonary function test, such as spirometry or PEF. FEV1 or PEF values >70% and with minimal symptoms can be criteria for discharge.532 If FEV1 or PEF values are between 50% and 70%, possible risk factors should be considered (table 33).

Before discharge from the hospital, it is necessary to deliver a minimum education plan including checking of the inhalation technique and the provision of a written action plan (see chapter 3.4.3.). Also, an appointment with the patient's attending physician will be scheduled within the next five days.488

Figure 9 shows an algorithm for the patients’ hospital admission or discharge.

Figure 9.

Algorithm for the site of care based on severity of the exacerbation episode.

FEV1: forced expiratory volume in one second; PEF: peak expiratory flow.

(0.18MB).
4.6Referral and control after discharge

The care of patients who have suffered an asthma attack does not finish at the time of hospital discharge, and all patients should be assessed after the acute episode.

All patients should be evaluated by his/her family physician within five days after discharge,488 as well as those who had suffered from a severe exacerbation by the pneumologist or allergologist within one month.531Table 34 shows criteria for referral to the next healthcare level.

Table 34.

Criteria for specialized evaluation of patients within a month after an asthma exacerbation episode.531

– Severe or life-threatening exacerbation.– Repeated exacerbations requiring care in the emergency department.473,476– Exacerbations that require in-patient care,476,533 uncontrolled severe asthma, particularly in corticosteroid-dependent asthma, allergic bronchopulmonary aspergillosis, vasculitis.– Pregnancy.534– Exacerbations triggered by NSAID, aeroallergens, food allergens or presenting with anaphylaxia.– Known associated comorbidities.– Clinical suspicion of vocal cord dysfunction, nasal polyposis, rhinosinusitis, gastroesophageal reflux, sleep apnea-hypopnea syndrome, asthma-COPD overlap syndrome 

NSAID: non-steroidal anti-inflammatory drug; COPD: chronic obstructive pulmonary disease.

4.3Recommendations

4.1. The initial assessment of the patient with an exacerbation episode should include the analysis of the life-threatening risk, level of severity, and degree of airflow obstruction.  R2 
4.2. Depending on the signs and degree of airflow obstruction, the patient with an asthma exacerbation episode should be classified into four levels of severity: mild, moderate, severe, and life-threatening.  R2 
4.3. The degree of airflow obstruction will be objectively established by means of spirometry (FEV1) or peak expiratory flow (PEF)measurement.  R2 
4.4. In patients with asthma exacerbation, it is recommended to consider the initial therapeutic response of airflow obstruction and signs of severity, in order to establish the approach that should be followed.  R2 
4.5. Treatment with SABA is recommended in mild exacerbation episodes.  R1 
4.6. In moderate or severe exacerbations, early administration of systemic glucocorticoids and oxygen at the lowest concentration ensuring SaO2 >90% is recommended.  R1 
4.7. The decision of hospital admission should be made within the first three hours after starting treatment of the exacerbation episode, because the level of bronchodilation achieved cannot be increased significantly beyond this period.  R2 
4.8. Patients with FEV1 or PEF >70% (predicted or best personal value) and with minimal symptoms can be discharged from the hospital.  R2 
4.9. Before hospital discharge, a minimum education plan including assessment of the patient's inhalation technique should be delivered, and a written action plan should be provided.  R2 
4.10. After an exacerbation, it is recommended that the patient should be evaluated by his/her family physician within five days and, if necessary, by a specialist within a month.  R2 

5Treatment of childhood asthma5.1Education

The education of the child with asthma and his/her family increases the quality of life, reduces the risk of exacerbations and the cost of healthcare, the reasons for which education is one the fundamental pillars of treatment. Its objective is for the child to achieve a normal life for his/her age including physical exercise and sport activities.535

Education is essential to improve treatment adherence and to achieve control of the disease.536,537

Education should be developed in all healthcare settings in which children with asthma are attended.538

Education will be primary addressed to the family during early childhood and, from 8-9 years, should be especially addressed to the child, in order to promote personal autonomy and to achieve the maximum degree of self-care.539

Home education programs may be beneficial for children with poorly controlled asthma and are potentially profitable.540

For education to be effective, it is essential to identify the educational needs and the factors that affect the behavior of the patient and/or his/her family.541

Key aspects of education are shown in table 35.535

Table 35.

Key aspects of the education of a child with asthma.

Topic area  Key points 
Asthma  – Concept of asthma (chronic disease, variability)– Symptoms exacerbation/between exacerbations– Bronchoconstriction– Inflammation 
Environmental measures  – Counselling against smoking– Triggering factors (allergens, viruses, exercise, etc.)– How to identify and avoidance measures 
Treatment  – Bronchodilators (rescue treatment)– Anti-inflammatory drugs (maintenance treatment)– Side-effects– Exacerbations (how to recognize initial symptoms and early action)– Inmunotherapy 
Inhalers  – Importance of inhaled medication– Inhalation technique– Maintenance of the system– Errors/forgetfulness 
Self-control  – PEF. Best personal value– Symptoms registry– Personalized written action plan 
Lifestyle  – School attendance– Practice of sports– Autonomy 

PEF: peak expiratory flow.

The education of children with asthma is more effective when accompanied by personalized written action plans (table 36),542,543 which address maintenance treatment (table 37)544 and management of asthma exacerbations (table 38).545 Every education plan should be reviewed periodically.

Table 36.

Components of a personalized action plan.

Action plan for treating asthma exacerbation at home 
• Recognize asthma symptoms and the onset of an exacerbation for using early short-acting bronchodilators and on-demand when symptoms appear.• Recognize warning signs and when to seek help from the doctor or go to the emergency department. 
Plan of self-control/control by the family 
• Rules for avoiding specific asthma triggers in children.• Daily use of preventive medication: doses, frequency and route of administration.• Changes of preventive medication according to the severity and frequency of symptoms (symptom diary) and/or measurement of peak expiratory flow (home recording of PEF).• When to go to his/her pediatrician because asthma is not controlled.• Prevention and treatment of exertional asthma. 

PEF: peak expiratory flow.

Table 37.

Written action plan to maintain asthma control.

Your usual treatment (preventive):Every day I take: ____________________________________________________Before exercise I take: ________________________________________________
WHEN TO INCREASE PREVENTIVE TREATMENT     
Assess your level of asthma control:     
In the last week you have had:     
Asthma symptoms more than twice a day?  No  Yes 
Activity or physical exercise limited by your asthma?  No  Yes 
Night awakenings due to asthma?  No  Yes 
Need of rescue medication more than twice a day?  No  Yes 
If you measure (PEF), your PEF is lower than _______________________________  No  Yes 
If you have answered “Yes” to 3 or more questions, your asthma is not well controlled and to increase a step in your treatment may be necessary     
HOW TO INCREASE TREATMENT     
Increased treatment from _____________________________________to ______________________________________________________________and assess improvement every day. Maintain this treatment for ___________days.
In case of an exacerbation, treatment will be started based on the action plan for the management of exacerbations and will attend a medical consultation for a new assessment.
Modified from GINA (www.ginasthma.com).
Table 38.

Action plan for treating an asthma exacerbation at home.

What is an ASTHMA EXACERBATION EPISODE and HOW TO ACT AT HOME? 
An asthma exacerbation episode is a sudden or progressive worsening of symptoms:– Increased cough (continuous, nocturnal or with exercise).– Whistling sound.– Fatigue (difficult breathing).– Feeling of chest tightness.– Decrease of PEF (if you use the peak-flow meter). 
There are symptoms that warn us that an exacerbation can be severe (warning signs):– Bluish color of the lips.– Ribs sink when breathing.– Difficulty speaking.– Numbness.Warning signs indicate that medical assistance should be immediately requested! 
What to do at home in the presence of an exacerbation episode?– Keep calm.– Treat symptoms as early as possible.– Start medication at home.– Never wait to see if symptoms disappear spontaneously.– After starting medication, observe for 1 hour and assess response. 
USE OF MEDICATION:Take your rapid rescue medication: salbutamol _____________ with spacer, 2-4 puffs, separated by 30-60 seconds. This dose can be repeated very 20 minutes, up to a maximum of 3 times.If symptoms do not improve in 1 hour, start taking oral glucocorticoids ___________________ (1 mg/kg/day, maximum 40 mg/day), during 3-5 days and go to the healthcare center or emergency department. 
Take your anti-inflammatory medication___________________ times a day, all days, according to the indications given by your pediatrician. 
ASSESS RESPONSE TO TREATMENTIf you improve in one hour and improvement is maintained for 4 hours, continue with salbutamol: 2-4 puffs every 4-6 hours (depending on symptoms) and visit your pediatrician in 24-48 hours.If you do not improve or the improvement is not maintained and you relapse again: go to an emergency departmenti 
Si If you know how to control exacerbations, the duration of symptoms will be shorter and your quality of life will improve. 

In children, written action plans based on measurement of PEF do not provide benefits as compared with plans based on monitoring of symptoms, so that PEF-based plans are not generally recommended.542,546 However, on an individual basis, children and adolescents with severe asthma and low perception of symptoms could benefit from plans based on PEF monitoring.547,548

5.2Maintenance treatment5.2.1Drugs

Inhaled corticosteroids (ICS). ICS are the first-line of treatment. In children older than 3 years of age, the efficacy of daily ICS has been conclusively established, with improvement of clinical, parameters and bronchial inflammation parameters, better quality of life, and decrease in the risk of both exacerbations and hospitalizations.549,550

Infants and preschool children treated with daily ICS experience fewer recurrent asthma/wheezing episodes,551,542 with a better treatment response being obtained by those showing risk factors of developing persistent asthma (Asthma Predictive Index [API]),552,553 while viral-induced episodic wheezing shows limited response.554 A treatment trial followed by evaluation of response is recommended.555

Treatment with ICS, either continuously or intermittently, does not modify the natural history of the disease.556,557

In preschool and children, the use of controller drugs (ICS or montelukast) at regular doses or intermittently at the onset of symptoms is not recommended.558–560

Early intermittent therapy with ICS at high doses given to infants and preschool children with moderate-severe episodic wheezing and risk factors (API+) at the onset of symptoms have shown to be effective in reducing the severity and duration of exacerbations,550,561,562 but further safety studies are needed to establish a generalized recommendation of this therapy. It could be an option in highly selected cases in which education and acceptance by families are guaranteed.563,564

When administered at usual doses, ICS are safe drugs for the management of childhood asthma. There is usually a decrease in the growth rate at the beginning of treatment (1-3 years), although this is a transient effect and does not influence final growth or final height. However, the final height of children treated with ICS over prolonged periods is lower, an effect proved to be dose-dependent.565,566

It is difficult to establish the equivalent doses of ICSs mostly used in pediatric age.567 Comparable doses of ICS drugs for use in the pediatric age group are tentatively shown in table 39., taking into account that the lowest dose that maintains patient's control must be sought.

Table 39.

Comparable doses of inhaled glucocorticoids commonly used in pediatric age (μg/day).

Children under 12 years of age
  Low doses  Medium doses  Hogh doses 
Budesonide  100-200  >200-400  >400 
Fluticasone propionate  50-100  >100-250  >250 

Leukotriene receptor antagonists (LTRA). In preschool children with virus-induced asthma/weezing episodes, LTRA are associated with a modest reduction of symptoms and the need of oral glucocorticoids as compared with placebo.559,568,569 Although a definite beneficial effect remains unclear, a clinical trial to assess response to LTRA may be conducted, which could be stopped if the expected response is not obtained.568 More evidence is needed to determine whether there is a responder phenotype to montelukast.570

If asthma symptom cannot be controlled with ICS at low doses, increasing ICS at medium doses is more effective than the association with montelukast.571

Association of long-acting ß2-adrenergic agonists and ICS. It has been approved for use in children over 4 years of age. The use of a LABA when administered with an ICSs is safe, but should never be used as monotherapy.572,573

One study showed a decrease in exacerbations and the need for systemic glucocorticoids in children aged 4-11 years with formoterol/budesonide administered in a single inhaler, both as maintenance treatment and as reliever (MART strategy),574 although some authors consider that the evidence in this age group is limited.575

En In children aged between 6 and 11 years with uncontrolled persistent asthma with low doses of ICS, doubling the ICS dose has a similar effect on clinical control and lung function than adding a LABA.576 However, the clinical phenotype and the heterogeneity of the individual response to ICS, LTRA and LABA should be assessed,539,577 therefore, it is necessary to closely monitor the response to treatment in children with uncontrolled asthma using ICS.

Tiotropium. It is a long-acting muscarinic antagonist. It can be used in children from 6 years of age with poorly controlled severe asthma treated with ICS at high doses plus a LABA. The dose is 5μg once a day.578 A study in children aged 1 to 5 years concluded that tolerability of tiotropium is good in preschool children and that this agent can reduce the number of exacerbations.579

Biologics. Biologics are drugs indicated in severe uncontrolled asthma that are aimed at treating the underlying inflammation by blocking different mediators. In the pediatric field, there are currently three biological agents available, all of them being monoclonal antibodies that are administered subcutaneously. Omalizumab targets IgE, mepolizumab blocks IL-5, and dupilumab targets IL-4 and IL-13. They have shown efficacy and safety and are recommended for children aged 6 and above.580,581 Their characteristics, indications, doses, and modes of administration are indicated in section 7.5 (Severe uncontrolled asthma in children).

Immunotherapy (IT). When biologically standardized extracts are used and appropriately selected in sensitized patients, immunotherapy has been shown to provide a beneficial effect by reducing symptoms, the need of reliever and maintenance medication, and decreasing bronchial hyperresponsiveness (both specific and non-specific).582

Also, IT prevents the development of new sensitizations and asthma in children with rhinitis.583,584

5.2.2Treatment according to the level of severity, control and future risk

Considering that the main objective is to achieve control with the least possible medication, the treatment should be continuously adjusted, escalating or de-escalating the therapeutic step based on the level of control, always considering non-pharmacological measures, therapeutic adherence, and modifiable risk factors (Figure 10).

Figure 10.

Stepwise treatment of asthma in the pediatric age according to the level of

ICS: inhaled glucocorticoids; LTRA: leukotriene receptor antagonists; LABA: long-acting β2-adrenergic agonist; GC: glucocorticoid. *From 6 years of age. **Off-label.

(0.37MB).

The maintenance treatment will be initiated based on the initial severity level (recurrence or intensity of symptoms). Subsequently and retrospectively, the severity will be classified according to the level of treatment necessary to maintain symptom control.

  • Step 1. Children who have occasional asthma symptoms, without nighttime symptoms, and without risk factors for exacerbation, should only use bronchodilators on-demand. In the case of infrequent asthma symptoms but with risk factor(s) for exacerbation (table 19), the corresponding treatment for step 2 should be initiated. It is important to perform a careful evaluation, ensuring that the symptoms are truly intermittent and not persistent. From the age of 12, the use of formoterol associated with ICS could be considered.

  • Step 2. Children who require treatment with SABA two or more times per month, without symptoms between episodes and with normal lung function, should initiate treatment with low doses of ICS or consider montelukast as an alternative.

  • Step 3-4. Children with more than 6-8 asthma episodes per year, symptoms between episodes, asthma-related awakenings once a week, and/or impaired pulmonary function should adjust their treatment to step 3 or 4. In this step, there are three options: ICS at medium doses, adding a LABA to ICS at low dose (step 3), or association of a LABA with ICS at medium doses (step 4), and in children under 4 years of age, montelukast associated with low doses of ICS.

  • Step 5-6. In children with persistent symptoms, the need for short courses of oral corticosteroids, wheezing with minimal exertion, and impaired pulmonary function, treatment should be initiated at step 5 (ICS at high doses/LABA). Once control is achieved, the treatment should be stepped down to the lowest effective dose.585

If control is not achieved in children less than 4 years of age, it is necessary to consider adding one or more of the following drugs: LABA (off-label use), tiotropium (off-label use in children under 6 years), macrolides, or even oral glucocorticoids. In children of more than 6 years of age, options include tiotropium, monoclonal antibodies, or oral glucocorticoids (see chapter 3, “Severe Asthma”).

In children older than 12 years, it is possible to apply the so-called MART therapy, that is, ICS/formoterol, both as maintenance and rescue treatment.

5.3Evaluation and treatment of exacerbations5.3.1Evaluation of severity

The following factors should be considered: time course of the exacerbation episode, pharmacological treatment administered, presence of associated diseases, and possible risk factors (previous intubation or ICU admission, hospitalizations in the previous year, frequent need of admission to the emergency department in the previous year and/or use of oral glucocorticoids, and excessive use of SABA in the preceding weeks).

Severity assessment is mainly based on clinical criteria (respiratory rate, presence of wheezing and sternocleidomastoid retractions). Although no clinical scale is considered to be well validated586,587 the Pulmonary Score (table 40)588 has been found to be useful and applicable to all ages. The combination of symptoms and arterial oxygen saturation (SaO2) allows completing an estimation of the severity of the exacerbation episode (table 41).

Table 40.

Pulmonary Score for clinical assessment of asthma exacerbation in children.*

Score  Respiratory rateWheezing  Use of sternocleidomastoid muscle 
  <6 years  ≥ 6 years     
<30  <20  No  No 
31-45  21-35  End of expiration  Slight increase 
46-60  36-50  Throughout expiration (stethoscope)  Increased 
>60  >50  Inspiration and expiration without stethoscope**  Maximum activity 
*

It is scored from 0 to 3 in each of the sections (minimum 0, maximum 9).

**

If wheezing is absent and the sternocleidomastoid activity is increased the wheezing section stolid be scored 3.

Table 41.

Overall evaluation of the severity of asthma exacerbation in children by integrating the Pulmonary Score and the arterial oxygen saturation.

  Pulmonary Score  SaO2 
Mild  0-3  >94% 
Moderate  4-6  91-94% 
Severe  7-9  <91% 

SaO2: arterial oxygen saturation. In case of disagreement between clinical score and arterial oxygen saturation, the score indicating higher degree of severity will be used.

5.3.2Drugs

Inhaled short-acting ß2-adrenergic agonists (SABA). These agents constitute the first-line treatment due to their higher effectiveness and lower incidence of side effects.589 They should preferably be administered via a pressurized inhaler with a spacer chamber, since this way of administration is as effective as nebulizers for treating an acute asthma episode.590–593

The recommended doses and dosing intervals depend on the severity of the exacerbation episode and the response to the initial doses.594 The most commonly used drug is salbutamol, which is available as a solution for use with a nebulizer and a pressurized inhaler. The latter must be administered in sequences of 2-10 puffs of 100μg until response is obtained. In mild exacerbations, a series of 2-4 puffs may be sufficient, although up to 10 puffs may be necessary for severe exacerbations.

Nebulized SABA should be restricted to those cases in which the patient requires oxygen supply for SaO2 normalization, although a recent randomized clinical trial (RCT) showed that even in severe exacerbations, the administration of salbutamol and ipratropium bromide with spacer chamber and facial mask with oxygen by means of a nasal cannula was more effective than using a nebulizer.595

Continuous nebulization does not offer greater advantages compared to intermittent nebulization when the same total doses are administered.596,597

Ipratropium bromide. The use of frequent doses, every 20minutes, of ipratropium bromide for the first 2hours in case of severe asthma exacerbations or moderate exacerbations not responding to initial treatment with SABA, has been shown to be effective and safe.588,598 The nebulized dose is 250μg for children weighing less than 30kg y 500μg for those weighing more than 30kg. The dose for inhaled use with a spacer chamber is 40-80μg (2-4 puffs). The maximum effect, which tends to decrease gradually, is observed with the first doses, so that inhalations beyond the first 24-48hours should not be maintained.599

In infants, the use of ipratropium combined with inhaled SABA has been shown to be effective in the treatment of the most severe exacerbations.600 The effect of this association administered by an inhaler seems to be higher as compared to that administered by nebulization.595

Systemic glucocorticoids. The efficacy of systemic glucocorticoids in preschool children with mild to moderate acute episodes of wheezing induced by viral infections has been questioned; hence, its use should be restricted to more severe exacerbations (1-2mg/kg/day).569,601,602 In children over 5 years of age, these agents have shown benefit after early use,603 with the oral route being preferred over the intravenous or intramuscular routes, except for circumstances in which oral intake is not feasible.604,605 Systemic glucocorticoids should be administered in moderate-severe exacerbations, and may be considered for mild exacerbations when sufficient improvement with bronchodilators has not been achieved or the child has a history of severe attacks (in this case, early administration). Prednisolone at doses of 1-2mg/kg/day (maximum 40mg) for 3 to 5 days or until resolution of the asthma attack is the most commonly used drug.606,607

Dexamethasone is being used as an alternative. The effect of administering a single dose of dexamethasone orally (at 0.3-0.6mg/kg) is not inferior to that of administering prednisolone orally (at 1mg/kg/day) during 3 days of treatment.608–611

Inhaled glucocorticoids. There is insufficient evidence to recommend the use of ICS as an alternative612 or additional treatment to systemic glucocorticoids613,614 in the management of asthma exacerbations. Larger studies are needed, with better methodological quality and cost-effectiveness analysis,615 as well as safety studies.612

Magnesium sulfate. It can be used in severe exacerbations with unsuccessful response to the initial treatment,616,617 but its use does not prevent hospitalizations.618 The drug is administered intravenously as a single dose of 40mg/kg (up to 2g) over 20minutes.

Nebulized magnesium sulfate together with a ß2-adrenergic agonist in the treatment of an asthma exacerbation seems to have benefits in the improvement of pulmonary function.619,620

5.3.3Therapeutic regimens

Treatment of an asthma exacerbation episode depends on its severity and follows the flow chart shown in Figure 11. Doses of drugs and duration of administration should be modified according to the severity of the exacerbation and the response to treatment.

Figure 11.

Treatment of asthma exacerbation in children.

SaO2: oxyhemoglobin saturation; max: maximum. SABA: short-acting β2-adrenergic gonist.

(0.4MB).

When SaO2 is below 94%, oxygen therapy is required to maintain SaO2 between 94-98%.621,622 An SaO2 <92% after initial treatment with inhaled bronchodilators can be used as a marker to select the most severely ill patients who should be hospitalized for starting intensive treatment.621,622

During the first 2 hour of treatment and in children with moderate/severe exacerbation unresponsive to first-line therapy, the use of a high-flow nasal cannula seems to be superior to conventional oxygen therapy to reduce breathing difficulty.623,624 However, more studies are needed to demonstrate the general efficacy of this approach in the management of asthma and respiratory insufficiency in the emergency setting.625

Regarding non-invasive ventilation (NIV), the current available evidence does not allow to confirm or exclude its use in exacerbation episodes refractory to the usual treatment.626

Mild and moderate exacerbations can be treated in the primary healthcare setting.

In the presence of severe exacerbation or suspicion of complications, history of high-risk exacerbations or lack of response to treatment, patients should be referred to the hospital in a medicalized ambulance.

Follow-up. It is necessary to evaluate the degree of the control of symptoms in the previous weeks, the presence of risk factors, possible triggering factors, and previous treatment. Also, it is important to assess the level of therapeutic adherence and to supervise that the inhalation technique is correct. A written action plan must be reviewed or provided and a follow-up visit arranged.544

4.4Recommendations

5.1. The education of the child with asthma and his/her family is recommended because increases the quality of life and reduces the risk of exacerbations and healthcare costs.  R1 
5.2. In the education of a child with asthma, it is recommended to include written personalized management action plans, addressing maintenance treatment and how to treat exacerbations.  R1 
5.3. Inhaled corticosteroids (ICS) is recommended as first-line treatment for the control of persistent asthma in children of any age.  R1 
5.4.Montelukast can be tried as an alternative to ICS for maintenance therapy.  R2 
5.5. Treatment with LABA can be considered in children older than 4 years of age but always combined with ICS. LABA monotherapy should never be administered.  R1 
5.6. In the treatment of children with allergic asthma, immunotherapy should be considered provided that biologically strandardized extracts are used and patients are appropriately selected.  R1 
5.7. In children aged 6 years or older with insufficiently controlled severe persistent asthma with high doses of ICS and LABA and/or LTRA and/or tiotropium, the use of biologics or monoclonal antibodies is recommended.  R1 
5.8. Before considering that an asthma patient is poorly controlled and stepping up treatment, the diagnosis of asthma should be confirmed, treatment adherence and inhalation technique should be evaluated, and other comorbidities excluded.  R1 
5.9. Early and repeated administration of SABA at high doses is the first-line of treatment of asthma exacerbations in children.  R1 
5.10. It is recommended to indidualize drug doses according to severity of the exacerbation and the response to treatment.  R2 
5.11. Early use of systemic glucocorticoids is recommended in moderate and severe exacerbations; in mild exacerbation, an individualized assessment of its use is recommended.  R1 
5.12. In the presence of SaO2 <92% after an initial treatment with inhaled bronchodilators, admission to the hospital to start intensive therapy is recommended.  R2 
5.13. A pMDI with spacer should preferably be used to administer bronchodilators, especially in mild-to-moderate attacks.  R1 
5.14. The degree of control, risk factors, therapeutic adherence, and inhalation technique should be assessed, a written action plan should be provided, and the follow-up of children with exacerbations should be ensured.  R2 

6Asthma-associated rhinitis and rhinosinusitis6.1Definition and epidemiology

The term rhinitis defines the inflammatory process of the nasal mucosa, which is characterized by the following clinical symptoms: anterior or posterior rhinorrhea, sneezing, block of nasal passages or congestion and/or nasal pruritus/itching. These symptoms should be present for two or more consecutive days and for more than one hour on most of the days.627–629

Rhinitis is a syndrome that encompasses several phenotypes. Rhinitis has the highest prevalence of all diseases, and it has been estimated that 100% of the population (children and adults) suffer from 1 to 10 episodes of infectious rhinitis annually630 (table 42). Allergic rhinitis (AR) is the most prevalent of all chronic diseases, affecting 22-41% of the European population631 and 12.6% of children aged 0-18 years.632 The prevalence of non-allergic rhinitis (NAR) is not so well estimated, with the highest rates in children under 6 years (up to 24.9%) and around 10% in children older than 15 years of age.633

Table 42.

Phenotypes of rhinitis.

InfectiousNon-infectious
Viral  Bacterial  Allergic/local allergic  Non-allergic 
    • Intermittent/persistent• Seasonal/perennial• Occupational• Mild/moderate/severe  • Occupational rhinitis• Drug-induced rhinitis• Gustatory rhinitis• Hormonal rhinitis• Reactive rhinopathy (nasal hyperreactivity/old vasomotor rhinitis)• Dry/atrophic/sicca rhinitis• Idiopathic rhinitis 

In Spain, rhinitis is the most common reason for consultation in Allergology (62% in adults and 53.8% in children).634,635 The ISAAC study reported a prevalence of rhinoconjunctivitis of 7.9% in Spanish children aged 6-7 years (with an annual increase of 0.33) and 15% among those aged 13-14 years (annual increase of 0.10).636

AR-associated costs are high. A study carried out in Spain (FERIN project) reported that the cost per patient per year was €2,326.70 (direct costs €553.80; indirect costs €1,772.90).637

6.2Diagnosis and classification

The diagnosis of AR is mainly based on clinical manifestations, although symptoms do not enable to assess the cause, pathophysiology or the specific rhinitis phenotype; therefore, complementary diagnostic tests are necessary to establish the etiological diagnosis in cases of rhinitis of moderate to severe intensity.627

An initial approach to the classification (phenotyping) of rhinitis should establish whether the patient presents an infectious or non-infectious rhinitis, and subsequently classify rhinitis based on positivity of allergy tests and the correlation with the patient's symptoms. Two main rhinitis phenotypes are defined: AR and NAR. NAR includes a heterogeneous group of phenotypes of different pathogenetic mechanisms638 (table 42).

Family history of allergy, seasonal manifestation of symptoms, concomitant ocular and nasal symptoms and its relationship with exposure to aeroallergens are clinical data with a high predictive value for suspicion of AR639 (fig. 12).

Figure 12.

Diagnostic algorithm of allergic rhinitis.

ENT: ear, nose and throat; CT: computerized tomography.

(0.32MB).

The most efficient complementary tests for the diagnosis of rhinitis are allergic tests: skin prick testing or intradermal puncture with standardized allergic extracts and determination of specific serum IgE against allergens, preferably against recombinant allergens.638 A high percentage of patients with positive allergic tests do not have the disease or positive allergens are not clinically relevant, so that clinical correlation is indispensable to establish the diagnosis.640

The specific nasal challenge (or provocation) test with allergens is the reference test for the diagnosis AR and can be necessary in the case of a high clinical suspicion and negative results of intradermal testing or specific serum IgE.641,642

A specific AR phenotype, named local AR, has been described, which is characterized by negativity of systemic allergic tests (intradermal tests or specific serum IgE) and positive specific nasal challenge test.643

Other complementary tests that can be useful in the study of nasal function include an objective assessment of obstruction (acoustic rhinometry, active anterior rhinomanometry, measurement of peak nasal inspiratory flow),644 assessment of nasal inflammation (nasal nitric oxide [nNO], nasal cytology, biopsy),645 and assessment of olfactory function by olfactometry.646

AR is an IgE-mediated chronic inflammatory immunological disorder of the nasal mucosa that causes a myriad of symptoms, including nasal obstruction/congestion, nasal and ocular itching, sneezing bouts, and rhinorrhea after inhalation of environmental allergens.647

AR can be classified according to different criteria. On the basis of triggering allergens, AR can be classified into seasonal (outdoors such as pollens and fungal spores mainly) or perennial (indoors such as dust mites, insects, animal dangers or other fungal spores), and on the basis of temporal criterium as intermittent or persistent (symptoms present for more than 4 days a week and for more than 4 consecutive weeks). This last classification has been validated and has been shown to better reflect the actual clinical condition of patients.648

The severity of AR is evaluated on the basis of the impact on the quality of life (sleep disturbance, impairment of daily life activities, leisure and/or sport activities, impairment of school or job tasks, and the consideration of symptoms as bothersome), differentiating into mild (none affected) moderate (one to three) or severe (all affected). This classification has been validated in children and adults, with and without treatment649,650 (table 43). A visual analogue scale can also be used to assess severity of AR.652

Table 43.

Classification of allergic rhinits.

1. According to duration   
Intermittent  Persistent 
Symptoms are present for ≤ 4 days a week or for ≤ 4 consecutive weeks  Symptoms are present for >4 days a week for >4 consecutive weeks. 
2. According to severity     
Mild  Moderate  Severe 
None of the following items is present:– Sleep disturbance– Impairment of daily, leisure and/or sports activities– Impairment of school and job tasks– Symptoms are bothersome  • - One,• - Two,• - or three of the aforementioned items are present  The four items are present 
Modified from Bousquet 2008, according to Valero 2007651.

In the last few years, and in a similar way to that established in asthma, it has been proposed to evaluate the control of rhinitis using validated questionnaires (such as the Rhinitis Control Assessment Test)653 or using a visual analogue scale (available as applications for mobile devices).654

6.3Rhinitis and asthma

Multiple epidemiological, physiopathological, and therapeutic studies have shown an association between rhinitis and asthma.627

Factors determining why some patients with AR will develop asthma are unclear (table 44), although it is known that both AR and NAR are risk factors for asthma.655,656

Table 44.

Interrelationship between rhinitis and asthma: risk factors for asthma.

– Allergic rhinitis.– Non-allergic rhinitis.– Characteristics of aeroallergens.– Number of sensitizations.– Intensity of sensitization.– Severity and duration of rhinitis.– Number of associated allergic diseases (rhinitis, conjunctivitis, dermatitis). 

Sensitization to different types of aeroallergens and specific profiles are associated with different allergic clinical features (rhinitis with/without conjunctivitis with/without asthma) and different levels of severity.649,657

According to some studies, the association with asthma would be greater in cases of more severe and prolonged AR,658,659 higher number of sensitizations,658,660,661 higher specific IgE levels662 and in the presence of various associated allergic diseases (rhinitis, conjunctivitis, dermatitis).663,664

The prevalence of rhinitis in patients with asthma is very high and much higher than in the general population.665 In Spain, two studies showed a prevalence of rhinitis in patients diagnosed with asthma of 71% and 89.5%, respectively.666 Also, it has been shown a parallel increase in the prevalence of asthma and rhinitis.667

Suffering from rhinitis aggravates asthma,668 worsens asthma control669 and asthma symptoms,670 and increases the use of healthcare resources.671,672

Inflammatory changes in the bronchial mucosa of non-asthmatic patients with AR have been observed,673 as well as nasal eosinophilic inflammation in asthma patients without nasal symptoms.674

Treatment of AR with intranasal glucocorticoids may improve some aspects of asthma, such as pulmonary function,675 symptom score, quality of life or the use of reliever or rescue medication,676 the level of asthma control,677 and exacerbations in the pediatric population.678,679

6.4Treatment of allergic rhinitis

The treatment strategy of allergic rhinitis includes patient education, avoidance of allergens and contaminants, pharmacotherapy and allergen-specific immunotherapy. At the time of selecting the pharmacological treatment, efficacy, safety, cost-effectiveness relationship, patients’ preferences, severity of disease and the presence of comorbidities should be evaluated. Pharmacological treatment of allergic rhinitis should include clear-cut recommendations that will have to be implemented in a stepwise approach according to severity (fig. 13).

Figure 13.

Algorithm of treatment of allergic rhinitis.627,675,676

LTRA: leukotriene receptor antagonists; GC: glucocorticoids. *In short time periods, usually less than 5 days.

(0.61MB).

Second generation H1-antihistamines (non-sedating) (bilastine, cetirizine, desloratadine, ebastine, fexofenadine, levocetirizine, loratadine, mizolastine and rupatadine) administered by the oral route, improve symptoms both in adults and children, such as rhinorrhea, szeening, nasal itching and ocular symptoms, although are less effective to relieve nasal obstruction, and should be preferred over sedating antihistamines for their favorable risk-benefit ratio.638

Topical H1-antihistamines (azelastine, emedastine, epinastine, levocabastine and olopatadine) have a rapid effect on symptoms, are more effective for nasal congestion than oral antihistamines and more effective for ocular symptoms, although are less effective for nasal congestion than intranasal glucocorticoids (INGC), and have been shown to reduce symptoms and improve quality of life versus placebo, without relevant side effects except for a bitter taste.638

INGC (budesonide, ciclesonide, fluticasone, mometasone and triamcinolone) are very effective drugs for reducing nasal and ocular symptoms, even when administered intermittently, and are superior to oral antihistamines and montelukast. Their use may be associated with some minor adverse effects, such as epistaxis or headache, but a relevant effect neither on the hypothalamic-pituitary axis nor on the growth of children has been demonstrated.638

The combination of a glucocorticoid and an intranasal antihistamine (fluticanose propionate and azelastine or mometasone furoate and olopatadine) in a single device has shown a rapid and more effective effect than the use of INGC or intranasal antihistamines in monotherapy, with the only relevant adverse effect of its bitter taste. It is recommended in more severe or uncontrolled cases or as a second-line treatment after failure of monotherapy.638,680–682

Montelukast has consistently shown to reduce symptoms and to improve quality of life as compared with placebo, although to a lower extent than INGC and similarly to oral antihistamines, with good safety data. It is neither recommended as monotherapy nor as first-line treatment.638

Decongestants, both oral and intranasal, have shown to be effective to reduce nasal congestion in the short-time, but adverse effects outweigh the benefits especially in the presence of other comorbidities, so that its generalized use is not recommended. Intranasal decongestants used for more than 5 days may cause rhinitis medicamentosa.638

Oral or parenteral glucocorticoids can improve the symptoms of RA, but should not be prescribed routinely because of their adverse effects on the hypothalamic-pituitary axis, growth and the musculoskeletal system, digestive system, control of glycemia, blood pressure and emotional status.638

Intranasal chromones (cromoglycate and nedocromil) have shown efficacy for reducing sneezing, rhinorrhea, and nasal congestion with fewer adverse effects, although these drugs are less effective than INGC.638

Intranasal anticholinergics (ipratropium bromide) reduce rhinorrhea, although are associated with some adverse effects, such as nasopharyngeal irritation, headache, and oral and nasal mucosa dryness. It is recommended to be added to INGC to improve excessive rhinorrhea.638

The anti-IgE monoclonal antibody, omalizumab, has show to reduce symptoms and the use of rescue medication as well as to improve quality of life as compared with placebo, with a low risk of local reactions at the site of injection or anaphylaxis. Its use could be considered as an add-on treatment in severe uncontrolled cases or to reduce the risk of anaphylaxis in patients treated with allergenic vaccines, although at the present time AR is not included as an indication in the technical specifications of the product.638

Immunotherapy with allergens is effective and cost-effective for the treatment of adult and pediatric AR caused by pollens and dust mites when administered both subcutaneously and orally (sublingual route). It may alter the natural course of the respiratory allergic disease, decreasing the frequency of appearance of asthma and preventing new sensitizations, and is effective for treating symptoms of both asthma and rhinitis.638

The combination of several avoidance measures of indoor allergens added to baseline pharmacological treatment is also and effective option.638

The principles of treatment of rhinitis in childhood are the same than in adulthood, but special attention should be paid to adverse events. Doses should be adequate and, in some cases, the age of the patient should be considered when prescribing certain drugs.683–685

6.5Rhinosinusitis. Nasal polyposis

Chronic rhinosinusitis (CRS) is defined as an inflammatory disorder of the nose and paranasal sinuses, characterized by the presence of at least two symptoms, one of which should be nasal obstruction and/or rhinorrhea, and/or facial pain/pressure, and/or hyposmia/anosmia for at least 12 weeks.686 There are two phenotypes of CRS, with nasal polyps (CRSwNP) and without nasal polyps (CRSsNP), with differences in the inflammatory profile and therapeutic response to treatment.686,687

In Europe, the prevalence of CRS is 10.7%.688

Patients with CRS have a 3.5-fold higher risk for asthma.631Acetylsalicylic acid-exacerbated respiratory disease (AERD) or NSAID- exacerbated respiratory disease associated with asthma, CRSwNP, and NSAID intolerance is more severe and has a poorer prognosis.689 In patients with asthma, the prevalence of AERD is 7-15%, which increases with a greater severity of asthma.690

The severity of CRS can be evaluated using a visual analogue scale, nasal endoscopy to assess the size of polyps, and/or using validated questionnaires, such as SNOT-22, to assess the impact on the quality of life.686,691

Imaging studies do not add value to endoscopic diagnosis692 and should be reserved for surgical planning (computerized tomography), suspicion of complications or nasal or sinus tumors (magnetic resonance).693

Medical treatment of chronic rhinosinusitis with nasal polyps (CRSwNP) is based on the continuous and prolonged use of INGC (beclomethasone, mometasone, fluticasone, budesonide) (fig. 14).694 A greater efficacy of one active principle compared to another has not been demonstrated, although high doses are more effective than low doses.695–697

Figure 14.

Treatment algorithm of nasosinusal polyposis based on the POLINA consensus.694

NSAID: non-steroidal anti-inflammatory drugs. AERD: aspirin-exacerbated respiratory disease/NSAID. COPD: chronic obstructive pulmonary disease. VAS: visual analogue scale. CRSwNP: chronic rhinosinusitis with nasal polyps. SNOT-22: Sino-Nasal Outcome Test 22.

aIn spray, drops or irrigations. bIrrigations with isotonic saline or Ringer lactate. cSee POLINA criteria for the control of CRSwNP (Figure 13.6). dShort courses from 5 días af doses 0.5-1mg/kg/day. eOpening of affected paranasal sinuses. fPossible selection according to endotype. gEvaluate more radical/extensive surgery according to doctor-patient consensus.

(0.53MB).

Short courses of oral glucocorticoids (prednisone, methylprednisolone or deflazacort, administered for 2 to 4 weeks) associated with intranasal glucocorticoids significantly improve nasal congestion and reduce the size of polyps.698

Endoscopic nasal and sinus surgery should be indicated in patients in which medical treatment has been unsuccessful to achieve an adequate control of the disease.699,700

INGC should be used after surgery for the prevention of relapses and to improve outcome.701 The need of revision surgery depends on the previous surgical procedures and postoperative medical treatment, the probability being greater in AERD.672,702

An adequate medical/surgical control of CRS improves clinical and functional parameters of asthma.703,704

Other treatment options associated with the use of INGC that have shown some efficacy are: montelukast (particularly in allergic patients or AERD)705 and clarithromycin.706

Up to 40% of patients have poor control of the disease,707 evidencing the need to identify specific phenotypes that allow predicting therapeutic success.684 Recent studies with different monoclonal antibodies, such as omalizumab (anti-IgE),708,709mepolizumab,710 reslizumab (anti-IL5),711 and dupilumab (anti-IL4-receptor a)712–714 have shown an improvement in the size of nasal polyps, nasal symptoms including olfaction, and quality of life. Mepolizumab and dupilumab have demonstrated a mild to moderate reduction in the indication of surgery.710,714 In patients with severe CRSwNP in whom pharmacological treatment and/or nasosinusal surgery have not achieved an adequate control of the disease, the use of biologics with dupilumab,714mepolizumab715 and omalizumab709 has been recently approved in the European Union.

The use of monoclonal antibodies for the treatment of CRSwNP should be considered for patients resistant to appropriate medical treatment (mainly intranasal and oral glucocorticoids) and failure of at least one endoscopic sinonasal surgical procedure716–720 as described in table 45 based on the POLINA consensus.694

Table 45.

Criteria for the indication of biologics in the treatment of nasal poliposis proposed by the POLINA consensus.694

However, it is necessary to conduct studies under clinical practice conditions and patient record analysis that allow for a better establishment of its effectiveness, as well as the duration of treatment and its cost-effectiveness. In the future, biomarkers should be available to identify the clinical and inflammatory characteristics of candidates to receive them.

4.5Recommendations

6.1. It is recommended to classify allergic rhinitis according to duration into intermittent and persistent, and according to severity into mild, moderate, and severe.  R1 
6.2. The diagnosis of rhinitis is established by clinical criteria and allergy testing.  R1 
6.3. Patients diagnosed with asthma should be assessed for the presence of chronic rhinitis and rhinosinusitus with nasal polyps and vice versa, to implement an integral trearment strategy.  R1 
6.4. For the pharmacological treatment of allergic rhinitis, it is recommended the use of oral and/or nasal topical second-generation antihistamines, intranasal glucocorticoids, or the association of these medications in case of lack of response or moderate to severe disease.  R1 
6.5. In appropriately selected patients (adults and children), immunotherapy with allergen extracts is recommended for the treatment of allergic rhinitis.  R1 
6.6. In patients with chronic rhinosinusitis with nasal polyposis, continuous use of intranasal glucocorticoids is recommended. The use of short-courses of oral glucocorticoids is indicated in severe cases and exacerbations.  R1 
6.7. En In patients with poor control of chronic rhinosinusitis with nasal polyposis despite maximum medical treatment, it is recommended to consider the surgical option followed by post-surgical treatment with intranasal glucocorticoids.  R1 

7Severe uncontrolled asthma7.1Concepts and definitions

Severe asthma is characterized by the need to be treated with multiple drugs at high doses (steps 5-6 of GEMA and step 5 of GINA; see chapter 2.5). Severe asthma includes both controlled and uncontrolled asthma patients.721

Severe asthma is associated with a higher consumption of economic resources as compared with moderate or mild asthma.722–724

Severe uncontrolled asthma (SUA) has received multiple and varied terms and there is no consistent agreement for its terminology.

SUA is defined as the asthma disease that remains poorly controlled despite treatment in the previous year with a combination of inhaled glucocorticoids at high doses/long-acting ß2-adrenergic agonists (ICS/LABA), and long-acting anticholinergics (LAMA) or requiring maintenance oral glucocorticoids therapy (treatment for 6 months a year, independently of the doses or cumulative doses >1g of prednisone or equivalent, independently of the duration).725 Lack of control will be identified by any of the following characteristics (table 46):

  • Asthma Control Test (ACT) <20 or Asthma Control Questionnaire (ACQ) >1.5.

  • ≥ 2 severe exacerbations or having been received ≥ 2 courses of oral glucocorticoids (≥ 3 days each) in the previous year.

  • ≥ 1 hospitalization for a severe exacerbation episode in the previous year.

  • Chronic airflow limitation (forced expiratory volume in one second/forced vital capacity [FEV1/FVC] ratio <0.7 or FEV1 <80% predicted) after the use of an adequate treatment (as long as the better FEV1 will be higher than 80%).

Table 46.

Severe uncontrolled asthma: definition and control.

It is defined as the asthma disease that persists poorly controlled despite treatment with a combination of ICS/LABA/LAMA at high doses in the previous year, or oral glucocorticoids for at least 6 months during the same period.The lack of control is shown by:– ACT <20 or ACQ >1.5.– ≥ 2 severe exacerbations or having being received ≥ 2 courses of oral glucocorticoids (≥ 3 days each) in the previous year.– ≥ 1 hospitalization for a severe exacerbation episode in the previous year.– Chronic airflow limitation (FEV1/FVC ratio <0.7 or FEV1 <80% predicted) after the use of an adequate treatment (as long as the better FEV1 will be higher than 80%). 

It is important to exclude external factors that may contribute to poor asthma control before defining SUA (section 7.2.2).726–730

Some studies have shown a prevalence of SUA between 3% and 4% among patients with asthma.731,732

SUA can be corticosteroid-dependent or corticosteroid-resistant to a higher or lesser extent.733–735

Corticosteroid-dependent SUA is defined when continuous treatment with oral or parenteral glucocorticoids for disease control is required, the disease is insensitive to glucocorticoids and shows a FEV1 ≤ 75% that does not improve significantly (≤ 15%) after treatment with oral prednisone, 40mg/day for 2 weeks.736,737

7.2Diagnosis and evaluation

When SUA is suspected, it is advisable to perform a systematic evaluation, preferably in specialized asthma centers or units, using a multidisciplinary approach, following a sequential stepwise diagnostic algorithm726,738–741 (fig. 15).

Figure 15.

Diagnostic algorithm based on the sequential stepwise approach for SUA.

(0.25MB).

The use of this multidimensional approach has shown good clinical results and to be cost-effective.742–744

7.2.1Diagnostic confirmation of asthma

It has been estimated that between 12% and 30% of patients with suspected SUA do not have asthma.726,745–747

It should be confirmed that the diagnosis of asthma has been made correctly and, in case of doubt, studies aimed to demonstrate objectively the presence of airflow obstruction, variability and/or bronchial hyperresponsiveness (see chapter 2.2) should be performed. If diagnosis cannot be confirmed, other diseases mimicking asthma should be excluded through the rational and progressive use of work-up studies summarized in table 47.

Table 47.

Differential diagnosis in asthma: diseases mimicking asthma and their corresponding diagnostic tests.

Differential diagnosis  Diagnostic tests 
– Organic upper airway diseases– Dynamic airway collapse– Bronchial obstruction  – Spirometry with inspiratory loop– Computerized tomography (CT) obtained in inspiration and expiration of the upper airway– Fiberoptic bronchoscopy 
– Inducible laryngeal obstruction (ILO)  – Laryngoscopy/video-stroboscopic during exacerbation or after challenge with methacholine or after ergometry 
– Chronic obstructive pulmonary disease (emphysema)  – Chest CT– Plethysmography and CO diffusing capacity 
– Bronchiolitis obliterans  – Chest CT obtained in inspiration/expiration– Plethysmography/air trapping– Biopsy transbronchial/pulmonary 
– Functional dyspneas/hyperventilation syndrome  – Cuestionario de hiperpercepción (de Nijmegen)– Psychological evaluation 
– Left heart failure  – Chest CT– Electrocardiogram/echocardiogram 
– Bronchiectasis– Cystic fibrosis– Allergic bronchopulmonary aspergillosis (ABPA)  – Chest CT– Sweat test/genetic study– Total and Arpergillus-specific IgE/precipitins 
– Eosinophilic granulomatosis with polyangiitis (EGPA)– Pulmonary eosinophilias  – pANCA/biopsy of organ(s) affected– Fiberoptic bronchoscopy (with bronchoalveolar lavage) 

pANCA: perinuclear anti-neutrophil cytoplasmic antibodies.

7.2.2Identification of external factors

It is necessary to identify and evaluate some factors unrelated to the disease, the presence of which can contribute to poor control of asthma. These factors can be grouped into the following categories:

  • Factors directly related to the patient: treatment adherence and inhalation technique. Up to 50% to 80% of cases of SUA are caused by inadequate adherence or by a deficient inhalation technique.732,745,748

    Therefore, adherence should always be evaluated (preferably using validated questionnaires or information on dispensing prescriptions in the the community pharmacy) and the inhalation technique (direct observation) (see chapter 3.4).

  • Factors related with comorbidities and aggravating conditions. Different diseases or processes when present concomitantly with asthma can contribute to an insufficient control of the disease. It has been shown that 92% of patients with SUA suffer from at least one of these conditions, which in turn are more prevalent than in patients without SUA.730

    Table 48 summarizes the most commonly cited comorbidities and their corresponding tests for evaluation, diagnostic confirmation, and treatment approach.738,740,741,749,750

    Table 48.

    Common comorbidities and aggravating factors of asthma with their corresponding diagnostic tests and treatment.

    Comorbidity  Diagnostic tests  Treatment 
    Sinonasal diseaseRhinoscopy/nasal endoscopy  Intranasal glucocorticoids 
    Sinus imaging studies (CT/MR)Nasal Iavages/antileukotrienes 
    Endonasal surgery 
    Gastroesophageal refluxpH-metry/esophageal manometry  Hygienic-dietetic counselling 
    Treatment test with PPI  Proton pump inhibitors 
    Upper digestive endoscopy  Surgical repair 
    ObesityBMIWeight loss 
    Bariatric surgery 
    Sleep apnea syndrome (SAS)PolysomnographyCPAP 
    Weight loss if necessary 
    Psychopathology (anxiety, depression)  Psychologist/psychiatrist evaluation  Psychotherapy/specific treatment
    Fibromyalgia  Rheumatological evaluation 
    Functional dyspneaSpecific questionnaires (Nijmegen questionnaire)Psychotherapy 
    Respiratory re-education 
    Inducible laryngeal obstruction (ILO)Laryngoscopy in exacerbation or methacholine/exercise challengeLogophoniatric rehabilitation 
    Treatment of comorbidities: reflux 
    Drugs: NSAID, non-selective β-blockers, ACE inhibitors  Clinical history  Substitution 
    Tobacco and other inhalation toxics  Questioning  Cessation/quit 

    NSAID: non-steroidal anti-inflammatory; ACE: angiotensin-converting enzyme; CT: computed tomography; MR: magnetic resonance; PPI: proton pump inhibitors; BMI: body mass index; CPAP: continuous positive airway pressure.

  • Factors related to triggers of exacerbations. It is necessary to identify whether exposure to triggers of exacerbations are present (see table 4), particularly active and passive smoking, e-cigarettes, cannabis inhalation, allergen exposure (mites, pollens, fungi, dander, cockroaches, etc.), indoor and outdoor air contamination, occupational agents, molds and harmful chemical products, drugs such as non-cardioselective ß-blockers, non-steroidal anti-inflammatory drugs (NSAID), and angiotensin-converting enzyme (ACE) inhibitors.738,740

    Moreover, lack of response due to SABA abuse (by downregulation of ß2 receptors and increase of bronchial hyperresponsiveness [BHR]) has been reported.751,752

7.2.3Establishment of phenotype

Classification into phenotypes aims to identify specific patients who are candidates for a particular treatment739,753 (see section 7.3). Currently, specific biomarkers for each phenotype/endotype are not available.754

The minimum follow-up period, by a specialist or a specialized asthma unit, to accept the diagnosis of SUA is 6 months.728,738,741

7.3Phenotypes of severe uncontrolled asthma

Severe asthma is a heterogeneous syndrome with multiple clinical variants. Over the past two decades, there has been intense research focusing on the study, discovery, and refinement of phenotypes of SUA.755–763

Phenotype is defined as an observable characteristic of severe asthma that can be associated with an underlying mechanism, named endotype. It is important to differentiate phenotype from comorbidities, since comorbidities coexist with SUA but their treatment is different.

Establishing the asthma phenotype in patients with SUA constitutes part of the diagnostic or assessment action to be carried out in these patients, as it may entail differential treatment modalities and has prognostic implications.727,764–766

Studies based on biostatistical analyses of cases clustered according to natural history, pathobiology, clinical features (age, onset, allergy symptoms, involvement of the upper respiratory tract, body mass index [BMI], acetyl salicylic acid-exacerbated respiratory disease [AERD], pulmonary function, biomarkers (peripheral blood and sputum eosinophils, immunoglobulin E [IgE], fractional exhaled nitric oxide [FENO], induced sputum neutrophil count) and therapeutic response have identified the existence of different phenotypes.739,753,767–770 Two inflammatory patterns have been defined: T2 (present in allergic and eosinophilic asthma) and non-T2. In clinical practice, three SUA phenotypes stand out with implications in treatment decision-making:

  • Type 2 (T2) allergic phenotype.

  • Type 2 (T2) eosinophilic phenotype.

  • Non-T2 phenotype (table 49).

    Table 49.

    Severe asthma phenotypes.

    Phenotypes  Clinical characteristics  Biomarkers  Treatment 
    Allergic (T2)  Allergic symptoms+Allergen sensitization(prick test and/or specific IgE)  Specific IgETh2 cytokines PeriostinSputum eosinophils and neutrophils  Glucocorticoids OmalizumabAnti-IL-5/anti-IL-5R- (mepolizumab, reslizumab, benralizumab)Dupilumab Tezepelumab 
    Eosinophilic (T2)  Chronic rhinosinusitis/nasalpolyposisAERDCorticoid-depedent or refractoryto glucocorticoids  Blood and sputum eosinophilsIL-5Cysteinyl-leukotrienes  LTRAAnti-IL-5/anti-IL-5R- (mepolizumab, reslizumab, benralizumab)Dupilumab Tezepelumab 
    Non-T2  Lower FEV1Greater trappingSmoking history  Neutrophils orpaucigranulocytic in sputumTH17 activationIL-8  Azythromycin TezepelumabThermoplasty 

    IgE: immunoglobulin E; AERD: acetyl salicylic acid-exacerbated respiratory disease. FEV1: forced expiratory volumen in one second; LTRA: letra leukotriene receptor antagonist; IL: interleukin.

However, both T2 phenotypes may show some degree of overlap.

7.3.1Allergic asthma (T2)

Allergic asthma accounts for 40-50% of severe cases of asthma, and has an atopic underlying mechanism mediated by the activation of type-2 helper T lymphocytes (Th2), the production of interleukin (IL) 4, IL-5 and IL-13, and an isotype shift within B lymphocytes towards IgE production. Allergic bronchopulmonary aspergillosis (ABPA) is a particularly severe variety of allergic asthma that shows a pure eosinophilic or mixed (eosinophilia and neutrophilia) inflammatory pattern in sputum. Periostin (an IL-13-induced cell matrix protein), which can be measured in blood and bronchial secretions, and the fractional exhaled nitric oxide (FENO) are good biomarkers of the “increased” T2 variant.771–774 The diagnosis requires the demonstration of sensitization to an allergen and the triggering of symptoms with exposure to such allergen.

7.3.2Eosinophilic asthma (T2)

It accounts for more than 25% of severe asthma cases and is characterized by the presence of eosinophils in bronchial biopsies and sputum samples despite treatment with glucocorticoids at high doses. Chronic rhinosinusitis and nasal polyps may also occur. A subset of patients develops AERD. Although eosinophilic asthma is associated with a lower prevalence of atopy, IgE and FENO may be increased. Alterations of the arachidonic acid metabolism are involved in the pathogenesis of this phenotype of asthma. A high production of IL-5 may explain the eosinophilic inflammation in the absence of the traditional allergy-mediated T2 mechanism.775–778

7.3.3Non-T2 asthma

This phenotype of asthma occurs without eosinophilia, neither in the peripheral blood, nor in sputum. It frequently shows a paucigranulocytic profile, neutrophilia, scarce local eosinophilia, low FENO levels, and a poor response to glucocorticoids. It can be accompanied by chronic airflow limitation with important air trapping and, frequently, history of smoking is present.779,780 It should be taken into account that inflammatory biomarkers of type T2 phenotype (peripheral blood eosinophils, sputum eosinophils, and FENO) are frequently suppressed by oral glucocorticoids. In our opinion, analysis of peripheral blood eosinophils and FENO should be repeated up to three times (e.g. when asthma worsens, before the administration of oral glucocorticoids), before assuming that asthma does not belong to the T2 phenotype.

In the GINA 2019, the possibility of type 2 refractory inflammation is considered, in the presence of any of the following findings in a patient taking ICS at high doses or daily oral glucocorticoids:740

  • -

    Peripheral blood eosinophils ≥ 150/μl, and/or FENO ≥ 20 ppb, and/or

  • -

    Sputum eosinophils ≥ 2%, and/or

  • -

    Asthma is clinically induced by allergens.

7.4Treatment7.4.1General measures

Asthma education. Asthma education activities are not different from that usually recommended for the remaining asthma population (see chapter 3.5). However, approaches such as maximizing avoidance measures and smoking cessation should be implemented, with special emphasis to confirm objectively that adherence to treatment and the inhalation technique are both correct. At present, there are different devices for remotely adherence monitoring.781,782

Background pharmacological treatment. According to the inclusion criteria defining SUA, in patients on maintenance therapy with a combination of ICS/LABA at high doses it is advisable to add, at least, a third controller drug, usually tiotropium783,784 (see chapter 3.2).

Treatment of comorbidities. If either an associated comorbid condition or an aggravating factor has been identified, the appropriate therapeutic measures should be initiated (table 48).738,740,749,750

7.4.2Phenotype-directed treatment

Patients with SUA according to the pathophysiological underlying mechanism (T2 or non-T2 asthma) and the presence or absence of different inflammatory markers are classified into the aforementioned phenotypes (see section 7.3).

Inflammation markers of T2 phenotype may be suppressed by treatment with oral glucocorticoids (OCS); therefore, they should be preferably measured before starting treatment with OCS or at the lowest possible dose, and at least on three occasions (e.g. during an exacerbation), prior to assume that a patient presents a non-T2 phenotype. In corticosteroid-dependent patients, it is important to check their historical values.

A phenotype-directed treatment algorithm is proposed in the present guideline (fig. 16 and table 50); the different monoclonal antibodies available for treating SUA are shown together with their main characteristics.

Figure 16.

Treatment of SUA according to phenotype.

SUA: severe uncontrolled asthma; ICS: inhaled glucocorticoids; LABA: long-acting β2-adrenergic agonists; LAMA: long-acting cholinergic agonists; CRSwNP: chronic rhinosinusitis with nasal polyposis; HES: hypereosinophilic syndrome; EGPA: eosinophilic granulomatosis with polyangiitis; PMN: polymorphonuclear neutrophils; ACT: Asthma Control Test; ACQ: Asthma Control Questionnaire; SNOT-22: Sinonasal Outcome Test; VAS: visual analogue scale; FENO: fractional exhaled nitric oxide; EOS: eosinophils; aSensitivity to allergens and presence of compatible clinical features, and total IgE ≥ 75 IU; bOmalizumab if IgE ≥ 75 U/l and EOS <150μl; cDupilumab if EOS ≥300μl and/or FENO50 ppb and between 150-300 EOS and FENO25 ppb. Not recommended if EOS ≥1,500μl. dTezepelumab if FENO25 ppb; eMepolizumab if current EOS ≥150μl and ≥300μl in the previous 12 months. fBenralizumab if current EOS ≥150μl and nasal polyposis or ≥3 severe exacerbations in the previous year or FCV <60%; gReslizumab if EOS ≥400μl.

Notes

Definitions

SUA: asthma requiring treatment with 5-6 therapeutic steps according to GEMA and presents ≥1 of the following criteria:

  • ACT <20 or ACQ ≥1.5.

  • 2 courses of oral corticoids (OCS) during ≥3 days

  • in the previous year.

  • 1 hospital admission due to asthma exacerbation in the previous year.

  • FEV1 ≤ 80% predicted.

Type 2 refractory inflammation:1 of the following criteria in a patient using inhaled glucocorticoids (ICS) at high doses or daily OCS:

  • 150 eosinophils per microliter in blood

  • FENO ≥25 ppb/ul (American Thoracic Society Committee).

  • 2% eosinophils in sputum.

  • Asthma is clinically induced by allergens.

Patients requiring maintenance treatment with oral glucocorticoids can also have an underlying type 2 inflammation. However, OCS often suppress type 2 inflammation biomarkers (blood and sputum eosinophils and FENO). Therefore, when possible, these tests should be performed before starting a short course or maintenance treatment with OCS, or when the patient receives the lowest possible dose of OCS.

Thresholds of peripheral blood eosinophilia: At least one analytical result of more than 300 Eos/μl in the last year. Low values of eosinophils may appear in patients recently treated or on chronic treatment with systemic glucorticoids. In this case, it can be useful to review the patient's historical values.

Thresholds of FENO. The cutoff value is established at 25 ppb. However, it should be considered that results of FENO measurement can altered by the recent use of systemic glucocorticoids and total dose of inhaled glucocorticoids, age. and current smoking (lower values in smokers). In the presence of high FENO levels, it is necessary to confirm that self-administration inhaled medication is correct (treatment adherence and inhalation technique).

Response to a biological drug. It is defined by:

  • ACT score equal or higher than 20 or a significant change as compared with baseline score (≥3 points).

  • Absence of hospital admissions or visits to the emergency room.

  • More than 50% reduction of exacerbations.

  • Suppression of the use of oral glucocorticoids or significant decrease of doses (≥50%).

Choice among monoclonals

The order in which biologics appear in the scheme when they coincide for the same indication only takes into account the time since each drug has been commercialized.

In the choice of biologics should be considered: blood eosinophil count, pulmonary function, use of maintenance treatment with oral glucocorticoids, presence of comorbidities: nasal polyposis/AERD, chronic urticaria, atopic dermatitis and asthma-associated diseases (eosinophilic granulomatosis with polyangiitis, eosinophilic pneumonia, allergic bronchopulmonary aspergillosis, eosinophilic esophagitis).

  • Benralizumab (higher efficacy ≥300 eosinophils/μl): patients with poor pulmonary function, polyposis, maintenance with oral glucocorticoids and difficult access to asthma unit living far away [long distances]).

  • Reslizumab (higher efficacy ≥400 eosinophils/μl): improves pulmonary function. Not effective for reducing OCS doses. Intravenous administration.

  • Mepolizumab (indication from 150 eosinophils/μl but higher efficacy ≥500 eosinophils/μl): indicated in patients with ≥150 eosinophils/μl if there are historical values of ≥300 eosinophils/μl. It has been shown that allows reduction of withdrawal of OCS.

  • Dupilumab (higher efficacy ≥300 eosinophils/μl and/or FENO ≥50 ppb): improves pulmonary function, nasal polyposis and severe dermatitis. It has been shown that allows reduction of withdrawal or OCS and increases eosinophil values. Administration every 15 days.

To choose between drugs with potential efficacy in a given patient, criteria of posology, patient's preferences, and costs should be also considered.

Thermoplasty is indicated in patients neither with emphysema/bronchiectasis/atelectasis nor with important comorbidities, without treatment with anticoagulants or immunosuppressants, and who do no present recurrent infections. FEV1 should be greater than 40% and any contraindication for fiberoptic bronchoscopy with sedation should be absent.

(0.38MB).
Table 50.

Biologics approved for the treatment of SUA and their characteristics.

Biologic(SUA)  Approval:TPR Spain  Mechanism of action  Evidences  Adverse events (“frequent” according to technical specification)  Administration 
Omalizumab  >6 years with severe allergic asthma and sensitization to perennial allergens with IgE between 30-1500 UI/ml and FEV1 <80%  Binds circulating IgE preventing binding to high and low affinity receptor (FcɛR1) for IgE  34% reduction of exacerbations but no improvement of symptoms, HRQoL and pulmonary function in RCT.Efficacy in nasal polyposis  Injection site reactions, headache, upper abdominal pain  75-600 mg s.c. route every2-4 weeks according to weight and IgE. Possible administration at home 
Mepolizumab  ≥ 6 years with refractory eosi nophilic asthma with Eos ≥ 500 or <500 with 2 severe exacerbations or 1 hospitalization in the previous year  Blocks IL-5 from binding to the IL-5 receptor  53% reduction of severe exacerbations and improvement of HRQoL, control of symptoms and pulmonary function in RCT. Reduces doses of maintenance OCS. Efficacy in nasal polyposis  Injection site reactions, headache, pharyngitis, pyrexia, upper abdominal pain, eczema, back pain, Hypersensitivity reactions  6-11 years: 40 mg every4 weeks≥ 12 years: 100 mg every4 weeksPossible administration at home 
Reslizumab  >18 years with severe eosinophilic asthma on treatment with ICS at high doses plus another controller with Eos ≥ 500 or between 400-500 and 2 severe exacerbations or 1 hospitalization in the previous year  Binds to the same domain that IL-5 receptor blocking binding of IL-5 to its receptor  54% reduction of exacerbations in patients with ≥ 400 Eos and ≥ 1 exacerbation in the past year  Increase of blood CPK  3 mg/kg i.v. route every 4 weeksDay hospital 
Benralizumab  >18 years with severe eosinophilic asthma on treatment with ICS at high doses plus LABA with Eos ≥ 500 or <500 with 2 severe exacerbations or 1 hospitalization in the previous year  Binds Fα of IL-5 receptorinhibiting its activation.Induces direct elimination (by Ac-mediated cytotoxicity) of eosinophils and basophils through natural killer (NK) cells  57% reduction of exacerbations in patients with ≥ 300 Eos and ≥ 3 exacerbations in the past year; and improvements of pulmonary function and reduction of OCS doses  Injection site reactions, pharyngitis, headache, hypersensitivity reactions  30 mg s.c. every 8 weeks (with the first 3 doses at one month interval)Possible administration at home 
Dupilumab  ≥ 6 years with severeasthma with T2 markers (Eos≥ 300 o FENO ≥ 25 ppb) or corticosteroid-dependent  Blocks subunit α of IL-4 receptor (anti-IL-4 and IL-13 effect)  50% reduction of severe exacerbations and improvement of HRQoL, control of symptoms and pulmonary function in RCT. Reduces maintenance doses of OCSEfficacy in nasal polyposis  Injection site reactions, transient blood eosinophilia (4-13%)  Initial dose 400 mg followed by:200 mg s.c. every 2 weeks (severe eosinophilic asthma/T2)300 mg in corticoid-dependent or with associated atopic dermatitis. Possible administration at home.Dosage in patients between 6 and 11 yeats of age is described in section 7.5 
Tezepelumab  Indicated as additional maintenance treatment in adults and adolescents ≥ 12 years of age with severe asthma not adequately controlled despite ICS at high doses combined with another drug for maintenance treatment*  Human monoclonal antibodu (IgG2λ) directed against TSLP, bronchial epithelial-cell derived cytokine member of the alarmin family  Significant reduction of exacerbations (66-71%) and bronchial hyperresponsiveness, improvement of pulmonary function, control of the disease, and HRQL. It is also effective in case of blood eosinophils <150 cells/μl and FENO<25 ppb.  Injection site reactions, pharyngitis, arthralgias, skin eruption  210 mg s.c. every 4 weeksPossible administration at home 

TPR: therapeutic positioning report; s.c.: subcutaneous; i.v.: intravenous; HRQoL: health-related quality of life; RCT: randomized controlled trial; Eos: eosinophils. FEV1: forced expiratory volumen in one second; ICS: inhaled corticosteroids; LABA: long-acting ß2-adrenergic agonist; IgE: immunoglobulin E; OCS: oral glucocorticoids; CPK: creatine phosphokinase; Ac: antibody.

*

Approved by the EMA (no TPR available in Spain at the time of publication of GEMA 5.3).

7.4.2.1Treatment of T2 asthma

Considering the level of peripheral blood or sputum eosinophils and the presence of relevant allergic clinical manifestations with confirmed sensitization to perennial aleroallergens, one of the available monoclonal antibodies will be selected (fig. 16).741

4.5.1.1Anti-IgE treatment: omalizumab

Monoclonal antibody blocking IgE, with more than 15 years in clinical practice that has shown its efficacy in randomized controlled trials (RCT) reducing severe exacerbations, intensity of symptoms, use of inhaled ICS, and improvement of quality of life.785–789

Omalizumab is indicated in allergic SUA with sensitization to perennial allergens in patients aged ≥ 6 years with serum total IgE values between 30-1500 IU. The dose varies according IgE levels and body weight. The administration route is subcutaneous (s.c.) every 2 or 4 weeks.

Subsequent studies carried out in daily practice conditions have shown a decrease of exacerbations, improvement of quality of life, and reduction of OCS,790 independently of the baseline value of biomarkers791 or the eosinophil count.790

In some cases, after a prolonged period of treatment (5 years), withdrawal of omalizumab is possible. Treatment discontinuation should be performed gradually, on an individual basis, in agreement with the patient and with close monitorization of the control of asthma.792–794

Good results with the use of omalizumab in allergic bronchopulmonary aspergillosis have been reported,795,796 but up to the present time RCTs have not been carried.

4.5.1.2Anti-IL-5/IL-5Ra treatment4.5.1.2.1Mepolizumab

Monoclonal antibody that bocks circulating IL-5. In RCTs, the use of mepolizumab has shown to reduce exacerbations in patients with ≥ 300 eosinophils/μl in peripheral blood during the previous year, or with ≥ 150/μl at the time of treatment but with high historical values.797,798 A post hoc analysis showed a greater reduction of exacerbations (70%) in the group of patients with >500 eosinophils/μl.799 Also, this drug has shown to be effective in reducing the doses of OCS in patients on maintenance treatment with systemic glucocorticoids.800,801 It is indicated in patients with eosinophilic asthma of ≥ 6 years of age, at doses of 100mg s.c. every 4 weeks for patients aged 12 years and older, and 40mg s.c. dose every 4 weeks to patients aged 6 to 11 years.

Studies carried out in routine daily practice802 and those extended a long time have confirmed the efficacy of mepolizumab shown in clinical trials, with a favorable safety profile and stable and long-lasting effect.803,804

The efficacy of this drug has been demonstrated in patients with partial response to Mepolizumab has been also approved for the treatment of hypereosinophilic syndrome (HES). In adolescents and adult patients with severe HES, the use of this drug achieved a reduction in the percentage of patients with exacerbations (worsening of symptoms or increase in eosinophil count requiring escalation to other treatments) and a decrease in annual exacerbations (the open-label extension study confirmed the results of the randomized trial).805,806

4.5.1.2.2Reslizumab

It is a monoclonal antibody against IL-5 that has shown a significant reduction of exacerbations and improvement of current control-related variables in severe asthma with ≥ 400 eosinophils/μl.807–809 The efficacy is independent of allergic sensitization.810 A post hoc study showed a reduction in OCS burden in corticosteroid-dependent asthma.811 It is indicated in patients with eosinophilic asthma >18 years of age, at doses of 3mg/kg i.v. every 4 weeks.

Some studies in small series of patients in which treatment with other monoclonal antibodies (omalizumab and mepolizumab) have been unsuccessful, showed improvement after the use of reslizumab.812,813 Studies at 2 years demonstrate a favorable safety profile.814

4.5.1.2.3Benralizumab

It is a monoclonal antibody binding subunit a of the IL-5 receptor preventing its activation and inducing direct elimination (by antibody-dependent cell-mediated cytotoxicity) of eosinophils and basophils through NK cells; so that, it is known as anti-eosinophilic effect. In pivotal RCTs carried our in eosinophilic SUA, benralizumab has shown to reduce severe exacerbations, improve pulmonary function (FEV1), and decrease asthma symptoms,815,816 particularly in patients with peripheral blood eosinophils ≥ 300μl or ≥ 150μl on maintenance treatment with OCS. It is indicated in patients with eosinophilic asthma aged ≥ 18 years, at doses of 30mg s.c. every 4 weeks for the first 3 doses, and every 8 weeks thereafter.

Also, it has been shown a significant reduction of the doses of OCS and even withdrawal of OCS.817,818

In phase III trials, a number of baseline clinical factors were associated with a greater response, including the use of OCS, history of nasal polyposis, reduced pulmonary function based on FVC <65% and frequent exacerbations.763,819–822

Follow-up studies at 2 and 5 years have confirmed the efficacy and safety results.823,824

Subsequent studies conducted in routine clinical practice have shown a decrease of exacerbations, improvement of quality of life, and a reduction of OCS.825

4.5.1.3Anti-IL4/IL-13 treatment4.5.1.3.1Dupilumab

It is a fully human monoclonal antibody binding receptor a of IL-4, blocking both IL-4 and IL-13. Initial RCTs with this drug have shown a reduction of exacerbations, improvements in quality of life, control of symptoms, and pulmonary function (FEV1) in patients with moderate to severe uncontrolled asthma over 12 years of age. These improvements were also observed in patients with peripheral blood eosinophils between 150 and 300/μl with FENO ≥ 25 ppb, although there was a higher improvement in patients with eosinophil count ≥ 300/μl and/or FENO ≥ 50 ppb.826–828 Further studies have extended the indication to patients ≥ 6 years of age with SUA and high eosinophil counts and/or FENO.829

Reduction and withdrawal of OCS has also been demonstrated in corticosteroid-dependent patients,830 and a better response in patients with higher values of eosinophils and/or FENO.828

The effect on reduction of exacerbations, pulmonary function, and reduction of OCS is maintained over time, according to an extension study of almost 3 years.831

Extension studies have confirmed a favorable safety profile.831 In relation to adverse effects, it should be noted that a patients treated with dupilumab showed eosinophilia, with counts higher than 3,000 cells/μl in a percentage of cases, although without clinical repercussion. In these studies, transient hypereosinophilia did not modify the response to treatment.828–831

4.5.1.4Treatment with anti-thymic stromal lymphopoietin (TSLP)4.5.1.4.1Tezepelumab

It is a human monoclonal antibody that binds thymic stromal lymphopoietin (TSLP), a bronchial epithelium-derived cytokine of the group of alarmins. At doses of 210mg by the subcutaneous route every 4 weeks significantly reduce exacerbations (66-71%) and bronchial hyperresponsiveness,832 improves pulmonary function, control of the disease, and quality of life, independently of baseline levels of T2 inflammation biomarkers (FENO, peripheral eosinophilia, IgE) without achieving a significant reduction of OCS doses.833,834

Extension studies have confirmed the effectiveness shown in clinical trials and a favorable safety profile.835

7.4.2.2Treatment of non-T2 asthma

In patients in whom there is no evidence of the presence of T2 inflammation biomarkers, other therapeutic options should be selected.

4.5.1.4.2Tezepelumab

Although it provides a reduction in the rate of severe exacerbations, more pronounced as more higher blood eosinophils and FENO values at baseline, it is also effective when the blood eosinophil count is <150 cells/μl and FENO <25 ppb. Therefore, it is the only biological drug that currently demonstrates efficacy in non-T2 asthma.833

4.5.1.4.3Azithromycin

Because of their immunomodulatory effect, macrolides have been used in asthma with inconsistent results.775,836,837 In the AMAZES study,838,839 it was found that azithromycin administered at doses of 500mg orally, 3 times a week during 48 weeks, reduced exacerbations and improved quality of life, independently of the inflammatory phenotype.

An individualized indication is recommended in SUA patients with triple therapy with non-T2 phenotype especially if they suffer from frequent exacerbation episodes.740,750

4.5.1.5Bronchial thermoplasty

This bronchoscopic procedure reduces the bronchial smooth muscle layer by heating the tissue through the deliver of radiofrequency energy.840,841

Results of studies of bronchial thermoplasty in patients with moderate and severe asthma showed a significant improvement of the quality of life, control of symptoms, and reduction of exacerbations.842–846 Efficacy regarding reduction of exacerbations is still present after 5 and 10 years of the procedure.840,847,848

This is a therapeutic option to be considered in patient with SUA with phenotypes unsuitable for the use of monoclonal antibodies or in which monoclonal antibodies have been unsuccessful, provided that there are no contraindications to the technique and it is applied in experienced centers.

4.5.1.6Systemic glucocorticoids

In some patients with SUA suffering from an exacerbation episode, treatment with OCS is necessary. Patients requiring OCS courses may present adverse effects, the risk of which increase with the use of ≥ 4 courses of OCS in a year or >30 days a year.849,850

The use of OCS at the minimum necessary dose and for the shortest time possible should be reserved as one of the last alternative for patients in whom control is not achieved with other therapeutic options.851 In these circumstances, preventive or treatment measures for possible adverse effects will be considered.

Some studies with not very robust designs, carried out in small samples of patients showed that intramuscular triamcinolone depot (glucocorticoid with the addition of a fluorine group), in patients with corticosteroid-dependent asthma, compared to the usual OCSs, provided a significant reduction of exacerbations, an increase in pulmonary function, and fewer side effects.852,853 However, they are not free of adverse effects and the pharmacokinetic profile is unknown.

4.5.1.6.1Recommendations for taperingl of oral glucocorticoids

Some studies have shown that the use of OCS is not uncommon.854,855

A significant dose-response relationship between chronic use of OCS and the risk of complications has been observed.856

Every patient undergoing treatment with OCS should undergo proper monitoring and management of possible side effects. A protocol to this purpose is shown in table 51.

Table 51.

Protocol of monitoring side effects of systemic glucocorticoids.

Osteoporosis  Annual height measurement.Evaluation of the risk of fractures.Bone densitometry before treatment with SGC and at one year:• If low BMD, repeat at one year.• If normal BMD, repeat at 2-3 years. 
Adrenal insufficiency  If the use of SGC exceeds 2 consecutive weeks continuously or 3 cumulative weeks in the previous 6 months:• Evaluation of symptoms.• Baseline cortisol assay (8-9 a.m). 
Ophthalmological  Annual ophthalmological examination.Early referral for ophthalmological examination (in the presence of cataracts and/or risk factors for glaucoma). 
Cardiovascular  Calculate the risk according to the Framingham index.Lipid profile after one month of starting treatment with SGC, repeat every 6-12 months. 
Hyperglycemia  Glycemia every 3-6 months on the first year, and then annually. 
Infections/pneumonias  Alert the patients to seek medical care in the presence of fever and/or symptoms of infection.Microbiological surveillance. Serial sputum cultures. 

BMD: bone mineral density; SGC: systemic glucocorticoids.

Modified from Liu et al., 2013857.

In a recent Dephi expert consensus study,858 the authors made a series of recommendations regarding the use of OCS:

  • Minimize OCS use as much as possible.

  • An annual cumulative dose of 0.5 to 1g would be indicative of poor asthma control.

  • Primary care physicians prescribing at least 3 courses of OCSs per year to a patient should consider specialist referral.

  • In all patients who have received a cumulative dose of OCS exceeding 2g in the past year, a fasting morning cortisol test (8 a.m.) should be performed when attempting to reduce the OCS dose below 5mg/day.

  • When no other alternative is available, a dose of ≤ 5mg/day of prednisone (or equivalent) would be considered acceptable.

Regarding tapering of OCS doses in patients starting treatment with biologics, the following should be considered:

  • The initial tapering of high OCS doses (e.g. 20mg/day) can proceed at a faster pace (e.g. 10mg/week or 30-50% reductions every 2-4 weeks). Reduce 2.5 to 5mg every 0.5 to 2 weeks until the physiological dose is achieved (e.g. 5-10mg/day) and then proceed to a lower pace (1-2.5mg every 1-2 weeks).

  • When a reduction in the OCS dose by 5mg/week fails, a slower and lower dose reduction of 1mg/week should be attempted.

  • If mild symptoms occur, maintain the current dosage; they are likely to resolve as endogenous-axis recovery occurs.

  • If intolerable symptoms occur, return to the previous (efficacious) dose and then later consider reattempting tapering at a slower pace.

Although there is no unanimous agreement, a scheme for reducing the dose of OCS in patients starting treatment with biologics has recently been proposed.859

Figure 17 shows the algorithm proposed for the treatment of corticosteroid-dependent severe asthma in adults725 and fig. 18 the algorithm for down-titration doses of OCS.

Figure 17.

Treatment of corticosteroid-dependent severe asthma in adults (based in part on the 2022 SEPAR consensus860 (OCS: oral glucocorticoids; ACT: Asthma Control Test; ACQ: Asthma Control Questionnaire; SNOT: Sinonasal Outcome Test Questionnaire; VAS: visual analogue scale).

(0.32MB).
Figure 18.

Algorithm for the evaluation of adrenal function during down-titration of OCS dosage.

(Modified from Menzies-Gow et al., 2022).859

*It can be directly substituted by hydrocortisone 20mg/day, preferable at breakfast.

**Do not take OCS on the previous night nor in the morning before blood sampling.

(0.32MB).
7.4.2.3New investigational treatments for SUA

Other new molecules such as antagonists of IL-33 and its receptor are under study. Of these, astegolimab, an anti-ST2 antibody, has shown efficacy in reducing exacerbations in eosinophil-high (> 300 cells/μl) and non-T2 inflammation (< 300 cells/μl) in a similar proportion.861

Interleukin-33 blockade with itepekimab led to a lower incidence of events indicating a loss of asthma control than placebo and improved lung function in patients with moderate-to-severe asthma.862

7.5Severe uncontrolled asthma in children7.5.1Epidemiology. Definition

Severe asthma in childhood is more common from school age863,864 with a prevalence of 2-5%.865,866 It is associated with a high morbidity,867 costs,868 and future risk of chronic obstructive pulmonary disease (COPD).869,870

The clinical presentation and response to treatment vary from infants to adolescents.871,872

In children with severe recurrent exacerbations, and in younger than 5 years of age, with or without symptoms between episodes, a diagnosis of SUA may be considered when despite a correct treatment with ICS at high doses, the following events are present:

  • 1)

    ≥ 1 admission to an intensive care unit,

  • 2)

    ≥ 2 hospital admissions requiring intravenous therapy or

  • 3)

    ≥ 2 courses of OCS in the previous year.873

The definition for children older than 5 years of age coincides with that for adults.726

7.5.2Evaluation

A cost-effective multidimensional, Multidisciplinary, and stepwise evaluation is necessary necesaria874,875 (fig. 19).

Figure 19.

Severe uncontrolled asthma in children: stepwise evaluation.

(0.19MB).

Up to 50% of patients present potentially avoidable factors and/or associated comorbidities responsible for difficult control of their asthma.726,876

4.5.1.7Diagnostic confirmation

Up to 12-30% of patients with SUA may be diagnosed with other diseases mimicking symptoms of asthma.726

A detailed medical history, physical examination, and pre- and post-bronchodilation spirometry are necessary. Many children with SUA have normal pulmonary function,877 requiring a bronchoprovocation test. In addition, other complementary tests oriented by the clinical suspicion or atypical presentation will be necessary. Also, in children with SUA under 5 years of age and in non-atopic children, the possibility of other diagnoses is high (table 52).

Table 52.

Diseases mimicking severe asthma in children.

– Bronchiolitis, bronchiolitis obliterans.– Persistent bacterial bronchitis.– Recurrent aspiration, gastroesophageal reflux, swallowing disorders.– Prematurity and related diseases (bronchopulmonary dysplasia).– Cystic fibrosis.– Endobronchial foreign body.– Congenital or acquired immunodeficiencies.– Primary ciliary dyskinesia.  – Obstruction/compression central airway.– Congenital abnormalities, including vascular rings– Tracheobronchomalacia.– Carcinoid tumor or other.– Mediastinal mass/lymphoid nodule.– Congenital heart disease.– Interstitial lung disease.– Connective tissue diseases.– Vocal cord dysfunction. 
4.5.1.8Identify the causes of por control

To this purpose, the presence of comorbidities (table 48) and/or avoidable associated factors that affect asthma control should be investigated.876 The following should be carefully evaluated: lack of adherence to treatment,878 inadequate inhalation technique,879 exposure to allergens,880 tobacco smoke, and other inhaled toxic substances881 as well as the presence of psychosocial factors.

4.5.1.9Resistance to glucocorticoids

Assessing the response to steroids after the administration of a course of OCS or a dose of triamcinolone, can help to make therapeutic decisions, such as adding tiotropium or monoclonal antibodies instead of increasing treatment with OCS.882

4.5.1.10Severe asthma phenotypes in children

Assessment of phenotypes is necessary for an adequate personalized treatment. The allergic phenotype is the most common, being frequent the presence of polysensitization, the association with other atopic comorbidities (allergic rhinitis, atopic dermatitis, food allergy) and a high T2 inflammatory profile (elevated IgE, peripheral blood eosinophilia, and increase of FENO.863,867

Non-allergic eosinophilic severe asthma is less common, and neutrophilic severe asthma is rare.

7.5.3Treatment

Children with SUA, despite adequate management of associated factors and comorbidities, are candidates for increasing the therapeutic step.

Inhaled glucocorticoids. A few children benefit from doses of fluticasone propionate or equivalent higher than 500μg/day, which in turn are related with adverse effects.879

Oral glucocorticoids. No data are available on the efficacy of OCS in the maintenance treatment of children with asthma treated with ICS at high doses plus LABA and/or montelukast. After the introduction of tiotropium and monoclonal antibodies, they have been relegated to a second step due to their adverse effects. If necessary, they should be used at the lowest dose for the shortest period of time and monitoring their adverse effects.

Triamcinolone. Triamcinolone could be useful in children with SUA, particularly in non-adherent patients to OCS or to determine the sensitivity or response to steroids.883 However, the use of triamcinolone should be very limited because of side effects and unknown pharmacokinetics.

Tiotropium. Associated with ICS/LABA in children aged 6 years or more is an option for trying to achieve asthma control884,885 prior to the use of monoclonal antibodies.

Omalizumab. It is an anti-IgE monoclonal antibody that has shown efficacy for treating children aged 6 years or more with persistent moderate or severe allergic asthma insufficiently controlled with high doses of ICS and LABA.886,887 It reduces exacerbations, symptoms, doses of ICS, the use of rescue medication, and improves quality of life.789,888

It is administered subcutaneously every 2-4 weeks, with the dose adjusted to total IgE values and body weight. In several studies carried out in routine clinical practice in children with severe allergic asthma, after the fifth month of treatment with omalizumab, improvement in asthma control, reduction in the rates of exacerbations and admissions, and in ICS doses were observed.889

Mepolizumab. It is an anti-IL5 monoclonal antibody, effective in severe eosinophilic asthma.890,891 Currently there is indication for its use after 6 years of age, with safety and efficacy data that support the use of mepolizumab in children with severe eosinophilic asthma, with a similar than adolescents and adults.829,891 The recommended dose is 40mg between 6-11 years and 100mg from 12 years, administered subcutaneously, once every 4 weeks.

Dupilumab. It is a monoclonal antibody that blocks the shared receptor of IL-4 and IL-13, reduces exacerbations, improves pulmonary function, and decreases the need for oral glucocorticoids in severe eosinophilic asthma in adults and adolescents. Its efficacy has also been proven in children from 6 to 11 years of age, with a dose of 100mg every 2 weeks of 300mg every 4 weeks in patients weighing less than 30kg, 200mg every 2 weeks or 300mg every 4 weeks in those weighing between 30 and 60 kf, and 200mg every 2 weeks in those with a body weight greater than 60 kg.892

Macrolides. They have an immunomodulator and antibacterial effect. In highly selected cases, azithromycin may be beneficial for improving some clinical symptoms and pulmonary function in children (< 6 years of age) with persistent uncontrolled asthma,893 as well as the quality of life, without differences between the eosinophilic and non-esoinophilic phenotypes.894

In infants and preschool children the level of evidence of the studies is even lower, although emerging studies are trying to define therapeutic position alternatives.

When symptoms remain uncontrolled despite ICS at high doses combined with montelukast, either LABA (off-label indication),895tiotropium,896 macrolides or even OCS may be added, although the best therapeutic option has not yet been established. The need to stepped-up treatment should be reevaluated at each visit, trying to maintain it during the shortest possible period of time.

4.6Recommendations

7.1. It is suggested to define severe uncontrolled asthma (SUA) as asthma disease that remains poorly controlled despite having been treated with a combination of ICS/LABA at high doses in the previous year, or oral glucocorticoids for at least 6 months during the same period.  R2 
7.2. The lack of control will be objectively determined by any of the following characteristics: ACT <20 or ACQ >1.5; ≥ 2 severe exacerbation or having being treated with ≥ 2 courses of oral glucocorticoids (≥ 3 days each) in the previous year; ≥ 1 hospital admission due to severe asthma in the previous year; chronic airflow limitation (FEV1/FVC ratio <0.7 or FEV1 <80% predicted) after use of adequate treatment (as long as the best FEV1 is higher than 80%).  R2 
7.3. It is recommended that diagnostic evaluation of SUA should be preferably undertaken in centers or specialized asthma units, and using a stepwise decision algorithm.  R2 
7.4. It is suggested to perform a protocolized diagnostic evaluation of SUA (in adults and children) based on three key actions: 1) to confirm the diagnosis of asthma objectively; 2) to identify those factors that are external to the asthmatic disease (treatment adherence, patient's inhalation technique, comorbidities o aggravating factors, triggers of exacerbations); and 3) to establish the phenotype of severe asthma.  R2 
7.5. In the absence of diagnostic confirmation, the presence of other possible disease mimicking asthma should be excluded.  R2 
7.6. It is recommended to establish asthma phenotype in patients with SUA as part of the diagnostic assessment. This identification can involve a differential treatment approach and have prognostic implications.  R2 
7.7. In daily clinical practice, it is suggested the use of three severe asthma phenotypes for treatment decision-making, which are the following: allergic asthma (T2), eosinophilic asthma (T2), and non-T2 asthma.  R2 
7.8. The general treatment of SUA includes: the prescription of drugs recommended in steps 5 and 6 (ICS/LABA combination at high doses and a third controller drug preferably tiotropium), adherence to an asthma education program, treatment of comorbidities/aggravating factors, and prevention/treatment of side effects of glucocorticoids.  R2 
7.9. Given that inflammation markers of phenotype T2 may be suppressed by treatment with OCS, it is recommended assessing these markers before starting treatment of OCS, or with the lowest possible dose, and at least on three occasions (e.g. during an exacerbation) prior to assuming that the patient presents a non-T2 asthma.  R2 
7.10. In the treatment of T2 SUA, on the basis of the level eosinophils in the peripheral blood and sputum, and the presence of relevant allergic clinical manifestations with confirmed sensitization to perennial aeroallergens, one or other of the available monoclonal antibodies will be chosen: omalizumab, mepolizumab, reslizumab, or benralizumab.  R1 
7.11. In case of non-T2 asthma, treatment with azithromycin or bronchial thermoplasty or systemic glucocorticoids is recommended.  R2 
7.12. In In children 6 years of age or older, depending on the inflammatory phenotype, one or the other of the monoclonal antibodies, omalizumab, mepolizumab or dupilumab, will be chosen.  R1 

8Special circumstances8.1ASTHMA and COPD overlap syndrome (ACOS)8.1.1Concept and definition

Asthma and chronic obstructive pulmonary disease (COPD) are two different chronic respiratory diseases,897 although it is common to find the characteristics of both diseases in a single patient.898

Asthma and smoking,899,900 low pulmonary function in childhood,901 exposure to irritants902 or environmental contamination903 can contribute to the development of associated COPD in adulthood.

The GesEPOC-GEMA consensus defines asthma-COPD overlap syndrome (ACOS) as the presence of persistent chronic airflow limitation (CAL) (crucial for diagnostic confirmation), in a current smoker or ex-smoker patient (main risk factor), who presents characteristics of asthma (clinical, biological or functional).904

Different definitions of ACOS have been proposed,905–912 the most recent of which are based on two types of patients:

  • Patients with asthma who smoke and develop chronic airway obstruction.

  • Patients with COPD and eosinophilia.904,911,913,914

The prevalence of ACOS varies according to the source considered and criteria used for definition,915–917 with estimates between 1.6% and 4.5% in the general population, and between 15% and 25% in patients with obstructive respiratory disease.907,918–932

Patients with ACOS have more symptoms, poorer quality of life, higher risk of exacerbations, more accelerated loss of pulmonary function, higher incidence of comorbidities and greater consumption of healthcare resources905,906,927,933–937 as compared to patients with asthma or COPD, but a better survival when treated with inhaled glucocorticoids (ICS).907,919,938,939

The mortality of chronic respiratory disease is higher in patients with ACOS or COPD than in those without chronic airway obstruction.940–942

8.1.2Diagnostic confirmation

The following sequential diagnostic evaluation is proposed (fig. 20):913,943

  • To confirm that the patient meets criteria for COPD (> 35 years, smoker >10 pack-years, post-bronchodilation forced expiratory volume in one second/forced vital capacity [FEV1/FVC] <70% [assessing the lower limit of normal, particularly at extreme ages]).909,944

  • If the patient also meets criteria for asthma,909,945 ACOS is confirmed.

Figure 20.

Diagnostic confirmation of asthma and COPD overlap syndrome (ACOS).

*Maintain after treatment with ICS/LABA (6 months). In some cases added after a course of oral steroids (15 days). ACOS: asthma-COPD overlap syndrome; ICS: inhaled glucocorticoids; LABA: long-acting ß2- adrenergic agonist; BDT: bronchodilation test; LNN: lower limit of normal.

(0.17MB).

If the patient does not meet complete criteria for asthma, the presence of a very positive bronchodilation test (FEV1 post-bronchodilation ≥ 15% and 400ml) or blood eosinophilia (≥ 300 eosinophils/μl), confirms the diagnosis of ACOS.

8.1.3Treatment

Although the initial treatment does not differ between patients with pure asthma and those with overlap syndrome, in patients with COPD, a diagnosis of ACOS predicts the response to ICS.946,947 There are proposals for the treatment of ACOS according to its treatable features948,949 that should be agreed upon.

Therapeutic recommendations in patients with ACOS

  • If the diagnostic evaluation only confirms asthma, it will be treated according to GEMA guidelines,943 avoiding monotherapy with long-acting ß2-adrenergic agonist (LABA).

  • If the diagnostic evaluation only confirms COPD, it will be treated according to GesEPOC guidelines944 avoiding monotherapy with ICS.

  • If the evaluation confirms ACOS: start with a combination of ICS at low or moderated doses according to symptoms,950 associated with a LABA.951–955

  • In case of persistence of exacerbations or relevant symptoms, it is recommended adding a long-acting muscarinic antagonist (LAMA).956,957

  • Treatment of comorbidities.

  • Treatment with biologics: the role of omalizumab958–963 or anti-leukin-5 (anti-IL-5) (benralizumab964,965 or mepolizumab963,966,967) in ACOS still remains unclear.968

  • Other treatments (when necessary): smoking cessation, respiratory rehabilitation, oxygen therapy.

  • Patients should be referred to specialized consultation in case of lack of response or partial response to the prescribed treatment.

  • Periodic follow-up assessments should be established.

8.2Asthma and pregnancy

Asthma is the most common respiratory disease in pregnancy and affects between 2% and 13% of all pregnant women.969 Up to 18% of pregnant women with asthma experienced worsening of asthma during gestation, increasing to 50% in case of severe asthma.969–971 This may be due to hormonal and mechanical changes, fear by pregnant women to use medications and the degree of previous control of the disease.972

8.2.1Effects of asthma on pregnancy

Although the risk is low, pregnant women with asthma may present maternal and fetal complications. Poor asthma control is associated with prematurity, loss of pregnancy, low birthweight, and increased perinatal mortality, whereas in the mother there is an increased risk of pre-eclampsia, placenta previa, and gestational diabetes.973,974 Also, asthma exacerbations during pregnancy are associated with a higher risk of complications during the gestational period, adverse perinatal events, and respiratory conditions during early infancy in their children.975 Prevention of asthma exacerbations is essential for reducing the risk of complications.976

Poor adherence to treatment977 and upper respiratory tract infections are the most common trigger factors for exacerbations.969 Women with other comorbidities, such as rhinitis, obesity, sudden increase of body weight during the first trimester of gestation, and smoking habit have a poorer control of asthma during pregnancy.978,979

8.2.2Treatment of asthma in pregnancy

Virtually all drugs used in the treatment of asthma cross the placental barrier; however, the advantage of treating asthma during pregnancy outweighs the potential shortcomings of the use of medication.969,972,978,979

The appropriate use of ICS, LABA, montelukast, and theophylline is not associated with an increase of fetal abnormalities.980

ICS prevent asthma exacerbations during pregnancy.981

Budesonide and other ICS are safe drugs.982,983 A study carried out in 2014 in neonates born from mothers treated with inhaled budesonide during pregnancy did not show a higher rate of teratogenesis (3.8%) as compared to the general population (3.5%).984

Although safety studies of ß2-agonists during pregnancy are not totally conclusive, and a recent study revealed a slightly higher risk for the incidence of cleft palate and gastroschisis985 but advice against the use of these compounds has not been established.986

Oral glucocorticoids (OCS) cause teratogenic effects, and their use should be restricted to asthma exacerbations and severe asthma.987 A study carried out in 250 pregnant women with asthma treated with omalizumab did not reveal a high risk of congenital malformations.988 However, starting the administration of omalizumab during pregnancy is not recommended because of the risk of anaphylaxis.988,989

The same algorithms for the treatment of exacerbations in non-pregnant women with asthma should be followed, but ensuring an adequate oxygenation (SaO2 >95%) and monitoring of the fetus.969,972

Control of asthma and prevention of exacerbations can be improved during pregnancy using measurement of FENO, questionnaires such as the Pregnancy Asthma Control Test (p-CAT) or the Asthma Control Questionnaire (ACQ) or telehealth.990–993

8.3Occupational asthma

Occupational asthma (OA) is asthma induced by work exposure and caused by agents exclusively found in the workplace (table 53). It is the most common occupational respiratory disease and the risk attributable to workplace exposure is 10% to 25%; it has been estimated that this etiology is present in one out 6 adults with asthma.996,997

Table 53.

Causative agents of occupational asthma.994,995

Class  Agent  Jobs/activities at risk of exposure 
High molecular weight     
Animals  Mites, rats, crustaceans, mammal dander, etc.  Laboratory workers, farmers, veterinarians, seafood processors 
Cereals and flours  Cereal powders, wheat, barley, oats, com  Bakery, baker's shop, pastry-making, beer industry 
Enzymes  Amylase, alcalase  Pharmaceutical companies, baker's shops 
Latex  Latex  Healthcare personnel 
Low molecular weight     
Diisocyanates  Toluene diisocyanate (TDI), methylene diisocyanate (MDI) and hexamethylene diisocyanate (HDI)  Polyurethane foams, varnish, plastics, insulators, gun spray painting 
Acid anhydrides  Phthalic acid, trimellitic acid, maleic anhydride, trimellitic anhydride  Resins and plastics, chemical and adhesive industries 
MetalsNickel, platinum, cobalt, chrome, stainless steel saltsPlatinum refinery, polishers, grinding, tanners 
Sanitary ware 
Glutaraldehyde and chlorhexidine  Carpentry, electronic welding 
Red cedar and tropical wood   
BiocidesPenicilin, spiramycin, tetracycline  Pharmaceutical industry 
Nickel, platinum, cobalt, chrome, stainless steel salts  Platinum refinery, polishers, grinding, tanners 
Antibiotics  Glutaraldehyde and chlorhexidine  Sanitary ware 
Irritants     
Bleach/hydrogen chloride  Chlorine, ammonia, CIH  Cleaning 
Smokers  Smokes  Firefighters 
Gases  NO2, SO2, ozone  Metallurgy, agriculture 
Other  Resin, acetic acid, caustic soda  Sanitary ware, chemical industry 
8.3.1Types of occupational asthma

  • Immunological OA: induced by sensitization to specific agents which are present in the workplace, through a mechanism associated with a specific immunological response.994 High molecular weight (HMW) agents (proteins or glycopeptides >10 kDa) causing production of specific IgE and the typical allergic response are the most common. Low molecular weight (LMW) agents are chemical products causing asthma through an unclear mechanisms suggesting sensitization. OA induced by HMW agents is associated with rhinitis and conjunctivitis and characterized by an earlier reaction, whereas OA induced by low molecular weight agents presents higher bronchial hyperreactivity and more severe clinical manifestations.998,999

  • Non-immunological OA: induced by irritants in the absence of sensitization.1000 The reactive airways dysfunction syndrome (RADS)1001 is the most representative form of this type of asthma. The term irritant-induced asthma is currently used, which includes cases of asthma occurring after one or more exposures to high concentration levels of irritants.1002

8.3.2Risk factors

  • Exposure level: the higher the level, the greater the risk of developing asthma caused by both HMW or LMW agents.1003,1004

  • Atopy: particularly in those exposed to HMW agents.1005

  • Rhinitis: often accompanying or preceding asthma produced by HMW.995,1006

  • Tobacco: an association may exist with the development of asthma caused by HMW and LMW agents, which act through an IgE-mediated mechanism.1007

8.3.3Diagnosis

The diagnosis of asthma and its relationship with the patient's workplace should be confirmed.1000 Diagnostic tests are shown in table 54 and the diagnostic algorithm is presented in fig. 21. Methacholine challenge test has a high negative predictive value for the diagnosis of OA due to its high sensitivity (87-95%), in particular, if the patient has been recently exposed, but the specificity is low (36-40%).1012,1013

Table 54.

Diagnostic tests in occupational asthma.

Diagnostic tests  Diagnostic value 
Clinical and work history  Essential but low positive predictive diagnostic value1008
Immunological tests  – IgE sensitization → intradermal tests/prick testIdentify the Allergen.– Positivity only indicates that sensitization exists995
PEF monitoring: working vs. non-working period  – Sensitivity: 81-87%.– Specificity: 74-89%.1009 
Non-specific bronchial hyperresponsiveness: working vs. non-working period  – Associated to PEF monitoring.– Added value but with no increase neither in sensitivity or specificity.1010 
Induced sputum  – Eosinophilic pattern in most cases (> 3%).– Improves the sensitivity of specific bronchoprovocation test.1000 
Fractional exhaled nitric oxide (FENO- Additional information to the specific bronchoprovocation test if induced sputum is not available.1000 
Specific bronchial provocation test  – Inhalation of the suspicious agent at increasing doses.– Serial FEV1 monitorization.– Is the most reliable and the reference test to confirm OA.1011 
Figure 21.

Diagnostic algorithm of occupational asthma.

OA: occupational asthma; RADS: reactive airway dysfunction syndrome; SBPT: specific bronchial provocation test; PEF: peak expiratory flow. *Measurements performed after 15 days of a working period and 15 days of sick leave; sputum: analysis of the change in the number of eosinophils.

(0.51MB).

Bronchial provocation using the specific agent is the most accepted test for the diagnostic confirmation of OA.1014

8.3.4Treatment

The patient with OA caused by a sensitizing agent should be removed from the source of exposure.1010,1015 Workers with irritant-induced asthma may continue to work provided they are transferred to lower exposure areas together with the implementation of industrial hygienic measures to reduce exposure.

In approximately 70% of patients, asthma symptoms and bronchial hyperresponsiveness (BHR) persist for several years after having been removed from the site of exposure.994

8.4Physical exercise-induced asthma

Exercised-induced asthma is defined as a narrowing of the lower airways that is triggered by strenuous physical exercise.1016

Exercise-induced bronchoconstriction is more frequent among patients diagnosed with asthma, but may be also be present in non-asthmatic subjects.1017,1018

Exercise-induced asthma is more common in patients with poorly controlled asthma.1019,1020

Exercise-induced asthma is caused by the increased osmolarity at the airway surface due to cooling and dehydration induced by hyperventilation.1021

It is associated with the release of mediators, such as prostaglandins, leukotrienes and histamine. Exercise-induced asthma may be the expression of a genetic predisposition and the environmental interaction of pollutants and the resulting oxidative stress,1022 among other factors.

The prevalence is higher in athletes, children and adolescents, females, urban environments, and among Afro-Americans and Asiatics.1023,1024

Symptoms (cough and dyspnea with wheezing) usually occur during or after finishing the exercise, with a 2-3 hour-refractory period after their onset.1025

Self-reported symptoms are unreliable for the diagnosis. The diagnostic test is the finding of a fall of FEV1 over 10% measured 30minutes after cessation of exercise and compared with the previous FEV1 value.1026

Differential diagnosis with laryngeal and glottic disorders should be made as well as with other conditions associated with exercise-induced dyspnea, such as COPD, restrictive pulmonary diseases, obesity, anatomical defects, paralysis of diaphragm, or pulmonary fibrosis.1027

It is necessary to evaluate the degree of control of asthma and to consider the possibility of increasing a therapeutic step.

Occasional use of short-acting ß2-agonists (SABA) approximately 10minutes before exercise1017 is the treatment of choice. However, when used regularly, these agents present a gradual loss of effectiveness.1028,1029

ICS should be added when a continuous treatment with SABA is needed, since this combination reduces both the frequency and intensity of exacerbations.1030

LTRA are a therapeutic option as they have a similar efficacy to LABA for preventing exercise-induced bronchial obstruction, but are not effective when the obstructions has been already established.1031

Increasingly intense warm-up exercise before starting any sports activity may decrease the intensity of bronchoconstriction.1032,1033

Reduction of dietary sodium intake and supplementation with ascorbic acid or fish oil may diminish the severity of exacerbations.1034

8.5Aspirin-exacerbated respiratory disease (AERD)

AERD or respiratory disease exacerbated by non-steroidal anti-inflammatory drugs (NSAID) refers to acute development of nasal and/or bronchial respiratory symptoms of any intensity between 30minutes and 3hours after the administration of acetylsalicylic acid (ASA) or other cyclooxygenase-1 (COX-1) inhibiting NSAIDs1013 It can be associated with cutaneous symptoms and hypotension, although this occurs rarely. The prevalence of AERD in the general population is of 0.3-2.5% but increases to 9% in subjects with asthma and is higher than 20% in patients with severe asthma.1035 In patients with concomitant asthma, chronic rhinosinusitis (CRS), and nasal polyposis (NP), the prevalence reaches 40%.1036 Avoidance of NSAID does not resolve asthma or NP.

There is a mechanism of non-IgE-mediated hypersensitivity with dysregulation of the arachidonic acid pathway by 5-LT-C4-synthase followed by overproduction of cysteinyl-leukotrienes (LT-C4, LT-D4, LT-E4) and a reduction of PG-E2.1037 There is inflammation of the mucosa with activated eosinophils and mast cells (in which the enzyme is overexpressed), basophils, and abundant platelets Blockage of COX-1 by NSAID contributes to formation and release of T lymphocytes, and to the release of preformed mediators (PGD2, histamine, and tryptase).1038 Mucous secretion, vascular permeability and bronchoconstriction are rapidly increased. IL-C2 cells of innate immune response are also involved producing type T2 cytokines.1039

8.5.1Diagnosis

AERD should be suspected in any subject with asthma, with or without CRS and NP, and confirmed through a detailed clinical history showing a relationship between ingestion of a NSAID and the appearance of respiratory symptoms.1040 At the present time, sufficiently validated in vitro diagnostic tests are lacking. The use of E4 leukotriene concentration in urine (uLTE4) together with clinical findings, slightly improves the diagnostic prediction.1041 The diagnosis is confirmed by means of controlled exposure challenge with a NSAID, preferably ASA. The administration route may be oral, bronchial (inhaled), or nasal. These latter two routes are safer, although negative results do not exclude diagnosis; in this case, the result must be confirmed by using the oral route, which is the definitive test to confirm or exclude the diagnosis of AERD.1042–1044 The diagnostic algorithm of AERD with asthma symptoms is shown in fig. 22.

Figure 22.

Diagnostic algorithm of aspirin-exacerbated respiratory disease (AERD) with asthma symptoms.954

L-ASL: lysine-acetylsalicylate.

(0.15MB).
8.5.2Treatment

The medical-surgical treatment of underlying diseases should be considered.1045 Improvement in patients with AERD and moderate or severe asthma after adding LTRAs to the standard treatment1046 or after endoscopic sinus surgery has been reported.1047 In addition, the administration of biological drugs can be useful in the treatment of patients with AERD. Omalizumab significant reduces the use of rescue medication in patients with severe allergic asthma and AERD1048 and leukotrienes (LT) in urine.1049 Also, some patients treated with omalizumab may finally tolerate NSAID, although this possibility should always be confirmed by means of controlled exposure tests.1050 Biological drugs targeting eosinophilic inflammation (mepolizumab,1051 reslizumab,1051 and benralizumab,1052 as well as dupilumab)1053 in patients with asthma and T2-high endotype may be potentially beneficial in patients with AERD.

COX-1 inhibitors should be avoided1054 (table 55). Selective COX-2 inhibitors (celecoxib, etoricoxib, perecoxib)1056 or partially selective COX-2 inhibitors (nabumetone, meloxicam)1057 are recommended, but in all cases after assessment of tolerability by oral controlled exposure testing. Doses of paracetamol higher than 500mg should not be recommended without assessment of tolerance.1040

Table 55.

Classification of some NSAIDs based on their capacity of inhibition of cyclooxygenase isoforms.1055

Potent COX-1 and COX-2 inhibitors  Acetylsalicylic acid, diclofenac, ibuprofen, metamizol 
Weak COX-1 and COX-2 inhibitors  Paracetamol 
COX-2 inhibitors   
– Partially selective (dose-dependent COX-1 inhibition)– Highly selective  Meloxicam, nabumetoneCelecoxib, etoricoxib, parecoxib 

In selected cases (patients with uncontrolled severe asthma, recurrent nasal polyposis with several endoscopic sinus surgeries despite receiving appropriate maintenance treatment), ASA desensitization could be considered.1058 It has been shown that ASA desensitization can improve nasal symptoms, asthma control, and quality of life in patients with AERD.1059,1060 Moreover, these effects are maintained over time despite requiring lower doses of ASA,1061 although the procedure is not free from associated adverse effects.1062 The maintenance dose should not be withdrawn, as the therapeutic effect is lost and adverse reactions reappear when taking NSAID.1063 However, the cost-benefit of chronic treatment with high doses of NSAID should be evaluated. While this treatment is maintained, the patient can also tolerate other NSAIDs different from ASA.1064

Both challenge and desensitization tests are not routine techniques and should be performed by qualified personnel and with the adequate equipment for the control reactions.1045

8.6Inducible laryngeal obstruction

The ERS/ELS/ACCP Working Group has defined inducible laryngeal obstruction (ILO), formerly known as vocal cord dysfunction, as a condition that causes sudden respiratory difficulties secondary to an obstruction of the airway at the level of the glottic or supraglottic larynx. These attacks are characterized by the presence of dyspnea, stridor of laryngeal origin, and other symptoms such as cough, pharyngeal globe or dysphonia.1065

The term inducible refers to the mechanism by which the obstruction crisis is triggered, which can include physical exercise or the presence of external (odors, chemicals) or internal (gastroesophageal reflux) irritants.

Its presentation may suggest an asthma exacerbation episode, as well as other laryngeal diseases such as paralysis or dystonia. Its association with asthma is possible, which makes the diagnosis difficult. ILO is observed in about 25% of individuals with asthma, with a trend towards a higher frequency in cases of severe asthma.1066

As triggers for ILO episodes, mechanical factors (talking, shouting, and swallowing) and the smell of vinegar are more frequent than exposure to pollens and humidity, which are more characteristic of asthma exacerbation.1067

Clinical suspicion is essential for the diagnosis of ILO. There are questionnaires that can help to distinguish between asthma and ILO.1068 Flattening of the inspiratory portion of the flow-volume loop is of little value in the diagnosis of ILO,1069 but it may be suggestive. The diagnosis is confirmed by laryngeal videoendoscopy showing paradoxical adduction of the larynx during inspiration, or less frequently, during expiration. A challenge test with exercise or inhalation of mannitol or methacholine is usually required.1070

The use of dynamic computerized tomography (CT) to demonstrate paradoxical laryngeal closure during attacks has been recently proposed.1066

In the acute phase of ILO, respiratory techniques for controlling inspiratory flow may be useful. Mild sedatives (ketamine, benzodiazepines) have shown to be of help, as well as inhaling heliox (a mixture of helium and oxygen) or non-invasive ventilation.1071

Long-term treatment aims to reduce the intensity and frequency of attacks. The first step includes logophoniatric rehabilitation focused on breathing techniques and relaxation of the laryngeal muscles.

In refractory cases or in patients who are not candidates for logophoniatric rehabilitation, infiltration of thyroarytenoid muscles with botulinum toxin may be used.1072

In selected cases of supraglottic ILO, transoral laser surgical techniques have been used successfully.1073

There is no solid evidence for the indication of tracheostomy in these patients; however, some single case reports have been published.1074

8.7Asthma and the coronavirus disease 2019 (COVID-19)

COVID-19 is caused by the betacoronavirus SARS-CoV-2. This airborne infection has high transmissibility and within a few weeks from the outbreak (Wuhan [Hubei, central China]) in December 19, it became a serious pandemic.1075

The disease has a broad clinical spectrum from mild forms with a few symptoms (or asymptomatic) to influenza-like symptoms (fever, cough, myalgia, asthenia) and severe forms with bilateral pulmonary infiltrates and severe acute respiratory failure (5-20%), and in some cases, causing death (2.3-3.8%).1076–1081 The disease is less common in children,1082 in which there is a different transmission pattern and only a minority of index cases (< 20%) is identified in the pediatric age.1083 Clinical manifestations are usually milder, although infants may be more vulnerable.1084–1086

The available evidence indicates that suffering from (non-severe) asthma or allergy is not associated with a greater probability of developing, becoming more severe or dying from COVID-19.1081,1087–1091 However, in severe asthma the information available is ambivalent, some series confirmed a higher morbimortality,1092,1093 or with exacerbations in the previous year,1094 while other did not.1095,1096

It is recommended not to perform pulmonary function and induced sputum tests in infected patients and in the entire population during the waves of the pandemic (peaks). However, outside of them, they can be carried out following certain biosafety standards.1097–1099

In the treatment of patients with asthma infected by SARS-CoV-2, nebulizers will not be used for the aerosolization of drugs (but devices coupled to spacers or inhalation chambers), nor non-invasive single-limb ventilator equipment and without bacterial filter placed before the outlet port.1100–1102

There is no evidence of the deleterious effect of maintenance treatments for asthma, particularly ICS, on the prognosis of COVID-19. Therefore, patients should continue to take previously prescribed medications for their asthma. Systemic glucocorticoids should even be administered in case of exacerbations.

On the other hand, some in vitro studies have shown that ICSs reduce the replication of SARS-CoV-2 in respiratory epithelial cells.1103 In addition, the data from in vivo studies seem to suggest that treatment with ICS may offer some protective capacity against infection.1104,1105

Although the information available is limited, there could be some pharmacological interaction between some of the drugs used to treat the infection and those for the treatment of asthma (table 56).1104,1105 Very close clinical monitoring is recommended when these drugs are administered together. It is possible that in some cases it would be necessary to consider up or down dose adjustments (table 56).1106

Table 56.

Possible pharmacological interactions between drugs used for the treatment of COVID-19 and those used for asthma (based on the proposals from the Neumo SEFH Group 2020).1106

Patients with severe asthma on treatment with biological drugs do not present a greater severity of SARS-CoV-2 infection.1095,1096

However, in case of infection, it is advisable to postpone the administration of the biological agent until its resolution.

There is no evidence against vaccination for COVID-19 in patients with asthma. It would be contraindicated in patients with previous anaphylaxis reactions to the vaccine or any of its components.1107

8.8Allergic bronchopulmonary aspergillosis (ABPA)8.8.1Concept and definition

ABPA is a respiratory disease due to a hypersensitivity reaction caused by bronchial colonization by Aspergillus fumigatus, which mainly affects patients with asthma and/or cystic fibrosis (CF).1108

Repeated inhalation of Aspergillus fumigatus spores generates a type I (IgE-mediated), type III (IgG-mediated immune complex), and type IV (cell-mediated) hypersensitivity response in susceptible hosts without tissue invasion.1109,1110

The Prevalence of ABPA ranges between 0.7-3.5% in asthma and 9% in CF.1111,1112

The most frequent symptoms are cough, respiratory distress, expectoration of mucus plugs, wheezing and more rarely chest pain or hemoptysis.1109,1113,1114

8.8.2Diagnostic confirmation

The diagnosis of ABPA is based on the combination of clinical, radiological, and laboratory data. The ISHAM criteria of 2013, updated in 2016, are currently the most widely used1114,1115 (table 57).

Table 57.

Diagnostic criteria ISHAM 2013, updated 2016.

Predisposing conditions 
– Asthma.– Cystic fibrosis.– COPD.– Post-tuberculous fibrocavitary disease. 
Obligatory criteria (both should be present) 
A. fumigatus-specific IgE >0.35 kUA/l (or positive cutaneous reaction if specific IgE is not available).– Elevated serum total IgE >1000 IU/ml (if patients fulfill all “other criteria”, an IgE value <1000 IU/ml can be accepted). 
Other criteria (at least two of three) 
A. fumigatus-specific serum IgG antibodies >27 mgA/l.– Total eosinophilia >500 cells/μl in steroid naïve patients.– Radiograhic lung opacities consistent with ABPA. 

More recently, Asano et al.1116 have proposed new diagnostic criteria for aspergillosis/allergic bronchopulmonary mycosis, with greater sensitivity and specificity compared to those of Patterson and Rosenberg and those of the ISHAM, even in atypical cases without asthma or without positive cultures for Aspergillus.

Total IgE serum level is a useful test both in the diagnosis and in the follow-up of ABPA. The most widely used cut-off point for diagnosis is >1,000 IU/ml1117,1118 and it decreases with treatment.1119

Most patients exhibit Aspergillus sensitization demonstrated by skin testing. Specific IgE >0.35 Ku/l has a sensitivity of 100% and a specificity of 66.2%.1114

Specific IgG against A. fumigatus is present in 69-90% of patients with ABPA. A cut-off point of 27mg/l has high sensitivity and specificity.1118

In relation to peripheral eosinophilia, a blood eosinophil count >500 cells/l has been considered to be a relevant criterion for diagnosis.1120

The most common finding on chest radiographs are infiltrates, whereas bronchiectasis, mucoid impactation, centrilobular nodules, and tree-in-bud opacities among others are frequent findings on chest CT.1121 The presence of high-attenuation mucus (HAM) visually denser than the paraspinal skeletal muscle is a pathognomonic finding of ABPA.

8.8.3Classification

On the basis of high-resolution CT findings of the lung, the disease is classified into:

  • Serological ABPA.

  • ABPA with bronchiectasis.

  • ABPA with high-attenuation mucus plugs.

  • ABPA with chronic fibrosis1122 (table 58).

    Table 58.

    Classification of ABPA based on radiological findings.

    – ABPA-S (serological ABPA): fulfills the diagnostic criteria of ABPA with an absence of radiological manifestations.– ABPA-B (ABPA with bronchiectasis).– ABPA-HAM (ABPA-High attenuation mucus): ABPA with high-attenuation mucus plugs in high-resolution CT– ABPA-CPF (ABPA-Chronic pleuropulmonary fibrosis): fulfills the diagnostic criteria of ABPA with at least two radiological features suggestive of fibrosis (including fibocavitary lesions, pulmonary fibrosis, pleural thickening) without the presence of mucoid impaction. 

Depending on the response to glucocorticoids, it is classified into 5 stages:

  • Acute.

  • Remission.

  • Exacerbation.

  • Glucocorticoid-dependent asthma.

  • Fibrotic lung disease.1115,1123

8.8.4Treatment

The aims of treatment of ABPA is to control symptoms, prevent exacerbations, and limit loss of pulmonary function and structural damage especially bronchiectasis.

A reduction of IgE levels of 25-50% correlates with clinical and radiological improvement.1119

Systemic glucocorticoids are the primary therapy for ABPA. The steroids help to relieve the symptoms and decrease airflow obstruction, decrease serum IgE and reduce peripheral blood eosinophils.1108

Prednisolone is a commonly used drug for treatment, 0.5 to 1mg/kg/day for 2 weeks, followed by 0.5mg/kg every other day for 8 weeks. A subsequent taper (by 5mg every 2 weeks) over the 3 to 5 months1124 or 0.75mg/kg/day for 6 weeks followed by 0.5mg/kg/day for 6 weeks, and subsequent taper by 5mg every 6 weeks and suppression in 8-10 months.1113,1125,1126

It is advisable to add antifungal treatment with itraconazole (200mg twice daily for 26 weeks) or voriconazole to decrease the fungal load and subsequent suppression of inflammatory response.1127,1128

There are no randomized double-blind placebo-controlled trials to establish the efficacy of biologics in the treatment of ABPA. However, the involvement of type I (IgE-mediated) hypersensitivity response and Th2 pathway suggest that they could have a prominent role.

Treatment with omalizumab has been associated with an improvement of asthma symptoms, reduction of FENO levels, exacerbations, serum IgE, and requirements of the doses of glucocorticoids.1129

Single cases reports of the combination of omalizumab and mepolizumab have been published.1130

The evidence with mepolizumab1131 and benralizumab1132,1133 is limited.

Some cases have been published with sequential treatments due to a lack of response to the initial biological treatment. After failure of omalizumab, there have been cases of response to mepolizumab1134 or benralizumab;1135 or initial failure of mepolizumab with posterior response to benralizumab.1136

8.9Eosinophilic granulomatosis with polyangiitis (EGPA)8.9.1Concept and definition

EGPA (formerly known as Churg-Strauss syndrome) is a rare form of vasculitis and according to the Chapel Hill Consensus Conference (CHCC 2012) included in the group of vasculitis associated with antineutrophil cytoplasmic antibodies (ANCA).

It is characterized by eosinophil-rich necrotizing granulomatous inflammation and necrotizing vasculitis of small and medium-sized blood vessels associated with eosinophilia and asthma.1137

EGPA has been recently considered an emergent clinical variant of ANCA-associated vasculitis, in which a Th2 autoimmune response is involved in its pathogenesis.1138

In general, ANCA are positive in 30-47% of patients with EGPA and are usually p-ANCA (MPO-ANCA). Rarely, c-ANCA are seen, which difficults the differential diagnosis with granulomatosis with polyangiitis (formerly known as Wegener's disease).1139

The incidence of EGPA is lower than 4 cases per million person-years and the prevalence is lower than 31 cases per million person-years.1140

It is estimated that the age at onset ranges between 40-59 years and at the time of diagnosis, history of asthma of more than5 years is present in more than 90% of patients.1141 Some studies have shown a higher prevalence in women than in men.1142

Clinical manifestations of EGPA are heterogeneous and the clinical course is characterized by three phases: prodomal (asthma, rhinosinusitis, and nasal polyposis), eosinophilic (peripheral eosinophilia and organ involvement), and vasculitis (clinical manifestations of vasculitis of small-sized vessels). These three phases are not necessarily always present, they may even overlap, or be absent in some patients.1143,1144

Asthma in EGPA is usually severe in more than 65% of patients, glucocorticoid-dependent, and precedes to the systemic disease.1140

The presence of severe refractory and/or late-onset asthma associated with eosinophilia >10% or pulmonary infiltrates requires ruling out EGPA.

8.9.2Diagnostic confirmation

Although biopsy (lung, nerve or skin) is the reference procedure for the diagnosis of EGPA, most patients are diagnosed according to clinical criteria.1145

The diagnosis of EGPA is based on the combination of clinical, radiological, and laboratory criteria. The presence of at least four out of the six criteria proposed in the classification of the American College of Rheumatology (ACR) in 19901146 (table 59) renders an 85% sensitivity and 99.7% specificity. This classification does not include the determination of ANCA, which can lead to overlaps with other vasculitis conditions.

Table 59.

Diagnostic criteria of EGPA according to the American College of Rheumatology 1990.1146

The presence of at least four out of the six criteria: 
1. Asthma.2. Eosinophilia (> 10% total leukocyte count)3. Neuropathy (mono- or polyneuropathy)4. Migratory infiltrates in lungs.5. Paranasal sinus abnormalities.6. Extravascular eosinophilic infiltration in biopsy. Histological features of vasculitis. 

Recently, the ACR/European Alliance of Associations for Rheumatology (EULAR) validated criteria to help differentiating EGPA for other small- or medium-vessel vasculitis once other mimicking diseases have been excluded (table 60). A score of >6 points allows classifying vasculitis as EGPA with a sensitivity of 85% (95% CI: 77 to 91) and a specificity of 99% (95% CI: 98 to 100).

Table 60.

Classification criteria of EGPA according to the American College of Rheumatology/European Alliance of Associations for Rheumatology 2022.1147

A cumulative score ≥ 6 points is necessary to classify EGPA as a small- or medium-vessel vasculitis*
Clinical criteria   
• Obstructive airway disease  +
• Nasal polyps  +
• Mononeuritis multiplex  +
Laboratory and biopsy criteria   
• Blood eosinophil count >1×109/liter  +
• Biopsy with extravascular eosinophilic predominant inflammation  +
• Cytoplasmic antineutrophil cytoplasmic antibody(c-ANCA) or anti–proteinase 3 (anti-PR3) positivity  -3 
• Hematuria  -1 
*

These criteria should be applied for classifying a patient with EGPA when the diagnosis of small- or medium-vessel vasculitis has been confirmed.

In an appropriate clinical context, the high diagnostic specificity of a positive ELISA test for MPO-ANCA or PR3-ANCA can obviate the need for biopsy.

Patients with positive ANCA (usually MPO-ANCA) typically have more renal and peripheral nerve involvement, whereas cardiomyopathy and possibly pulmonary infiltrates are more common in ANCA-negative cases.

Multiple prospective and retrospective studies have confirmed cardiac involvement as one of the most important risk factor for specific mortality in patients with EGPA (standardized mortality rate 3.06).1144,1146

The 2021 ACR and the Vasculitis Foundation guideline defined EGPA as active disease in the presence of new, persistent clinical signs and/or symptoms and not related to prior damage; severe disease in the presence of vasculitis with life- or organ-threatening manifestations (e.g. alveolar hemorrhage, glomerulonephritis, central nervous system vasculitis, mononeuritis multiplex, cardiac involvement, mesenteric ischemia, limb/digit ischemia); and non-severe disease in the presence of vasculitis without life- or organ-threatening manifestations (e.g., rhinosinusitis, asthma, mild systemic symptoms, uncomplicated cutaneous disease, mild inflammatory arthritis).1148

The update of the EULAR recommendations for the management of ANCA-associated vasculitis has recently been published, which classifies the disease for treatment purposes according to the presence of vital organ involvement or not (instead of severity).1149

8.9.3Treatment

When deciding on treatment, it is important to consider the severity and whether EGPA is active or in remission.

The treatment is divided into two parts: 1) induction phase to achieve remission, and 2) maintenance phase.

Recently, the 2021 ACR/VF guideline has established new recommendations, the majority of which are conditional, for the treatment of EGPA (fig. 23), in particular:

  • a.

    Induction of remission for active and severe EGPA treatment with doses of oral glucocorticoids (1mg/kg/day) or intravenous pulse (e.g. methylprednisolone i.v. 500mg/day for 3-5 days followed by oral glucocorticoids with progressive reduction up to 10mg or less), generally associated with cyclophosphamide (2mg/kg/day orally or 15mg/kg i.v. every 2 weeks for 3 doses, followed by 15mg/kg every 3 weeks for 3 doses).1148–1151

    Experiences with rituximab have been published for refractory forms of EGPA,1152,1153 but controlled studies are lacking. It has been used in EGPA with renal involvement, but its efficacy is less established compared to other ANCA-associated vasculitis (especially if ANCA-negative), with cases of recurrent nasal and sinus disease and refractory asthma despite treatment with rituximab. The 2021 ACR/VF guideline consider rituximab in the following situations: ANCA-positive patients, glomerulonephritis, failure of cyclophosphamide treatment, or to preserve fertility.1148

  • b.

    For patients with active non-severe EGPA, induction of remission treatment includes the association of glucocorticoids (prednisone 0.5-1mg/kg/day with progressive reductuion)1145 with mepolizumab, and the association of azathioprine, mycophenolate or methotrexate as a second choice.

  • c.

    In patients with severe EGPA who achieve remission with cyclophosphamide (generally 3-6 months), the maintenance treatment recommended includes oral glucocorticoids at doses of 10mg or less, associated with azathioprine (2mg/kg/day), methotrexate (7.5mg/week oral or subcutaneous with 2.5mg increases every week up to a maximum dose of 20-25mg/week accompanied by folic acid 1mg/day), or mycophenolate (experience in case series only) for 12-18 months or at long-term in case of frequent relapses.999,1011,1043,1103,1105

  • d.

    In patients with non-severe EGPA with relapse on treatment with azathioprine, methotrexate or mycophenolate, the use of mepolizumab is recommended.

Figure 23.

Treatment of EGPA.

OCS: oral glucocorticoids; RTX: rituximab; AZA: azathioprine; MTX: methotrexate; MMF: mycophenolate mofetil. *Severe: alveolar hemorrhage, glomerulonephritis (GNF), central nervous system vasculitis, mononeuritis multiplex, cardiac involvement, mesenteric ischemia, limb/digit ischemia. **Non-severe: rhinosinusitis, asthma, mild systemic symptoms, uncomplicated cutaneous disease, mild inflammatory arthritis. RTX in cases of ANCA+, GNF, failue of cyclophosphamide, and to preseve fertility.

In selected cases consider OCS only. Modified from the 2021 ACR/VF guideline.1148

(0.43MB).

Regarding mepolizumab both the FDA and the EMA have approved its use at doses of 300mg subcutaneously every 4 weeks as add-on treatment in patients with EGPA aged 6 years and older with relapsing-remitting or refractory disease.1154 In a phase III 52-week randomized placebo-controlled trial in patients with relapsing or refractory EGPA, mepolizumab 300mg subcutaneously added to standard treatment increased the percentage of patients who achieved remission, improved their quality of life, and decreased the doses of oral glucocorticoids as compared with placebo.1155

Severe uncontrolled asthma may persiste even after conventional treatment of EGPA, being a reason for the indication of mepolizumab.

8.10Idiopathic hypereosinophilic syndrome8.10.1Concept and definition

Idiopathic hypereosinophilic syndrome (HES) is a rare disease characterized by bone marrow overproduction of eosinophils resulting in persistent high blood eosinophil levels (≥ 1500 cells/μl) with involvement of tissues and organs, without a known underlying cause.1156,1157

The diagnosis of idiopathic HES requires excluding other secondary causes of hypereosinophilia (parasitosis, drugs, entities such as EGPA, ABPA, eosinophilic pneumonia, etc.) including myeloid/lymphoid neoplasms with eosinophilia associated with FIP1L1-PDGFRA or PDGFRA, PDGFRB, FGFR1, PCM-JAK2 mutations.1158,1159

The incidence of all types of HES is estimated between 0.04-0.17 per 100,000 person-years and the prevalence is 0.15-6.3.1159,1160

In patients with chronic or persistent tissue infiltration, the release of effector molecules by activated eosinophils can result in tissue damage and organ dysfunction.1161

The clinical presentation is variable, the most frequent symptoms are cutaneous (intractable pruritus, urticaria, angioedema), pulmonary (asthma, sinusitis, pulmonary infiltrates), gastrointestinal, cardiovascular (endomyocardial fibrosis, thromboembolism), and neurological.1157,1162–1164

8.10.2Diagnostic confirmation

The diagnosis of idiopathic HES is based on the involvement and/or dysfunction of a systemic organ, directly related to an eosinophil count ≥ 1,500/μl in two or more determinations separated by 4 weeks and/or tissue eosinophilia without a secondary cause.1161

8.10.3Treatment

The goal of treatment is to achieve sustained reduction in blood and tissue eosinophil counts to reverse and prevent damage and improve symptoms.1158

Treatment of idiopathic HES includes systemic glucocorticoids and cytotoxic and/or immunosuppressive drugs. However, they can fail to achieve complete remission of the disease and have important side effects.1165,1166

Patients receiving standard treatment for HES frequently have periods of poor disease control, with worsening symptoms and increased eosinophil counts, sometimes with organ involvement.1159

Previous studies with mepolizumab (administered at a dose of 750mg i.v. every 4 or more weeks) demonstrated a reduction in the dose of glucocorticoids and eosinophil count, with good tolerance.1167–1171

A phase III randomized, double-blind, placebo-controlled study for 32 weeks demonstrated that mepolizumab (300mg subcutaneously every 4 weeks), added to existing HES therapy, reduced disease flares in patients with uncontrolled FP1L1-PDGFRA negative disease (defined as ≥ 2 flares within the past 12 months and a blood eosinophil count ≥ 1000 cells/μL) compared with placebo. The proportion of patients who experienced one or more flares during the study was 50% lower in the mepolizumab group than in the placebo group (15 of 54 [28%] vs. 30 of 54 [56%]; p=0.002); and the relapse rate was 66% lower with mepolizumab compared with placebo (0.50 vs 1.46 flares per year, respectively; p<0.001). A 92% reduction in eosinophil count was also observed.1172

An open-label extension study in patients with FIP1L1-PDGFRA negative HES who had been previously treated with placebo or mepolizumab in the phase III trial and received mepolizumab 300mg subcutaneously every 4 weeks for 20 weeks, confirmed the reduction of flares, need of OCS, and blood eosinophil count also in the long-term.1173

4.7Recommendations

8.1. The diagnosis of ACOS will be establish in patients with persistent chronic airflow limitation, current smokers or ex-smokers, with documented diagnosis of asthma, or in whom there is a very positive bronchodilation test or eosinophilia.  R2 
8.2. All patients with ACOS will be initially treated with a combination of ICS and LABA.  R2 
8.3. In patients with ACOS treated with a combination of ICS and LABA who remain symptomatic or with exacerbations, a LAMA will be added.  R2 
8.4. E Drugs usually administered, LABA plus ICS, are recommended for the maintenance treatment of asthma in pregnant women.  R1 
8.5. In the treatment of exacerbations in pregnant women the same algorithms than in non-pregnant women should be followed, ensuring adequate oxygenation (SaO2 >95%) and monitoring of the fetus.  R1 
8.6. In order to reduce the risk of maternal and fetal complications, pregnant women with asthma should be adequately controlled for preventing severe exacerbations.  R1 
8.7. In adult-onset asthma or if there is a deterioration of previous asthma, it is recommended to exclude occupational asthma.  R2 
8.8. The diagnosis of occupational asthma should be confirmed by objective tests, and in cases of allergic etiopathogenesis, by immunological tests.  R2 
8.9. The specific challenge test is the reference diagnostic test for immunological occupational asthma.  R2 
8.10. In the treatment of immunological occupational asthma, removal of exposure to the causative agent is recommended.  R2 
8.11. In exercise-induced asthma, warm-up exercises before starting the sport activity are recommended.  R1 
8.12. In exercise-induced asthma, SABA used occasionally are the most effective short-term treatment option.  R1 
8.13. In exercise-induced asthma, ICS reduce the frequency and intensity of symptoms, so that its use is advisable in patients usually treated with SABA.  R1 
8.14. In exercise-induced asthma, LTRA is a less effective therapeutic option than ICS for preventing bronchoconstriction and are not useful to reverse an already established obstruction.  R1 
8.15. It is recommended to evaluate the degree of control to determine the need for increasing a therapeutic step in known asthma patients with exercise-induced asthma.  R1 
8.16. In patients with asthma and chronic rhinosinusitis with nasal polyps, it is advisable to exclude aspirin-exacerbated respiratory disease (AERD), particularly in cases of severe asthma.  R1 
8.17. Patients with AERD should avoid receiving treatment any NSAIDs that are COX-1 inhibitors.  R1 
8.18. In the analgesic or anti-inflammatory treatment of patients with AERD, an alternative medication of choice (opiates, systemic corticosteroids) should be used. After demonstrating their tolerability, paracetamol at doses lower than 500mg and selective COX-2 inhibitors (celecoxib, etoricoxib, parecoxib) can be used.  R2 
8.19. In patients with moderate or severe asthma and AERD, adding LTRA should be considered.  R2 
8.20. Desensitization with acetylsalicylic acid may be useful in selected cases.  R2 
8.21. Biological drugs can be used in patients with severe uncontrolled asthma and AERD, especially in the presence of concomitant nasal polyposis.  R2 
8.22. The diagnosis of inducible laryngeal obstruction (ILO), formerly known as vocal cord dysfunction, should be established after clinical suspicion and confirmation by laryngeal videoendoscopy.  R1 
8.23. Treatment of the acute phase of ILO should include respiratory logophoniatric reeducation (laryngeal muscle relaxation) techniques.  R2 
8.24. In the treatment of the acute phase of ILO, sedatives may be useful, whereas type A botulinum toxin or surgery is reserved for refractory cases.  R2 
8.25. It is recommended to rule out ABPA in all patients with severe uncontrolled asthma.  R1 
8.26. For the diagnosis of ABPA in patients with asthma, it is recommended to use a combination of clinical, laboratory, and radiological data following the ISHAM 2016 criteria.  R1 
8.27. In acute ABPA it is recommended to start treatment with glucocorticoids generally associated with antifungals.  R1 
8.28. In case of recurrent exacerbations of ABPA or glucocorticoid dependence, a therapeutic trial with biological drugs is recommended.  R2 
8.29. It is recommended that EGPA should be ruled out in all patients with long-lasting severe uncontrolled eosinophilic asthma.  R2 
8.30. For the diagnosis of EGPA in patients with asthma, it is recommended to use a combination of clinical, laboratory (eosinophil count and ANCA determination in blood) and radiological data.  R1 
8.31. In severe and active EGAP, it is recommended to start treatment with systemic glucocorticoids in pulses or at high oral doses associated with cyclophosphamide or rituximabR2 
8.32. In non-severe and active EGAP, it is recommended to start treatment with glucocorticoids associated with mepolizumab as the first choice.  R2 
8.33. It is recommended to rule out secondary causes of hypereosinophilia and myeloproliferative hematological disease before diagnosing idiopathic HES.  R1 
8.34. The diagnosis of idiopathic HES is established (by arbitrary expert criteria) in the presence of involvement and/or dysfunction of a systemic organ, together with an eosinophil count ≥ 1,500/μl in 2 or more determinations separated by 4 weeks, and/or tissue eosinophilia of unknown cause.  R2 
8.35. The first-line treatment of idiopathic HES is glucocorticoids to which immunosuppressants or other glucocorticoid-sparing drugs can be added, but in some patients their effect may be insufficient to reduce eosinophilia.  R1 
8.36. It is recommended to add mepolizumab to glucocorticoid treatment in idiopathic HES with the aim of reducing disease flares, the need for OCS, and the eosinophil count.  R1 

9Organizational aspects. GEMA diffusion9.1Continuity of care

Healthcare professionals should provide asthma patients with continuous care in order to ensure adequate prevention, diagnosis, control, treatment and follow-up,1174 so that coherence of coordinated healthcare over time (continuity of care)1175 is perceived by the users.

It is a priority to identify the current status of healthcare for patients with asthma1176–1181 to provide solutions in the three types of continuity of care: information (availability of data of previous episodes at different levels of care), relationship (between patients and providers), and management (coordination of actions).1182

The multidisciplinary approach, the coordination between the levels of care, the patient's involvement, and appropriate management of social and healthcare resources are the essential elements to establish an integrated care network that provides quality care to patients with asthma.1183–1185 The involvement of nursing, as demonstrated by the Finish program, is essential to achieve good asthma control.1186 Also, the collaborative practice between physicians and community pharmacists has a positive impact on the patients’ health, improving the knowledge they have of their disease, their quality of life, adherence to treatment, and control of the disease.1187,1188

Actions to be implemented for improving continuity of care in asthma are shown in table 61.

Table 61.

Actions aimed to improve continuity of care in asthma.

Healthcare professionals  Patients  Administration 
GEMA implementation1180,1189  Education1190,1191  National Strategic Plan in Asthma (nonexistent) 
Coordination between healthcare levels1192,1193  Adherence to treatment1194,1195  Integrated healthcare processes1196 
Referral criteria established by consensus1193  Action plans1189,1191  Universal electronic medical history1197 
Asthma units1198  Self-control1199–1201  National registry of patients with severe asthma1202,1203 
Importance of Nursing1186 and Community Pharmacy1188 in healthcare programs    Strategic plans adapted to local characteristics1183 
Use of computerized tools for asthma control1204,1205    Provide necessary resources 

Referral to specialized care has shown to be effective for adequate management of patients with asthma in selected cases.1206–1209

Clinical practice guidelines should describe the criteria by which a patient with asthma should be referred to an asthma specialist, but an effective referral system requires good coordination between healthcare providers at the different levels of care.1209

In Spain, the consensus document on referral criteria for asthma,1193,1210 developed by professionals of Primary Care Medicine, Pneumology and Allergology, establishes the circuit to be followed by the primary care physician in the event of suspected asthma, in the evaluation of the control and follow-up of asthma patients, as well the referral of patients with asthma from primary care to specialized care in the following circumstances:

  • To confirm the diagnosis of asthma when this is not possible with the resources available in the primary care setting.

  • To study comorbidities when this cannot be completed in the primary care setting.

  • Patients with severe asthma and uncontrolled asthma.

  • Special circumstances (allergy study, occupational asthma, aspirin-exacerbated respiratory disease [AERD], exercise-induced asthma, and asthma in pregnancy).

  • To study other diseases for the differential diagnosis with asthma.

For an adequate bidirectional communication between both levels of care and to improve continuity of care, the document proposes specific electronic referral templates and a minimum data set that should be included in specialized care reports of asthma patients.1193

9.2Asthma unit

Prospective data from a UK registry showed that management of patients with difficult asthma in asthma centers specialized in severe asthma resulted in improved related quality of life and less use of healthcare resources.1210 Some authors indicate that 1-day visit with extensive assessment in a specialized asthma center is beneficial and sufficient for a large group of patients with uncontrolled asthma, reducing the need of high-cost special treatments.1211

En 2015, the asthma area of the Spanish Society of Pneumology and Thoracic Surgery (SEPAR) addressed the task of establishing the necessary requirements for the provision of official accreditation standards of the different levels of care for asthma units already existing in hospitals of the Spanish National Healthcare System. Accreditation levels included basic units, specialized units, or specialized units of high complexity, with or without the distinctive of excellence, according to the fulfillment of a series of criteria.1212 Also, recently the Spanish Society of Allergology and Clinical Immunology (SEAIC) have established criteria for accreditation of Severe Asthma Units (SAU) in the Allergology Services.1213

These units coordinate the strategies aimed at improving the follow-up of patients with asthma, particularly those with severe asthma, interacting with other levels of care and with all other specialists involved in care of asthma, as well as the use of complex diagnostic and therapeutic techniques that require rigorous knowledge and application. This strategy results in a personalized clinical approach that makes it possible to recognize individual needs and carry out special pharmacological or behavioral interventions (education, follow-up of adherence to treatment).1214

Given the complexity of asthma, different specialties (Otorhinolaryngology, Gastroenterology, Endocrinology, Psychology, Pharmacy, etc.) are involved to a greater or lesser extent in the care of asthma patients. It is indispensable to have available a specialized nurse who can perform all education tasks, including training and review of the inhalation technique, treatment adherence, self-management, written action plan, and knowledge of the disease.1215

The distribution of tasks that should be assumed by the Asthma Unit is shown in fig. 24.

Figure 24.

Working tasks and distribution of activities in a specialized Asthma Unit in the hospital.

(0.48MB).

The development of an Asthma Unit in a healthcare areas is associated with important clinical benefits for the patient (increases considerably the percentage of patients with well-controlled asthma and reduces exacerbations substantially), with a highly favorable cost-effectiveness balance. In this respect, implementation of an Asthma Units is a beneficial option both from the perspective of efficiency for the healthcare system, and from the perspective of the patient, improving health outcomes and quality of care.1198,1216

9.3Implementation of GEMA

For a clinical practice guideline (CPG) to be applied and adopted by healthcare professionals, three indispensable sequential key steps should be addressed: diffusion, implementation, and evaluation. The diffusion of a CPG (be means of medical and scientific publications, mailing, workshops, symposia and computer-based tools via Internet) will not be effective if is not accompanied by a proper implementation.1217–1219

However, CPGs for asthma do not seem to meet this requirement. A study that aimed to evaluate the quality of these CPGs using the AGREE II instrument, found that none of them reached a score higher than 60% (minimum recommended level) in the evaluation of their corresponding implementation plans (domain 5 of AGREE checklist: applicability or implementation).1220

The correct application and implementation of a CPG, Graham proposes a series of structured and stepwise planning in order to transfer knowledge into action (knowledge-to-action).1221 The diffusion and implementation of GEMA is based in part on such principles and includes the following 8 actions:

  • 1.

    Specific healthcare area. For the implementation, a specific healthcare territorial area will be defined in order to assign a selected zone to a reference hospital and the various primary care teams assigned to the hospital.

  • 2.

    Analysis of needs and local deficiencies. An audit will be performed in order to detect weak points and deficiencies in disease management within that territory.

  • 3.

    Executive committee. A multidisciplinary group of experts in asthma pertaining to the implementation area will be set up. The committee will comprise expert physicians in asthma (pneumologists, allergologists, primary care physicians, and pediatricians) as well as professional representatives from the local nursing and pharmacy settings of the area.

  • 4.

    Development of a functional document based on GEMA.5.3 The Executive Committee will adapt evidences and recommendations of GEMA5.3 to the local healthcare reality according to the resources assigned to the area, the type of professionals, and their training level.

  • 5.

    Material resources. A minimal amount of material resources should be available in the area in order to ensure the application of the guideline. Specific resources will include: spirometries (of good quality throughout the area) in all centers; electronic medical history (EMH) shared among healthcare levels; standardized asthma symptom questionnaires (ACT, ACQ); placebo-containing inhalation devices to be used in education programs to instruct patients in the inhalation technique; an accredited specialized Asthma Unit in the hospital fitted with a complete technical equipment (bronchoprovocation tests, FENO, allergy skin tests, computerized tomography [CT]).

  • 6.

    Training plan. An educational intervention on asthma will be performed among both medical and nursing professionals working in the area.

  • 7.

    Professional motivation plan. Administrative authorities will be engaged in promoting adherence of professionals involved in the “Implementation Plan” by setting up appropriate motivational interventions.

  • 8.

    Evaluation and follow-up plan. To determine the impact of the “Implementation Plan” a set of health-related results (health outcomes) will be used in order to determine whether the proposed objectives have been achieved, and to establish appropriate adjustments if objectives were not meet. The eight indicators of healthcare quality for asthma proposed by a multidisciplinary expert group that participated in GEMA are shown in table 62.

    Table 62.

    Healthcare quality indicators for asthma proposed by the multidisciplinary expert group (Asmaforum II).

    Indicator groups  Indicator  Calculationo 
    I. Diagnosis1. C Diagnostic confirmation by means of spirometry with bronchodilation test. The diagnostic confirmation of patients with asthma is established by spirometry and bronchodilation test as an objective measurement of functional involvement.  Number of patents with asthma undergoing spirometry × 100/number of patients diagnosed with asthma. 
    2. Sensitization study in allergic asthma. Patients with suspicion of allergic asthma should undergo a study of possible sensitization to different allergens.  Number of patients diagnosed with suggestive history of allergic asthma with sensitization study performed at different allergens × 100/number of patients diagnosed with asthma. 
    II. Non-pharmacological treatment3. Smoking cessation. Smoking cessation is recommended in smokers with asthma.  Number of smoking patients with asthma and registered recommendation to quit smoking × 100/smoking patients with asthma. 
    4. Education plan for patients with asthma. Patients with asthma should follow a basic education program (including knowledge of the disease and its treatment, written action, plan and inhalation technique) as part of their management.  Number of patients with asthma with an asthma education program × 100/number of patients with asthma. 
    III. Pharmacological treatment5. Treatment of choice in persistent asthma. The treatment of choice in persistent asthma includes the use of inhaled glucocorticoids (ICS) on a daily basis. In some cases, an alternative treatment with leukotriene receptor antagonists may be justified.  Number of patients on control treatment due to persistent asthma receiving ICS × 100/number of patients on control treatment due to persistent asthma. 
    6. Treatment of asthma in the pregnant woman. In the maintenance treatment of asthma in pregnancy, it is recommended to maintain the usually administered medication (β2-adrenergic agonists and inhaled glucocorticoids).  Number of women with asthma who maintain their usual treatment (β2-adrenergic agonists and inhaled glucocorticoids) during pregnancy×100/number of pregnant women with asthma on maintenance therapy. 
    IV. Follow-up7. Periodic follow-up of patients. Need to establish periodic follow-up of patients based on scheduled medical visits, even in the absence of exacerbations.  Number of scheduled follow-up visits (unexpected visits excluded) per patient per year × 100/number of patients with asthma on follow-up by year. 
    8. Periodic registry of exacerbations. Specific assessment of exacerbations is periodically evaluated.  Number of patients with asthma in whom exacerbations have been evaluated and documented × 100/number of patients with asthma. 

9.4Telemedicine and asthma

Advances in knowledge and information technology make it possible to provide medical care for chronic conditions such as asthma. The terminology used to define healthcare based on the new technologies is continually evolving. It has been proposed to use the term telehealthcare as a general term, encompassing all the different forms of telemedicine-related healthcare. This term includes.1222

  • Tele-monitoring that involves storing and transference of patient's data.

  • Tele-consultation is the use of technology allowing remote consultation between a patient and a clinician.

  • Telemedicine that involves consultation among healthcare professionals.

The technology used is based on 3 main strategies:1223

  • Support for patients’ self-management through the use of automatic medication-taking reminders (tele-reminder) to improve adherence, educational games to improve knowledge or modify the attitude towards the disease, and tele-monitoring of clinical variables (PEF, use of medication, etc.).

  • Remote consultation with a healthcare professional.

  • Computerized systems to aid decision-making for both physicians and patients.

The combined use of these strategies, which includes tele-case management or tele-consultation, improves the control of the disease and the quality of life of patients with asthma.1223,1224

4.8Recommendations

9.1. To achieve quality in continuing care of asthma, coordination of different healthcare levels, involvement of the patient and nursing professionals, as well as the rational use of resources are recommended.  R1 
9.2. It is suggested to promote the development of Asthma Units because they provide a better control of the disease, decreasing exacerbations, and improving health-related quality of life of patients, with a favorable cost-effectiveness balance.  R2 
9.3. It is recommended to include a diffusion and implementation plan of this guideline to achieve the objectives of improving the level of training of healthcare professionals.  R2 
9.4. The GEMA implementation plan proposes: implementation of actions in a local specific healthcare area; identification of local opinion leaders and engage them in this endeavor; adaptation of GEMA to the healthcare reality of the area; arrangement of an education plan for the professionals involved; and adjustment of actions according to whether objectives assessed by health outcomes have been attained.  R2 
9.5. The use of telemedicine or medical tele-assistance based on strategies of “tele-cases” or tele-consultation is proposed, given that it improves control of disease and quality of life of patients with asthma.  R2 

Authors’ contributions

All the coauthor's contributed to the search for evidence, participated in the debates for decision-making and in the critical review of the final document.

Funding

The GEMA5.3 project was funded by pharmaceutical companies ALK, AstraZeneca, Chiesi, GlaxoSmithKline, Menarini and Sanofi. The viewpoints of these funding bodies did not influence the content of the guide.

Conflicts of interest

The authors of this guide declare that in the past two years, they have received honoraria for their participation in meetings, congresses, or research projects organized by the following pharmaceutical companies: ALK, AstraZeneca, Bial, Boehringer-Ingelheim, Chiesi, Esteve, GlaxoSmithKline, Leti, Menarini, MSD, Mundipharma, Novartis, Orion, Pfizer, Sanofi, Teva, and Zambón.

Acknowledgments

The authors would like to thank Marta Pulido, MD, for the English translation of the guideline and Luzan5 for their contribution in its diffusion.List of abbreviations:ABPA

Allergic bronchopulmonary aspergillosis

ABPA-B

ABPA with bronchiectasis

ABPA-CPF

ABPA-Chronic pleuropulmonary fibrosis

ABPA-S

Serological ABPA

ABPA-HAM

ABPA-High attenuation mucus

Ac

Antibody

ACE

Angiotensin-converting enzyme inhibitors

ACOS

Asthma-COPD overlap syndrome

ACQ

Asthma Control Questionnaire

ACR

American College of Rheumatology

ACT

Asthma Control Test

AE

Adverse effects

AEMPS

Spanish Agency of Medicines and Sanitary Products

AEPap

Spanish Association of Primary Care Pediatrics

AERD

Aspirin (acetylsalicylic acid)-exacerbated respiratory disease

AIRQ

Asthma Impairment and Risk Questionnaire

ALAT

Latin American Thoracic Association

ANCA

Antineutrophil cytoplasmic antibodies

API

Asthma Predictive Index

AR

Allergic rhinitis

ASA

Acetylsalicylic acid

ATS

American Thoracic Society

AZA

Azathioprine

BAI

Breath-actuated inhaler

Bd

Bronchodilation

BDT

Bronchodilation test

BHR

Bronchial hyperresponsiveness

BMD

Bone mineral density

BMI

Body mass index

c-ACT

Childhood Asthma Control Test

CAN

Asthma Control Questionnaire in Children

CAL

Chronic airflow limitation

CF

Cystic fibrosis

CO

Carbon monoxide

COPD

Chronic obstructive pulmonary disease

COVID-19

Coronavirus disease 2019

COX-1

Cyclooxigenase-1

COX-2

Cyclooxigenase-2

CPAP

Continuous positive airway pressure

CPG

Clinical practice guideline

CPK

Creatine phosphokinase

GRAP

Primary Care Respiratory Society

CRS

Chronic rhinosinusitis

CRSsNP

Chronic rhinosinusitis without nasal polyps

CRSwNP

Chronic rhinosinusitis with nasal polyps

CT

Computed tomography

DAC

Difficult-to-control asthma

DPI

Dry powder inhaler

EGPA

Eosinophilic granulomatosis with eosinophilia

EMA

European Medicines Agency

EMH

Electronic medical history

ENT

Ear, nose and throat

Eos

Eosinophils

ERS

European Respiratory Society

ERS/ATS

European Respiratory Society/American Thoracic Society

FDA

Food and Drug Administration

FEF25-75

Forced expiratory flow between 25% and 75% of forced vital capacity (FVC)

FENAER

National Federation of Associations of Patents with Allergic and Respiratory Diseases

FENO

Fractional exhaled nitric oxide

FEV1

Forced expiratory volume in one second

FEOS

Weighted FEV1. Exacerbations, oral corticosteroids and asthma symptoms to determine the response to a biological drug

FIO

Forced impulse oscillometry

FVC

Forced vital capacity

GC

Glucocorticoids

GEMA

Spanish guideline for asthma management

GM-CSF

Granulocyte-macrophage colony-stimulating factor

GNF

Glomerulonephritis

GRAP

Primary Care Respiratory Society

HES

Hypereosinophilic syndrome

HFC

Hydrofluorocarbon

HMW

High molecular weight

HR

Hazard ratio

HRQoL

Health-related quality of life

ICS

Inhaled corticosteroids

ICU

Intensive care unit

IE

Immunosuppressive effect

ICS

Inhaled glucocorticoids

IgE

Immunoglobulin E

IL

Interleukin

ILO

Inducible laryngeal obstruction

INGC

Intranasal glucocorticoids

INPECS

Institute for Clinical and Healthcare Excellence

IOS

Impulse oscillometry

ISAAC

International Study of Asthma and Allergens in Childhood

IT

Immunotherapy

i.v.

Intravenous

kg

kilogram

LABA

Long-acting ß2-adrenergic agonists

LAMA

Long-acting muscarinic antagonist

L-ASL

Lysine-acetylsalicylate

LLN

Lower limit of normality

LMW

Low molecular weight

LPT

Lipid transfer protein

LTRA

Leukotriene receptor antagonists

MA

Meta-analysis

MADM

Mean aerodynamic diameter mass

MART

Maintenance and reliever therapy

Mg

Magnesium

MMF

Mycophenolate mofetil

MR

Magnetic resonance

MTX

Methotrexate

NAR

Non-allergic rhinitis

NEB

Nebulized

NIV

Non-invasive ventilation

NK

Natural killer

nNO

Nasal nitric oxide

NO2

Nitric oxide

NP

Nasal polyposis

NSAID

Non-steroidal anti-inflammatory drug

OA

Occupational asthma

OCS

Oral glucocorticoids

OR

Odds ratio

pACT

Pregnancy Asthma Control Test

pANCA

Perinuclear anti-neutrophil cytoplasmic antibodies

PaCO2

arterial partial pressure of carbon dioxide

PaO2

arterial oxygen partial pressure

PEF

Peak expiratory flow

PD

Provocation dose

pMDI

Pressurized metered-dose inhaler

PMN

Polymorphonuclear neutrophils

ppb

Parts per billion

PPI

Proton pump inhibitor

RADS

Reactive airways dysfunction syndrome

RCT

Randomized controlled trial

RR

Risk ratio

RTX

Rituximab

RSV

Respiratory syncytial virus

SABA

Short-acting (2-adrenergic agonists

SaO2

Arterial oxygen saturation

SARS-Cov-2

Severe acute respiratory syndrome coronavirus 2

SBPT

Specific bronchial provocation test

s.c.

Subcutaneous

SEAIC

Spanish Society of Allergology and Clinical Immunology

SEDISA

Spanish Society of Healthcare Directors

SEFAC

Spanish Society of Clinical Family and Community Pharmacy

SEFC

Spanish Society of Clinical Pharmacology

SEFH

Spanish Society of Hospital Pharmacy

SEICAP

Spanish Society of Clinical Immunology, Allergology and Pediatric Asthma

SEMERGEN

Spanish Society of Primary Care Physicians

SEMES

Spanish Society of Emergency Medicine and Emergency Care

SEMFYC

Spanish Society of Family and Community Medicine

SEMG

Spanish Society of General and Family Physicians

SENP

Spanish Society of Pediatric Pneumology

SEORL-CCC

Spanish Society of Otorhinolaryngology and Head and Neck Surgery

SO2

Sulfur dioxide

SEPAR

Spanish Society of Pneumology and Thoracic Surgery

SEPEAP

Spanish Society of Outpatient and Primary Care Pediatrics

SGC

Systemic glucocorticoids

SMI

Soft mist inhaler

SNOT-22

Sino-Nasal Outcome Test 22

SPP

Portuguese Society of Pneumology

SR

Systematic review

SUA

Severe uncontrolled asthma

TAI

Test of Adherence to Inhalers

Th2

Type 2 helper T cells

TL

T lymphocytes

TNF

Tumor necrosis factor

TPR

Therapeutic positioning report

TSLP

Thymic stromal lymphopoietin

uLTE4

E4 leukotriene concentration in urine

USAA

Uncontrolled severe allergic asthma

VAS

Visual analogue scale

References
[1]
Plaza V (Coord). Guía Española para el Manejo del Asma 4.0 (GEMA4.0). Madrid: Luzán 5; 2015.
[2]
Grupo de trabajo sobre actualización de GPC. Actualización de Guías de Práctica Clínica en el Sistema Nacional de Salud. Manual Metodológico. Plan de Calidad para el Sistema Nacional de Salud del Ministerio de Sanidad y Política Social. Instituto Aragonés de Ciencias de la Salud-I+CS; 2009. Guías de Práctica Clínica en el SNS: I+CS N° 2007/02-01.
[3]
Grupo de trabajo sobre GPC. Elaboración de Guías de Práctica Clínica en el Sistema Nacional de Salud. Manual Metodológico. Madrid: Plan Nacional para el SNS del MSC. Instituto Aragonés de Ciencias de la Sa- lud-I+CS; 2007. Guías de Práctica Clínica en el SNS: I+CS N° 2006/0I.
[4]
2019 GINA Report, Global Strategy for Asthma Management and Prevention (GINA 2019). Disponible en https://ginasthma.org/gina-reports/.
[5]
BTS/SIGN British Guideline on the Management of Asthma 2019 (BTS 2019). Disponible en https://www.brit-thoracic.org.uk/quality-improvement/guidelines/asthma/.
[6]
G.H. Guyatt, A.D. Oxman, G.E. Vist, R. Kunz, Y. Falck-Ytter, P. Alonso-Coello, GRADE Working Group, et al.
GRA- DE: an emerging consensus on rating quality of evidence and strength of recommendations.
[7]
P. Alonso-Coello, D. Rigau, A. Juliana Sanabria, V. Plaza, M. Miravitlles, L. Martinez.
Calidad y fuerza: el sistema GRADE para la formulación de recomendaciones en las guías de práctica clínica.
Arch Bronconeumol., 49 (2013), pp. 261-267
[8]
Página web oficial de The GRADE working grsoup. Disponible en http://www.gradeworkinggroup.org/.
[9]
Guía Española para el Manejo del Asma (GEMA4.4). Madrid: Luzán 5; 2019.
[10]
Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention (GINA 2019). Disponible en: www.ginasthma.org.
[11]
ECRHHS 1996.
Variations in the prevalence of respiratory symptoms, self-reported asthma attacks, and use of asthma medication in the European Community Respiratory Health Survey.
Eur Respir, 9 (1996), pp. 687-695
[12]
ECRHHS 2002.
The European Community Respiratory Health Survey II.
EurRespir J., 20 (2002), pp. 1071-1079
[13]
GBD 2015 Chronic Respiratory Disease Collaborators.
Global, regional, and national deaths, prevalence, disability-adjusted life years, and years lived with disability for chronic obstructive pulmonary disease and asthma, 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015.
Lancet Respir Med., 5 (2017), pp. 691-706
[14]
B. Lundbäck, H. Backman, J. Lötvall, E. Rönmark.
Is asthma prevalence still increasing?.
Expert Rev Respir Med., 10 (2016), pp. 39-51
[15]
Grupo Español del Estudio Europeo en Asma.
Estudio europeo del asma Prevalencia de hiperreactividad bronquial y asma en jóvenes en 5 regiones de España.
Med Clin (Barc)., 106 (1996), pp. 761-767
[16]
N. Álvarez, F. Guillén, I. Aguinaga, J. Hermoso de Mendoza, B. Marín, I. Serrano, et al.
Estudio de prevalencia y asociación entre síntomas de asma y obesidad en la población pediátrica de Pamplona.
Nutr Hosp., 30 (2014), pp. 519-525
[17]
I. Elizalde, F. Guillén, I. Aguinaga.
Factores asociados al asma en los niños y adolescentes de la zona rural de Navarra (España).
Aten Primaria., 50 (2018), pp. 332-339
[18]
P. López, A.M. Gandarilla, L. Díez, M. Ordobás.
Evolución de la prevalencia de asma y factores demográficos y de salud asociados en población de 18-64 años de la Comunidad de Madrid (1996-2013).
Rev Esp Salud Pública., 91 (2017), pp. e1-e14
[19]
R. Vila-Rigat, R. Panadès, E. Hernandez, J. Sivecas, X. Blanché, L. Muñoz-Ortiz, et al.
Prevalence of Work-Related Asthma and its Impact in Primary Health Care.
Arch Bronconeumol., 51 (2015), pp. 449-455
[20]
S.J. Arias, H. Neffen, J.C. Bossio, C.A. Calabrese, A.J. Videla, G.A. Armando, et al.
Prevalencia y características clínicas del asma en adultos jóvenes en zonas urbanas de Argentina.
Arch Bronconeumol., 54 (2018), pp. 134-139
[21]
S.J. Arbes Jr., P.J. Gergen, B. Vaughn, D.C. Zeldin.
Asthma cases attributable to atopy: results from the Third National Health and Nutrition Examination Survey.
J Allergy Clin Immunol., 120 (2007), pp. 1139-1145
[22]
C. Minelli, D.A. van der Plaat, B. Leynaert, R. Granell, A.F.S. Amaral, M. Pereira, et al.
Age at puberty and risk of asthma: A Mendelian randomisation study.
PLoS Med., 15 (2018), pp. e1002634
[23]
K.B. Egan, A.S. Ettinger, M.B. Bracken.
Childhood body mass index and subsequent physiciandiagnosed asthma: a systematic review and meta-analysis of prospective cohort studies.
BMC Pediatr., 13 (2013), pp. 121
[24]
V.J. Carey, S.T. Weiss, I.B. Tager, S.R. Leeder, F.E. Speizer.
Airways responsiveness, wheeze onset, and recurrent asthma episodes in Young adolescents. The East Boston Childhood Respiratory Disease Cohort.
Am J Respir Crit Care Med., 153 (1996), pp. 356-361
[25]
S. Guerra, D.L. Sherrill, F.D. Martinez, R.A. Barbee.
Rhinitis as an independent risk factor for adult-onset asthma.
J Allergy Clin Immunol., 109 (2002), pp. 419-425
[26]
J.A. Burgess, E.H. Walters, G.B. Byrnes, M.C. Matheson, M.A. Jenkins, C.L. Wharton, et al.
Childhood allergic rhinitis predicts asthma incidence and persistence to middle age: a longitudinal study.
J Allergy Clin Immunol., 120 (2007), pp. 863-869
[27]
R. Shaaban, M. Zureik, D. Soussan, C. Neukirch, J. Heinrich, J. Sunyer, et al.
Rhinitis and onset of asthma: a longitudinal population-based study.
Lancet., 372 (2008), pp. 1049-1057
[28]
F. Gómez, J.A. Burgess, S. Villani, J. Dratva, J. Heinrich, C. Janson, et al.
Maternal age at delivery, lung function and asthma in offspring: a population-based survey.
[29]
J. Stokholm, A. Sevelsted, U.D. Anderson, H. Bisgaard.
Preeclampsia Associates with Asthma, Allergy, and Eczema in Childhood.
Am J Respir Crit Care Med, 195 (2017), pp. 614-621
[30]
J.V. Been, M.J. Lugtenberg, E. Smets, C.P. van Schayck, B.W. Kramer, M. Mommers, et al.
Preterm birth and childhood wheezing disorders: a systematic review and metaanalysis.
PLoS Med., 11 (2014), pp. e1001596
[31]
C. Leps, C. Carson, M.A. Quigley.
Gestational age at birth and wheezing trajectories at 3-11 years.
Arch Dis Child., 103 (2018), pp. 1138-1144
[32]
M.C. Tollånes, D. Moster, A.K. Daltveit, L.M. Irgens.
Cesarean section and risk of severe childhood asthma: a populationbased cohort study.
J Pediatr., 153 (2008), pp. 112-116
[33]
M.S. Ku, H.L. Sun, J.N. Sheu, H.S. Lee, S.F. Yang, K.H. Lue.
Neonatal jaundice is a risk factor for childhood asthma: a retrospective cohort study.
Pediatr Allergy Immunol., 23 (2012), pp. 623-628
[34]
K.M. Silvers, C.M. Frampton, K. Wickens, P.K. Pattemore, T. Ingham, D. Fishwick, New Zealand Asthma, Allergy Cohort Study Group, et al.
Breastfeeding protects against current asthma up to 6 years of age.
J Pediatr., 160 (2012), pp. 991-996
[35]
M. Gdalevich, D. Mimouni, M. Mimouni.
Breast-feeding and the risk of bronchial asthma in childhood: a systematic review with meta-analysis of prospective studies.
J Pediatr., 139 (2001), pp. 261-266
[36]
D.P. Strachan, B.K. Butland, H.R. Anderson.
Incidence and prognosis of asthma and wheezing illness from early childhood to age 33 in a national British cohort.
BMJ., 312 (1996), pp. 1195-1199
[37]
H. Burke, J. Leonardi-Bee, A. Hashim, H. Pine-Abata, Y. Chen, D.G. Cook, et al.
Prenatal and passive smoke exposure and incidence of asthma and wheeze: systematic review and meta-analysis.
Pediatrics., 129 (2012), pp. 735-744
[38]
J. Cunningham, G.T. O’Connor, D.W. Dockery, F.E. Speizer.
Environmental tobacco smoke, wheezing, and asthma in children in 24 communities.
Am J Respir Crit Care Med., 153 (1996), pp. 218-224
[39]
Å. Neuman, C. Hohmann, N. Orsini, G. Pershagen, E. Eller, H.F. Kjaer, et al.
ENRIECO Consortium. Maternal smoking in pregnancy and asthma in preschool children: a pooled analysis of eight birth cohorts.
Am J Respir Crit Care Med, 186 (2012), pp. 1037-1043
[40]
A.A. Litonjua, S.L. Rifas-Shiman, N.P. Ly, K.G. Tantisira, J.W. Rich-Edwards, C.A. Camargo Jr., et al.
Maternal antioxidant intake in pregnancy and wheezing illnesses in children at 2 y of age.
Am J Clin Nutr., 84 (2006), pp. 903-911
[41]
H.M. Wolsk, B.L. Chawes, A.A. Litonjua, B.W. Hollis, J. Waage, J. Stokholm, et al.
Prenatal vitamin D supplementation reduces risk of asthma/recurrent wheeze in early childhood: A combined analysis of two randomized controlled trials.
PLoS One., 12 (2017 Oct 27), pp. e0186657
[42]
G. Devereux, A.A. Litonjua, S.W. Turner, L.C. Craig, G. McNeill, S. Martindale, et al.
Maternal vitamin D intake during pregnancy and early childhood wheezing.
Am J Clin Nutr., 85 (2007), pp. 853-859
[43]
L. García-Marcos, J.A. Castro-Rodriguez, G. Weinmayr, et al.
Influence of Mediterranean diet on asthma in children: a systematic review and meta-analysis.
Pediatr Allergy Immunol., 24 (2013), pp. 330-338
[44]
A.M. Hibbs, K. Ross, L.A. Kerns, C. Wagner, M. Fuloria, S. Groh-Wargo, et al.
Effect of Vitamin D Supplementation on Recurrent Wheezing in Black Infants Who Were Born Preterm: The D-Wheeze Randomized Clinical Trial.
JAMA., 319 (2018), pp. 2086-2094
[45]
G. Håland, K.C. Carlsen, L. Sandvik, C.S. Devulapalli, M.C. Munthe-Kaas, M. Pettersen, et al.
Reduced lung function at birth and the risk of asthma at 10 years of age.
N Engl J Med., 355 (2006), pp. 1682-1689
[46]
M. Kerkhof, A.H. Wijga, B. Brunekreef, H.A. Smit, J.C. de Jongste, R.C. Aalberse, et al.
Effects of pets on asthma development up to 8 years of age: the PIAMA study.
Allergy., 64 (2009), pp. 1202-1208
[47]
M. Kogevinas, J.P. Zock, D. Jarvis, H. Kromhout, L. Lillienberg, E. Plana, et al.
Exposure to substances in the workplace and new-onset asthma: an international prospective population-based study (ECRHS-II).
Lancet., 370 (2007), pp. 336-341
[48]
J.A. Hoppin, D.M. Umbach, S.J. London, P.K. Henneberger, G.J. Kullman, M.C. Alavanja, et al.
Pesticides and atopic and nonatopic asthma among farm women in the Agricultural Health Study.
Am J Respir Crit Care Med., 177 (2008), pp. 11-18
[49]
S. Illi, E. von Mutius, S. Lau, R. Bergmann, B. Niggemann, C. Sommerfeld, MAS Group, et al.
Early childhood infectious diseases and the development of asthma up to school age: a birth cohort study.
BMJ., 322 (2001), pp. 390-395
[50]
F.D. Gilliland, T. Islam, K. Berhane, W.J. Gauderman, R. McConnell, E. Avol, et al.
Regular smoking and asthma incidence in adolescents.
Am J Respir Crit Care Med., 174 (2006), pp. 1094-1100
[51]
P.F. Coogan, N. Castro-Webb, J. Yu, G.T. O’Connor, J.R. Palmer, L. Rosenberg.
Active and passive smoking and the incidence of asthma in the Black Women's Health Study.
Am J Respir Crit Care Med., 191 (2015), pp. 168-176
[52]
P. Orellano, N. Quaranta, J. Reynoso, B. Balbi, J. Vasquez.
Association of outdoor air pollution with the prevalence of asthma in children of Latin America and the Caribbean: A systematic review and meta-analysis.
J Asthma., 55 (2018), pp. 1174-1186
[53]
J.E. Sordillo, C.V. Scirica, S.L. Rifas-Shiman, M.W. Gillman, S. Bunyavanich, C.A. Camargo Jr., et al.
Prenatal and infant exposure to acetaminophen and ibuprofen and the risk for wheeze and asthma in children.
J Allergy Clin Immunol., 135 (2015), pp. 441
[54]
T. Lai, M. Wu, J. Liu, M. Luo, L. He, X. Wang, et al.
Acid-Suppressive Drug Use During Pregnancy and the Risk of Childhood Asthma: A Meta-analysis.
Pediatrics., 141 (2018), pp. e20170889
[55]
F. Marra, L. Lynd, M. Coombes, K. Richardson, M. Legal, J.M. Fitzgerald, et al.
Does antibiotic exposure during infancy lead to development of asthma?: a systematic review and metaanalysis.
Chest., 129 (2006), pp. 608-610
[56]
E. Goksör, B. Alm, R. Pettersson, P. Möllborg, L. Erdes, N. Aberg, et al.
Early fish introduction and neonatal antibiotics affect the risk of asthma into school age.
Pediatr Allergy Immunol., 24 (2013), pp. 339-344
[57]
K. Loewen, B. Monchka, S.M. Mahmud, G. ’t Jong, M.B. Azad.
Prenatal antibiotic exposure and childhood asthma: a population-based study.
[58]
L. Hoskin-Parr, A. Teyhan, A. Blocker, A.J. Henderson.
Antibiotic exposure in the first two years of life and development of asthma and other allergic diseases by 7.5 yr: a dose-dependent relationship.
Pediatr Allergy Immunol., 24 (2013), pp. 762-771
[59]
I. Romieu, A. Fabre, A. Fournier, F. Kauffmann, R. Varraso, S. Mesrine, et al.
Postmenopausal hormone therapy and asthma onset in the E3N cohort.
Thorax., 65 (2010), pp. 292-297
[60]
S.F. Thomsen.
Genetics of asthma: an introduction for the clinician.
Eur Clin Respir J., 2 (2015 Jan 16),
[61]
World Health Organization. (2016). Ambient air pollution: a global assessment of exposure and burden of disease. World Health Organization. Disponible en https://apps.who.int/iris/handle/10665/250141.
[62]
Health Effects Institute. 2019. State of Global Air 2019: Special Report on global exposure to air pollution and its disease burden. Boston, MA: Health Effects Institute. Disponible en https://www.stateofglobalair.org/sites/default/files/soga_2019_report.pdf.
[63]
WMO-No. 1233. World Meteorological Organization, 2019.
[64]
S.J. Levine, S.E. Wenzel.
Narrative review: the role of Th2 immune pathway modulation in the treatment of severe asthma and its phenotypes.
Ann Intern Med., 152 (2010), pp. 232-237
[65]
B.N. Lambrecht, H. Hammad.
The airway epithelium in asthma.
Nat Med., 18 (2012), pp. 684-692
[66]
C.J. Koziol-White, R.A. Panettieri Jr..
Airway smooth muscle and mmunomodulation in acute exacerbations of air way disease.
Immunol Rev., 242 (2011), pp. 178-185
[67]
M. Komai-Koma, D. Xu, Y. Li, A.N. McKenzie, I.B. McInnes, F.Y. Liew.
IL-33 is a chemoattractant for human Th2 cells.
Eur J Immunol., 37 (2007), pp. 2779-2786
[68]
B. Zhou, M.R. Comeau, T. De Smedt, H.D. Liggitt, M.E. Dahl, D.B. Lewis, et al.
Thymic stromal lymphopoietin as a key initiator of allergic airway inflammation in mice.
Nat Immunol., 6 (2005), pp. 1047-1053
[69]
C.M. Lloyd, E.M. Hessel.
Functions of T cells in asthma: more than just T(H)2 cells.
Nat Rev Immunol., 10 (2010), pp. 838-848
[70]
S.J. Galli, M. Tsai.
IgE and mast cells in allergic disease.
Nat Med., 18 (2012), pp. 693-704
[71]
H.F. Rosenberg, K.D. Dyer, P.S. Foster.
Eosinophils: changing perspectives in health and disease.
Nat Rev Immunol., 13 (2013), pp. 9-22
[72]
B.N. Lambrecht, H. Hammad.
The role of dendritic and epithelial cells as master regulators of allergic airway inflammation.
Lancet., 376 (2010), pp. 835-843
[73]
M. Yang, R.K. Kumar, P.M. Hansbro, P.S. Foster.
Emerging roles of pulmonary macrophages in driving the development of severe asthma.
J Leukoc Biol., 91 (2012), pp. 557-569
[74]
P. Sui, D.L. Wiesner, J. Xu, Y. Zhang, J. Lee, S. van Dyken, et al.
Pulmonary neuroendocrine cells amplify allergic asthma responses.
[75]
M. Veldhoen.
Interleukin 17 is a chief orchestrator of immunity.
Nat Immunol., 18 (2017), pp. 612-621
[76]
S. Al-Muhsen, J.R. Johnson, Q. Hamid.
Remodeling in asthma.
J Allergy Clin Immunol., 128 (2011), pp. 451-462
[77]
C.L. Grainge, L.C. Lau, J.A. Ward, V. Dulay, G. Lahiff, S. Wilson, et al.
Effect of bronchoconstriction on airway remodeling in asthma.
N Engl J Med., 364 (2011), pp. 2006-2015
[78]
E.M. Dunican, B.M. Elicker, D.S. Gierada, S.K. Nagle, M.L. Schiebler, J.D. Newell, et al.
Mucus plugs in patients with asthma linked to eosinophilia and airflow obstruction.
J Clin Invest., 128 (2018), pp. 997-1009
[79]
D.J. Jackson, S.L. Johnston.
The role of viruses in acute exacerbations of asthma.
J Allergy Clin Immunol., 125 (2010), pp. 1178-1187
[80]
J.M. Weiler, J.D. Brannan, C.C. Randolph, T.S. Hallstrand, J. Parsons, W. Silvers, et al.
Exercise-induced bronchoconstriction up to date.
J Allergy Clin Immunol., 138 (2016), pp. 1292-1295
[81]
K.B. Aguiar, M. Anzolin, L. Zhang.
Global prevalence of exercise-induced bronchoconstriction in childhood: A meta-analysis.
Pediatr Pulmonol., 53 (2018), pp. 412-425
[82]
A. Izquierdo, I. Bobolea, I. Doña, P. Campo, C. Segura, N. Ortega, et al.
Position Statement of the Spanish Society of Allergology and Clinical Immunology on Provocation Tests with Aspirin/Nonsteroidal anti-inflammatory drugs.
J Investig Allergol Clin Immunol, 30 (2020), pp. 1-13
[83]
S. Shrewsbury, S. Pyke, M. Britton.
Meta-analysis of increased dose of inhaled steroid or addition of salmeterol in symptomatic asthma (MIASMA).
BMJ., 320 (2000), pp. 1368-1373
[84]
P.M. O’Byrne, M.D. Inman.
Airway hyperresponsiveness.
Chest., 123 (2003), pp. 411S-416S
[85]
A.R. West, H.T. Syyong, S. Siddiqui, C.D. Pascoe, T.M. Murphy, H. Maarsingh, et al.
Airway contractility and remodeling: links to asthma symptoms.
Pulm Pharmacol Ther., 26 (2013), pp. 3-12
[86]
I. Carvajal-Urueña, L. García-Marcos, R. Busquets-Monge, M. Morales, N. García de Andoin, J. Batlles-Garrido, et al.
Variaciones geográficas en la prevalencia de síntomas de asma en los niños y adolescentes españoles. International Study of Asthma and Allergies in Childhood (ISAAC) fase III España.
Arch Bronconeumol., 41 (2005), pp. 659-666
[87]
L. García-Marcos, A.B. Quirós, G.G. Hernández, F. Guillén-Grima, C.G. Díaz, I.C. Ureña, et al.
Stabilization of asthma prevalence among adolescents and increase among schoolchildren (ISAAC phases I and III) in Spain.
Allergy., 59 (2004), pp. 1301-1307
[88]
A. López-Silvarrey-Varela, S. Pértega-Díaz, S. Rueda-Esteban, J.M. Sánchez-Lastres, M.A. San-José-González, M. Sampedro-Campos, et al.
Prevalence and geographic variations in asthma symptoms in children and adolescents in Galicia (Spain).
Arch Bronconeumol., 47 (2011), pp. 274-282
[89]
A. Bercedo, C. Redondo, L. Lastra, M. Gómez, E. Mora, M. Pacheco, et al.
Prevalencia de asma bronquial, rinitis alérgica y dermatitis atópica en adolescentes de 13-14 años de Cantabria.
Bol. Pediatr., 44 (2004), pp. 9-19
[90]
A. Tai, H. Tran, M. Roberts, N. Clarke, A.-M. Gibson, S. Vidmar, et al.
Outcomes of childhood asthma to the age of 50 years.
J Allergy Clin Immunol., 133 (2014), pp. 1572-1578
[91]
L. Moral, G. Vizmanos, J. Torres-Borrego, M. Praena-Crespo, M. Tortajada-Girbés, F.J. Pellegrini, et al.
Asthma diagnosis in infants and preschool children: a systematic review of clinical guidelines.
Allergol Immunopathol (Madr)., 47 (2019), pp. 107-121
[92]
S.C. Dharmage, J.L. Perret, A. Custovic.
Epidemiology of Asthma in Children and Adults.
Front Pediatr., 7 (2019), pp. 246
[93]
A.F. Pennington, M.J. Strickland, K.A. Freedle, M. Klein, C. Drews-Botsch, C. Hansen, et al.
Evaluating early-life asthma definitions as a marker for subsequent asthma in an electronic medical record setting.
Pediatr Allergy Immunol., 27 (2016), pp. 591-596
[94]
D. Hines, N. Modi, S.K. Lee, T. Isayama, G. Sjörs, L. Gagliardi, et al.
Scoping review shows wide variation in the definitions of bronchopulmonary dysplasia in preterm infants and calls for a consensus.
Acta Paediatr., 106 (2017), pp. 366-374
[95]
T. Isayama, S.K. Lee, J. Yang, D. Lee, S. Daspal, M. Dunn, et al.
Revisiting the Definition of Bronchopulmonary Dysplasia: Effect of Changing Panoply of Respiratory Support for Preterm Neonates.
JAMA Pediatr., 171 (2017), pp. 271-279
[96]
D.G. Hancock, B. Charles-Britton, D.L. Dixon, K.D. Forsyth.
The heterogeneity of viral bronchiolitis: A lack of universal consensus definitions.
Pediatr Pulmonol., 52 (2017), pp. 1234-1240
[97]
O. Dumas, J.M. Mansbach, T. Jartti, K. Hasegawa, A.F. Sullivan, P. Piedra, et al.
A clustering approach to identify severe bronchiolitis profiles in children.
[98]
B.A. Kuzik.
Maybe this is just asthma.
Pediatr Pulmonol., 52 (2017), pp. 1531
[99]
D.S. Balekian, R.W. Linnemann, K. Hasegawa, R. Thadhani, C.A. Camargo Jr..
Cohort Study of Severe Bronchiolitis during Infancy and Risk of Asthma by Age 5 Years.
J Allergy Clin Immunol Pract, 5 (2017), pp. 92-96
[100]
M. Ruotsalainen, M.K. Hyvärinen, E. Piippo-Savolainen, M. Korppi.
Adolescent asthma after rhinovirus and respiratory syncytial virus bronchiolitis.
Pediatr Pulmonol., 48 (2013), pp. 633-639
[101]
R.T. Stein, F.D. Martinez.
Asthma phenotypes in childhood: lessons from an epidemiological approach.
Paediatr Respir Rev., 5 (2004), pp. 155-161
[102]
C. Oksel, R. Granell, S. Haider, S. Fontanella, A. Simpson, S. Turner, et al.
STELAR investigators, breathing Together investigators. Distinguishing Wheezing Phenotypes from Infancy to Adolescence. A Pooled Analysis of Five Birth Cohorts.
Ann Am Thorac Soc., 16 (2019), pp. 868-876
[103]
J. Bousquet, J.E. Gern, F.D. Martinez, J.M. Anto, C.C. Johnson, P.G. Holt, et al.
Birth cohorts in asthma and allergic diseases: Report of a NIAID/NHLBI/MeDALL joint workshop.
J Allergy Clin Immunol., 133 (2014), pp. 1535-1546
[104]
J.A. Howrylak, A.L. Fuhlbrigge, R.C. Strunk, R.S. Zeiger, S.T. Weiss, B.A. Raby, for the Childhood Asthma Management, Program Research Group.
Classification of childhood asthma phenotypes and long-term clinical responses to inhaled anti-inflammatory medications.
J Allergy Clin Immunol., 133 (2014), pp. 1289-1300
[105]
M. Depner, O. Fuchs, J. Genuneit, A.M. Karvonen, A. Hyvärinen, V. Kaulek, The PASTURE Study Group, et al.
Clinical and epidemiologic phenotypes of childhood asthma.
Am J Respir Crit Care Med., 189 (2014), pp. 129-138
[106]
J.A. Castro-Rodríguez, C.J. Holberg, A.L. Wright, F.D. Martinez.
A clinical index to define risk of asthma in young children with recurrent wheezing. Am J Respir Crit Care.
Med., 162 (2000), pp. 1403-1406
[107]
J.A. Castro-Rodriguez, L. Cifuentes, F.D. Martinez.
Predicting Asthma Using Clinical Indexes.
Front Pediatr, 7 (2019), pp. 320
[108]
D.R. Holleman, D.L. Simel.
Does the clinical examination predict airflow limitation?.
JAMA., 274 (1995), pp. 1051-1057
[109]
Proceso Asistencial Integrado Asma. Coord: García Polo C. Consejería de salud y familias de la Junta de Andalucía, 2012. Disponible en https://www.juntadeandalucia.es/organismos/saludyfamilias/areas/cali- dad-investigacion-conocimiento/gestion-conocimiento/paginas/pai-asma.html.
[110]
Martín P (Coord.). El Asma en Atención Primaria. Guía de práctica clínica basada en la evidencia. Grupo de respiratorio de la Sociedad Andaluza de Medicina Familiar y Comunitaria. Granada: Ed SAMFYC; 2001.
[111]
SIGN 158. British guideline on the management of asthma. NHS Scotland. British Thoracic Society. July 2019. Disponible en: http://www.sign.ac.uk.
[112]
W. Buke, M. Fesinmeyer, K. Reed, L. Hampon, C. Christen.
Family history as a predictor of asthma risk.
Am J Prev Med., 24 (2003), pp. 160-169
[113]
GINA 2019. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention NHL-BI/WHO Workshop Report. Disponible en: http://www.ginasthma.com.
[114]
P.M. O’Byrne, C. Jenkins, E.D. Bateman.
The paradoxes of asthma management: time for a new approach?.
Eur Respir J, 50 (2017 Sep 9), pp. 1701103
[115]
NICE guideline. Asthma: diagnosis, monitoring and chronic management. Nov 2017. Disponible en: http://www.nice.org.uk//guideline//ng80.
[116]
K. Tomita, H. Sano, Y. Chiba, R. Set, A. Sano, O. Nishiyama, et al.
A scoring algorithm for predicting the presence of adult asthma: a prospective derivation study.
Prim Care Respir J., 22 (2013), pp. 51-58
[117]
E.H. Bel.
Clinical phenotypes of asthma.
Curr Opin Pulm Med., 10 (2004), pp. 44-50
[118]
H.H. Van der Heijden, M.L. Brouwer, F. Hoekstra, P. van der Pol, P.J. Merkus.
Reference values of exhaled nitric oxide in healthy children 1-5 years using off-line tidal breathing.
Pediatr Pulmonol., 49 (2014), pp. 291-295
[119]
R. Louis, I. Satia, I. Ojanguren, F. Schleich, M. Bonini, T. Tonia, et al.
European Respiratory Society Guidelines for the Diagnosis of Asthma in Adults.
Eur Respir J., (2022 Feb 15), pp. 2101585
[120]
Plaza V (Coord.). GEMA 4.4. Guía Española para el Manejo del Asma (GEMA 2019. 4.4). Disponible en: https://www.gemasma.com/acceso-restringido/?redirect_to=https://www.gemasma.com/profesionales/.
[121]
R. Pellegrino, G. Viegi, V. Brusasco, R.O. Crapo, F. Burgos, R. Casaburi, et al.
Interpretative strategies for lung function tests.
Eur Respir J., 26 (2005), pp. 948-968
[122]
P.H. Quanjer, S. Stanojevic, T.J. Cole, X. Baur, G.L. Hall, B.H. Culver, et al.
Multi-ethnic reference values for spirometry for the 3-95-yr age range: the global lung function 2012 equations.
Eur Respir J., 40 (2012), pp. 1324-1343
[123]
S. Stanojevic, D.A. Kaminsky, M.R. Miller, B. Thompson, A. Aliverti, I. Barjaktarevic, et al.
ERS/ATS technical standard on interpretive strategies for routine lung function tests.
Eur Respir J., 60 (2022 Jul 13), pp. 2101499
[124]
B.T. Kitch, A.D. Paitiel, K.M. Kuntz, D.W. Dockery, J.P. Schouten, S.T. Weiss, et al.
A single measure of FEV1 is associated with risk of asthma attacks in long-term follow-up.
Chest., 126 (2004), pp. 1875-1882
[125]
F.W. Dekker, A.C. Schrier, P.J. Sterk, J.H. Dijkman.
Validity of peak expiratory flow measurement in assessing reversibility of airflow obstruction.
Thorax., 47 (1992), pp. 162-166
[126]
K. Phillips, J. Oborne, S. Lewis, T.W. Harrison, A.E. Tattersfield.
Time course of action of two inhaled corticosteroids, fluticasone propionate and budesonide.
Thorax., 59 (2004), pp. 26-30
[127]
H.K. Reddel, C.M. Salome, J.K. Peat, A.J. Woolcock.
Which index of peak expiratory flow is most useful in the management of stable asthma?.
Am J Respir Crit Care Med., 151 (1995), pp. 1320-1325
[128]
H.M. Boezen, J.P. Schouten, D.S. Postma, B. Rijcken.
Distribution of peak expiratory flow variability by age, gender and smoking habits in a random population sample aged 20-70 yrs.
Eur Respir J., 7 (1994), pp. 1814-1820
[129]
M. Perpiñá, F. García, F.J. Álvarez, C. Cisneros, L. Compte, L.M. Entrenas, et al.
Guidelines for the study of nonspecific bronchial hyperresponsiveness in asthma.
Spanish Society of Pulmonology and Thoracic Surgery (SEPAR). Arch Bronconeumol., 49 (2013), pp. 432-446
[130]
D.W. Cockcroft.
Bronchoprovocation methods: direct challenges.
Clin Rev Allergy Immunol., 24 (2003), pp. 19-26
[131]
M. Van den Berge, R.J. Meijer, H.A. Kerstjens, D.M. de Reus, G.H. Koëter, H.F. Kauffman, D.S. Postma.
PC20 adenosine 5′-monophosphate is more closely associated with airway inflammation in asthma than PC20 methacholine.
Am J Respir Crit Care Med., 163 (2001), pp. 1546-1550
[132]
S.D. Anderson, J. Brannan, J. Spring, N. Spalding, L.T. Rodwell, K. Chan, et al.
A new method for bronchialprovocation testing in asthmatic subjects using a dry powder of mannitol.
Am J Respir Crit Care Med., 156 (1997), pp. 758-765
[133]
R.O. Crapo, R. Casaburi, A.L. Coates, P.L. Enright, J.L. Hankinson, C.G. Irvin, et al.
Guidelines for methacholine and exercise challenge testing.
Am J Respir Crit CareMed., 161 (2000), pp. 309-329
[134]
A.L. Coates, J. Wanger, D.W. Cockcroft, B.H. Culver, Z. Diamant, the Bronchoprovocation Testing Task Force: KaiHåkon Carlsen, et al.
ERS technical standard on bronchial challenge testing: general considerations and performance of methacholine challenge tests.
Eur Respir J., 49 (2017), pp. 1601526
[135]
D.W. Cockcroft, K.Y. Murdock, B.A. Berscheid, B.P. Gore.
Sensitivity and specificity of histamine PC20 determination in a random selection of young college students.
J Allergy Clin Immunol., 89 (1992), pp. 23-30
[136]
ATS/ERS2005.
American Thoracic Society/European Respiratory Society. Recommendations for standardized procedures for the online and offline measurement of exhaled lower respiratory nitric oxide and nasal nitric oxide, 2005.
Am J Respir Crit Care Med, 171 (2005), pp. 912-930
[137]
C.R. Kuoa, M. Spearsb, J. Haughney, A. Smithd, J. Millere, T. Bradshawf, et al.
Scottish consensus statement on the role of FeNO in adult asthma.
Respiratory Medicine., 155 (2019), pp. 54-57
[138]
L.J. Dupont, M.G. Demedts, G.M. Verleden.
Prospective evaluation of the validity of exhaled nitric oxide for the diagnosis of asthma.
Chest., 123 (2003), pp. 751-756
[139]
A.D. Smith, J.O. Cowan, S. Filsell, C. McLachlan, G. Monti-Sheehan, P. Jackson, et al.
Diagnosing asthma: comparisons between exhaled nitric oxide measurements and conventional tests.
Am J Respir Crit Care Med., 169 (2004), pp. 473-478
[140]
D.R. Taylor, M.W. Pijnenburg, A.D. Smith, J.C. de Jongste.
Exhaled nitric oxide measurements: clinical application and interpretation.
Thorax., 61 (2006), pp. 817-827
[141]
L.B. Bacharier, R.C. Strunk, D. Mauger, D. White, R.F. Lemanske Jr., C.A. Sorkness.
Classifying Asthma Severity in Children: Mismatch Between Symptoms.
Medication Use, and Lung Function. Am J Respir Crit Care Med, 170 (2004 15), pp. 426-432
[142]
A.M. Fitzpatrick, W.G. Teague, D.A. Meyers, S.P. Peters, X. Li, H. Li, et al.
Heterogeneity of severe asthma in childhood: confirmation by cluster analysis of children in the National Institutes of Health/National Heart.
Lung, and Blood Institute Severe Asthma Research Program. J Allergy Clin Immunol., 127 (2011), pp. 382-389
[143]
E.A. Gaillard, C.E. Kuehni, S. Turner, M. Goutaki, K.A. Holden, C.A.M. de Jong, et al.
European Respiratory Society clinical practice guidelines for the diagnosis of asthma in children aged 5-6 years.
Eur Respir J, 58 (2021), pp. 2004173
[144]
C. Van Dalen, E. Harding, J. Parkin, S. Cheng, N. Pearce, J. Douwes.
Suitability of forced expiratory volume in 1 s/forced vital capacity vs. percentage of predicted forced expiratory volumen in 1 s for the classification of asthma severity in adolescents.
Arch Pediatr Adolesc Med., 162 (2008), pp. 1169-1174
[145]
S.P. Galant, T. Morphew, S. Amaro, O. Liao.
Value of the broncodilator response in assessing controller naïve asthmatic children.
J Pediatr., 151 (2007), pp. 457-462
[146]
A.M. Tse, D.R. Gold, J.E. Sordillo, E.B. Hoffman, M.W. Gillman, S.L. Rifas-Shiman, et al.
Diagnostic accuracy of the broncodilator response in children.
A Allergy Clin Immunol., 132 (2013), pp. 554-559
[147]
G. Müller-Brandes, U. Krämer, M. Gappa, G. Seitner-Sorge, A. Hüls, A. von Berg, et al.
LUNOKID: can numerical American Thoracic Society/European Respiratory Society quality criteria replace visual inspection of spirometry?.
Eur Respir J., 43 (2014), pp. 1347-1356
[148]
M.R. Miller, J. Hankinson, V. Brusasco, F. Burgos, R. Casaburi, A. Coates.
’ATS/ERS TASK force: standardization of lung function testing’. Standardisation of spirometry.
Eur Respir J, 26 (2005), pp. 319-338
[149]
P.H. Quanjer, D.J. Weiner, J.J. Pretto, D.J. Brazzale, P.W. Boros.
Measurement of FEF 25-75% and FEF75% does not contribute to clinical decision making.
Eur Respir J., 43 (2014), pp. 1051-1058
[150]
O. Asensio, A. Cordón, J. Elorz, A. Moreno, J.R. Villa, Grupo de Técnicas de la Sociedad Española de Neumología Pediátrica.
Estudio de la función pulmonar en el paciente colaborador.
Parte II. An Pediatr (Barc)., 66 (2007), pp. 518-530
[151]
E.G. Pérez-Yarza, J.R. Villa, N. Cobos, M. Navarro, A. Salcedo, C. Martín, et al.
Espirometría forzada en preescolares sanos bajo las recomendaciones de la ATS/ERS: estudio CANDELA.
An Pediatr (Barc)., 70 (2009), pp. 3-11
[152]
S. Stanojevic, A. Wade, S. Lum, J. Stocks.
Reference equations for pulmonary function tests in preschool children: A review.
Pediatric Pulmonology., 42 (2007), pp. 962-972
[153]
C. Martín de Vicente, I. de Mir, S. Rovira, A. Torrent, S. Gartner, Iglesias, et al.
Validation of Global Lung Function Initiative and All Ages Reference Equations for Forced Spirometry in Healthy Spanish Preschoolers.
Arch Bronconeumol., 54 (2018), pp. 24-30
[154]
N. Beydon, S.D. Davis, E. Lombardi, J.L. Allen, H. Arets, P. Aurora, on behalf of the American Thoracic Society/European Respiratory Society Working Group on Infant, Young Children Pulmonary Function Testing, et al.
An Official American Thoracic Society/European Respiratory Society Statement: Pulmonary Function Testing in Preschool Children.
Am J Respir Crit Care Med., 175 (2007), pp. 1304-1345
[155]
L.M. Borrego, J. Stocks, I. Almeida, S. Stanojevic, J. Antunes, P. Leiria-Pinto, et al.
Broncodilator responsiveness using spirometry in healthy and asthmatic preschool children.
Arch Dis Child., 98 (2013), pp. 112-117
[156]
L.E. Busi, S. Restuccia, R. Tourres, P.D. Sly.
Assessing bronchodilator response in presschool children using spirometry.
[157]
H. Knihtilä, A. Kotaniemi-Syrjänen, M.J. Mäkelä, J. Bondestam, A.S. Pelkonen, L.P. Malmberg.
Preschool oscillometry and lung function at adolescence in asthmatic children.
Pediatric Pulmonology., 50 (2015), pp. 1205-1213
[158]
S.B. Batmaz, S. Kuyucu, T. Arikoglu, O. Tezol, A. Aydogdu.
Impulse oscillometry in acute and stable asthmatic children: a comparison with spirometry.
J Asthma., 53 (2016), pp. 179-186
[159]
P. Jara-Gutierrez, E. Aguado, M.G. del Potro, M. Fernandez-Nieto, I. Mahillo, J. Sastre.
Comparison of impulse oscillometry and spirometry for detection of airway hyperresponsiveness to methacholine, mannitol, and eucapnic voluntary hyperventilation in children.
Pediatr Pulmonol., 54 (2019), pp. 1162-1172
[160]
N. Cobos, E.G. Pérez-Yarza, O. Sardón, C. Reverté, S. Gartner, J. Korta.
Óxido nítrico exhalado en niños: un indicador no invasivo de la inflamación de las vías aéreas.
Arch Bronconeumol., 44 (2008), pp. 41-51
[161]
D. Caudri, A.H. Wijga, M. Hoekstra, M. Kerkhof, G.H. Koppelman, B. Brunekreef, et al.
Prediction of asthma in symptomatic preschool children using exhaled nitric oxide, Rint and specific IgE.
Thorax., 65 (2010), pp. 801-807
[162]
K.C. See, D.C. Christiani.
Normal vàlues and thresholds for the clinical interpretation of exhaled nitric oxide levels in the US general population.
Chest., 143 (2013), pp. 107-116
[163]
R.A. Dweik, P.B. Boggs, S.C. Erzurum, C.G. Irvin, M.W. Leigh, J.O. Lundberg, et al.
American Thoracic Society Committee on Interpretation of Exhaled Nitric Oxide Levels (FENO) for Clinical Applications. An official ATS clinical practice guideline: interpretation of exhaled nitric oxide levels (FENO) for clinical applications.
Am J Respir Crit Care Med, 184 (2011), pp. 602-615
[164]
A. Michils, S. Baldassarre, A. Van Muylem.
Exhaled nítric oxide and asthma control: a longitudinal study in unselected patients.
Eur Respir J., 31 (2008), pp. 539-546
[166]
M.W. Pijnenburg, W. Hofhuis, W.C. Hop, J.C. de Jonste.
Exhaled nitric oxide predicts asthma relapse in children with clinical asthma remission.
Thorax., 60 (2005), pp. 215-218
[167]
F. Singer, I. Luchsinger, D. Inci, N. Knauer, P. Latzin, J.H. Wildhaber, et al.
Exhaled nitric oxide in symptomatic children at preschool age predicts later asthma.
Allergy., 68 (2013), pp. 531-538
[168]
X. Wang, X. Tan, Q. Li.
Effectiveness of fractional exhaled nitric oxide for asthma management in children: A systematic review and meta-analysis.
Pediatr Pulmonol., 55 (2020), pp. 1936-1945
[169]
H.L. Petsky, K.M. Kayleigh, A.B. Chang.
Exhaled nitric oxide levels to guide treatment for children with asthma.
Cochrane Database Syst Rev., 11 (2016 Nov 9), pp. CD011439
[170]
Expert Panel Working Group of the National Heart, Lung, Blood Institute (NHLBI) administered, coordinated National Asthma Education, Prevention Program Coordinating Committee(NAEPPCC).
2020 Focused Updates to the Asthma Management Guidelines: A report from the National Asthma Education and Prevention Program Coordinating Committee Expert Panel Working Group.
J Allergy Clin Immunol, 146 (2020), pp. 1217-1270
[171]
G.J. Burbach, L.M. Heinzerling, G. Edenharter, C. Bachert, C. Bindslev-Jensen, S. Bonini, et al.
GA(2)LEN skin test study II: clinical relevance of inhalant allergen sensitizations in Europe.
Allergy., 64 (2009), pp. 1507-1515
[172]
P. Ojeda, J. Sastre, J.M. Olaguibel, T. Chivato.
investigators participating in the National Survey of the Spanish Society of Allergology and Clinical Immunology Alergológica 2015. Alergológica 2015: A National Survey on Allergic Diseases in the Adult Spanish Population.
J Investig Allergol Clin Immunol., 28 (2018), pp. 151-164
[173]
J. Bousquet, L. Heinzerling, C. Bachert, N.G. Papadopoulos, P.J. Bousquet, P.G. Burney, et al.
Practical guide to skin prick tests in allergy to aeroallergens.
[174]
I.L. Bernstein, J.T. Li, D.I. Bernstein, R. Hamilton, S.L. Spector, R. Tan, et al.
Allergy diagnostic testing: an updated practice parameter.
Ann Allergy Asthma Immunol., 100 (2008), pp. S1-S148
[175]
E. González-Mancebo, J. Domínguez-Ortega, S. Blanco-Bermejo, E. González-Seco, M.J. Trujillo, F. de la Torre, et al.
Comparison of two diagnostic techniques, skin-prick test and component resolved diagnosis in the follow-up of a cohort of paediatric patients with pollinosis. Multicentre pilot study in a highly exposed allergenic area.
Allergol Immunopathol (Madr)., 45 (2017), pp. 121-126
[176]
C. Moreno, J.L. Justicia, J. Quiralte, A. Moreno-Ancillo, A. Iglesias-Cadarso, M. Torrecillas, et al.
Olive, grass or both? Molecular diagnosis for the allergen immunotherapy selection in polysensitized pollinic patients.
Allergy., 69 (2014), pp. 1357-1363
[177]
NAEPP-EP 2007. Nacional Asthma Education and Prevention Program. Expert Panel Report Guidelines for the diagnosis and management of asthma. Bethesda: National Institutes of Health, National Heart, Lung, and Blood Institute, 2007. Disponible en https://www.nhlbi.nih.gov/science/national-asthma-education-and-prevention-program-naepp.
[178]
D.W. Cockcroft, V.A. Swystun.
Asthma control versus asthma severity.
J Allergy Clin Immunol., 98 (1996), pp. 1016-1018
[179]
S.W. Stoloff, H.A. Boushey.
Severity, control, and responsiveness in asthma.
J Allergy Clin Immunol., 117 (2006), pp. 544-548
[180]
D.R. Taylor, E.D. Bateman, L.P. Boulet, H.A. Boushey, W.W. Busse, T.B. Casale, et al.
A new perspective on concepts of asthma severity and control.
Eur Respir J., 32 (2008), pp. 545-554
[181]
D. Dusser, D. Montani, P. Chanez.
Mild asthma: an expert review on epidemiology, clinical characteristics and treatment recommendations.
[182]
N. Shahidi, J.M. FitzGerald.
Current recommendations for the treatment of mild asthma.
Journal of Asthma and Allergy., 3 (2010), pp. 169-176
[183]
H.K. Reddel, J.M. FitzGerald, E.D. Bateman, L.B. Bacharier, A. Becker, G. Brusselle, et al.
GINA 2019: a fundamental change in asthma management Treatment of asthma with short-acting bronchodilators alone is no longer recommended for adults and adolescents.
Eur Respir J., 53 (2019), pp. 1901046
[184]
J. Muneswarao, M.Z. Hassali, B. Ibrahim, B. Saini, I.A. Hyder, A.K. Verma.
It is time to change the way we manage mild asthma: an update in GINA 2019.
Respiratory Research., 20 (2019), pp. 183
[185]
M.L. Osborne, W.M. Vollmer, K.L. Pedula, J. Wilkins, A.S. Buist, M. O’Hollaren.
Lack of correlation of symptoms with specialist-assessed long-term asthma severity.
Chest., 115 (1999), pp. 85-91
[186]
E.D. Bateman, H.A. Boushey, J. Bousquet, W.W. Busse, T.J. Clark, R.A. Pauwels, GOAL Investigators Group, et al.
Can guideline-defined asthma control be achieved? The Gaining Optimal Asthma ControL study.
Am J Respir Crit Care Med., 170 (2004), pp. 836-844
[187]
R.A. Nathan, C.A. Sorkness, M. Kosinski, M. Schatz, J.T. Li, P. Marcus, et al.
Development of the asthma control test: a survey for assessing asthma control test.
J Allergy Clin Immunol., 113 (2004), pp. 59-65
[188]
J.M. Vega, X. Badia, C. Badiola, A. López-Viña, J.M. Olaguíbel, C. Picado, Covalair Investigator Group, et al.
Validation of the Spanish version of the Asthma Control Test (ACT).
J Asthma., 44 (2007), pp. 867-872
[189]
E.F. Juniper, P.M. O’Byrne, G.H. Guyatt, P.J. Ferrie, D.R. King.
Development and validation of a questionnaire to measure asthma control.
Eur Respir J., 14 (1999), pp. 902-907
[190]
C. Picado, C. Badiola, N. Perulero, J. Sastre, J.M. Olaguíbel, A. López, Covalair Investigator Group, et al.
Validation of the Spanish version of the Asthma Control Questionnaire.
Clin Ther., 30 (2008), pp. 1918-1931
[191]
M. Schatz, M. Kosinski, A.S. Yarlas, J. Hanlon, M.E. Watson, P. Jhingran.
The minimally important difference of the Asthma Control Test.
J Allergy Clin Immunol., 124 (2009), pp. 719-723
[192]
E.F. Juniper, J. Bousquet, L. Abetz, E.D. Bateman, G.O.A.L. Committee.
Identifying ‘well-controlled’ and ‘not well-controlled’ asthma using the Asthma Control Questionnaire.
Respir Med., 100 (2006), pp. 616-621
[193]
J.M. Olaguibel, S. Quirce, B. Julia, C. Fernandez, A.M. Fortuna, J. Molina, et al.
Measurement of asthma control according to global initiative for asthma guidelines: a comparison with the asthma control questionnaire.
Respir Res., 13 (2012), pp. 50
[194]
C.E. Jia, H.P. Zhang, Y. Lv, R. Liang, Y.Q. Jiang, H. Powell, et al.
The Asthma Control Test and Asthma Control Questionnaire for assessing asthma control: Systematic review and meta-analysis.
J Allergy Clin Immunol, 131 (2013), pp. 695-703
[195]
K.R. Murphy, B. Chipps, D.A. Beuther, R.A. Wise, W. McCann, I. Gilbert, et al.
Development of the Asthma Impairment and Risk Questionnaire (AIRQ): A Composite Control Measure.
J Allergy Clin Immunol Pract., 8 (2020 Jul-Aug), pp. 2263-2274.e5
[196]
L. Pérez de Llano, E. Martínez Moragón, L.M. Entrenas, C. Martínez Rivera, C. Cisneros, M. Blanco-Aparicio, et al.
Validation of the Asthma Impairment and Risk Questionnaire (AIRQ) in Spain: a useful tool to assess asthma control in adolescent and adult patients.
J Investig Allergol Clin Immunol., (2022 Dec 15),
[197]
D.B. Price, A. Rigazio, J.D. Campbell, E.R. Bleecker, C.J. Corrigan, M. Thomas, et al.
Blood eosinophil count and prospective annual asthma disease burden: a UK cohort study.
Lancet Respir Med., 3 (2015), pp. 849-858
[198]
D.B. Price, F. Trudo, J. Voorham, X. Xu, M. Kerkhof, J. Ling Zhi Jie, et al.
Adverse outcomes from initiation of systemic corticosteroids for asthma: long-term observational study.
J Asthma Allergy., 11 (2018), pp. 193-204
[199]
S. Vedel-Krogh, S. Fallgaard Nielsen, P. Lange, J. Vestbo, B.G. Nordestgaard.
Association of Blood Eosinophil and Blood Neutrophil Counts with Asthma Exacerbations in the Copenhagen General Population Study.
Clin Chem., 63 (2017), pp. 823-832
[200]
J. Belda, J. Giner, P. Casan, J. Sanchis.
Mild exacerbations and eosinophilic inflammation in patients with stable, well-controlled asthma after 1 year of follow-up.
Chest., 119 (2001), pp. 1011-1017
[201]
R.S. Zeiger, M. Schatz, F. Zhang, W.W. Crawford, M.S. Kaplan, R.M. Roth, et al.
Elevated exhaled nitric oxide is a clinical indicator of future uncontrolled asthma in asthmatic patients on inhaled corticosteroids.
J Allergy Clin Immunol., 128 (2011), pp. 412-414
[202]
H.L. Petsky, C.J. Cates, K.M. Kew, A.B. Chang.
Tailoring asthma treatment on eosinophilic markers (exhaled nitric oxide or sputum eosinophils): a systematic review and meta-analysis.
Thorax., 73 (2018), pp. 1110-1119
[203]
M.L. Osborne, K.L. Pedula, M. O’Hollaren, K.M. Ettinger, T. Stibolt, A.S. Buist, et al.
Assessing future need for acute care in adult asthmatics: the Profile of Asthma Risk Study: a prospective health maintenance organization-based study.
Chest., 132 (2007), pp. 1151-1161
[204]
M. Ferrer, F.J. Alvarez, A. Romero, B. Romero, A. Sáez, J.F. Medina.
Is the bronchodilator test an useful tool to measure asthma control?.
Respir Med., 126 (2017), pp. 26-31
[205]
A. Menzies-Gow, S.J. Szefler, W.W. Busse.
The Relationship of Asthma Biologics to Remission for Asthma.
J Allergy Clin Immunol Pract, 9 (2021 Mar), pp. 1090-1098
[206]
J.M. Vonk, D.S. Postma, H.M. Boezen, M.H. Grol, J.P. Schouten, G.H. Koëter, et al.
Childhood factors associated with asthma remission after 30 year follow up.
Thorax., 59 (2004 Nov), pp. 925-929
[207]
G.A. Westerhof, H. Coumou, S.B. de Nijs, E.J. Weersink, E.H. Bel.
Clinical predictors of remission and persistence of adult-onset asthma.
J Allergy Clin Immunol., 141 (2018 Jan), pp. 104-109.e3
[208]
S. Khatri, W. Moore, P.G. Gibson, R. Leigh, A. Bourdin, J. Maspero, et al.
Assessment of the long-term safety of mepolizumab and durability of clinical response in patients with severe eosinophilic asthma.
J Allergy Clin Immunol., 143 (2019), pp. 1742-1751
[209]
E.G. Pérez-Yarza, X. Badía, C. Badiola, N. Cobos, J. Garde, M. Ibero, on behalf of the CAN Investigator Group, et al.
Development and validation of a questionnaire to assess asthma control in pediatrics.
Pediatr Pulmonol., 44 (2009), pp. 54-63
[210]
H.K. Reddel, D.R. Taylor, E.D. Bateman, L.-P. Boulet, H.A. Boushey, W.W. Busse, on behalf of the American Thoracic Society/European Respiratory Society Task Force on Asthma Control, Exacerbations, et al.
An Official American Thoracic Society/European Respiratory Society Statement: Asthma Control and Exacerbations. Standardizing Endpoints for Clinical Asthma Trials and Clinical Practice.
Am J Respir Crit Care Med, 180 (2009), pp. 59-99
[211]
A.H. Liu, R. Zeiger, C. Sorkness, T. Mahr, N. Ostrom, S. Burgess, et al.
Development and cross-sectional validation of the Childhood Asthma Control Test.
J Allergy Clin Immunol., 119 (2007), pp. 817-825
[212]
C.E. Rodríguez-Martínez, A. Melo-Rojas, S.M. Restrepo-Gualteros, M.P. Sossa-Briceño, G. Nino.
Validation of the Spanish versión of the childhood astma control test (cACT) in a population of Hispanic children.
J Asthma., 51 (2014), pp. 855-862
[213]
E.G. Pérez-Yarza, J.A. Castro, J.R. Villa, J. Garde, J. Hidalgo, on behalf of the VESCASI Group.
Validation of a Spanish version of the Childhood Asthma Control Test (Sc-ACT) for use in Spain.
An Pediatr (Barc)., 83 (2015), pp. 94-103
[214]
H.T. Den Dekker, A.M.M. Sonnenschein-van der Voort, J.C. de Jongste, I. Anessi-Maesano, S.H. Arshad, H. Barros, et al.
Early growth characteristics and the risk of reduced lung function and asthma. A meta-analysis of 25.000 children.
J Allergy Clin Immunol., 137 (2016), pp. 1026-1035
[215]
A. Buelo, S. McLean, S. Julious, J. Flores-Kim, A. Bush, J. Henderson, et al.
At-risk children with asthma (ARC):a systematic review.
[216]
D. Price, S. Bosnic-Anticevich, A. Briggs, H. Chrystyn, C. Rand, G. Scheuch, et al.
Inhaler Error Steering Committee. Inhaler competence in asthma: common errors, barriers to use and recommended solutions.
Respir Med., 107 (2013), pp. 37-46
[217]
G. Taylor, S. Warren, S. Dwivedi, M. Sommerville, L. Mello, C. Orevillo, et al.
Gamma scintigraphic pulmonary deposition study of glycopyrronium/formoterol metered dose inhaler formulated using co-suspension delivery technology.
Eur J Pharm Sci., 111 (2018), pp. 450-457
[218]
S. Israel, A. Kumar, K. DeAngelis, et al.
Pulmonary deposition of budesonide/glycopyrronium/formoterol fumarate dihydrate metered dose inhaler formulated using co-suspension delivery technology in healthy male subjects.
Eur J Pharm Sci., 153 (2020), pp. 105472
[219]
Wu L, Holsbeke CV, Mack P. Consistent lung delivery of inhaled triple ICS/LAMA/LABA fixed-dose combination using co-suspension delivery technology: an in silico modeling study [poster]. Presented at American Association of Pharmaceutical Scientists PharmSci 360 Congress; November 3-6, 2019; San Antonio, TX. Poster T1530-13-89.
[220]
A. Doty, J. Schroeder, K. Vang, M. Sommerville, M. Taylor, B. Flynn, et al.
Drug delivery from an innovative LAMA/LABA co-suspension delivery technology fixed-dose combination MDI: evidence of consistency, robustness, and reliability.
AAPS PharmSciTech., 19 (2018), pp. 837-844
[221]
R. Polosa, J.D. Knoke, C. Russo, G. Piccillo, P. Caponnetto, M. Sarvà, et al.
Cigarette smoking is associated with a greater risk of incident asthma in allergic rhinitis.
J Allergy Clin Immunol., 121 (2008), pp. 1428-1434
[222]
S.P. Newman, S.W. Clarke.
Therapeutic aerosols 1–physical and practical considerations.
Thorax., 38 (1983), pp. 881-886
[223]
L.P. Boulet, A. Becker, D. Berubé, R. Beveridge, P. Ernst, on behalf of the Canadian Asthma Consensus Group.
Summary of recommendations from the Canadian Asthma Consensus report 1999.
CMAJ., 161 (1999), pp. S1-S12
[224]
P.G. Gibson, H. Powell, F.M. Ducharme.
Differential effects of maintenance long-acting beta-agonist and inhaled corticosteroid on asthma control and asthma exacerbations.
J Allergy ClinImmunol., 119 (2007), pp. 344-350
[225]
J.D. Blakey, K. Woolnough, J. Fellows, S. Walker, M. Thomas, I.D. Pavord.
Assessing the risk of attack in the management of asthma: a review and proposal for revision of the current control-centred paradigm.
Prim Care Respir J., 22 (2013), pp. 344-352
[226]
E.D. Bateman, L. Jacques, C. Goldfrad, T. Atienza, T. Mihaescu, M. Duggan.
Asthma control can be maintained when fluticasone propionate/salmeterol in a single inhaler is stepped down.
J Allergy Clin Immunol., 117 (2006), pp. 563-570
[227]
L. Pérez de Llano, J.L. García-Rivero, I. Urrutia, E. Martínez-Moragón, J. Ramos, P. Cebollero, et al.
A Simple Score for Future Risk Prediction in Patients with Controlled Asthma Who Undergo a Guidelines-Based Step-Down Strategy.
J Allergy Clin Immunol Pract, 7 (2019), pp. 1214-1221.e3
[228]
Global Initiative for Asthma: Global Strategy for Asthma Management and Prevention (2019 update). Disponible en: https://ginasthma.org/wp-content/uploads/2019/06/GINA-2019-main-report-June-2019-wms.pdf.
[229]
R. Beasley, M. Holliday, H.K. Reddel, I. Braithwaite, S. Ebmeier, R.J. Hancox, et al.
Novel START Study Team.
Controlled trial of budesonide-formoterol as needed for mild asthma. N Engl J Med., 380 (2019), pp. 2020-2030
[230]
T. Haahtela, K. Tamminen, L.P. Malmberg, O. Zetterstrom, J. Karjalainen, H. Yla-Outinen, et al.
Formoterol as needed with or without budesonide in patients with intermittent asthma and raised NO levels in exhaled air: A SOMA study.
Eur Respir J., 28 (2006), pp. 748-755
[231]
A. Papi, G.W. Canonica, P. Maestrelli, P. Paggiaro, D. Olivieri, E. Pozzi, BEST Study Group, et al.
Rescue use of beclomethasone and albuterol in a single inhaler for mild asthma.
N Engl J Med., 356 (2007), pp. 2040-2052
[232]
Global Initiative for Asthma: Global Strategy for Asthma Management and Prevention 2018. Disponible en: www.ginasthma.org.
[233]
SIGN-158-british-guideline-on-the-management-of-asthma. html Disponible en https://www.sign.ac.uk/sign-158-british-guideline-on-the-management-of-asthma.
[234]
T. Sriprasart, G. Waterer, G. Garcia, A. Rubin, M.A.L. Andrade, A. Roguska, et al.
Safety of SABA Monotherapy in Asthma Management: a Systematic Review and Meta-analysis.
Adv Ther., 40 (2023 Jan), pp. 133-158
[235]
H.K. Reddel, J.M. FitzGerald, E.D. Bateman, L.B. Bacharier, A. Becker, G. Brusselle, et al.
GINA 2019: a fundamental change in asthma management: treatment of asthma with short-acting bronchodilators alone is no longer recommended for adults and adolescents.
Eur Respir J., 53 (2019), pp. 1901046
[236]
L. De las Vecillas, S. Quirce.
Landscape of short-acting beta-agonists (SABA) overuse in Europe.
Clin Exp Allergy., 53 (2023), pp. 132-144
[237]
J.K. Quint, S. Arnetorp, J.W.H. Kocks, M. Kupczyk, J. Nuevo, V. Plaza, et al.
Short-Acting Beta-2-Agonist Exposure and Severe Asthma Exacerbations: SABINA Findings From Europe and North America.
J Allergy Clin Immunol Pract., 10 (2022), pp. 2297-2309.e10
[238]
F. Montero-Arias, J.C.H. García, M.P. Gallego, M.A. Antila, P. Schonffeldt, W.J. Mattarucco, et al.
Over-prescription of short-acting ß2-agonists is associated with poor asthma outcomes: results from the Latin American cohort of the SABINA III study.
J Asthma., 60 (2023), pp. 574-587
[239]
D.W. Cockcroft.
As-needed inhaled beta2-adrenoceptor agonists in moderate-to-severe asthma: current recommendations.
Treat Respir Med., 4 (2005), pp. 169-174
[240]
R.A. Pauwels, S. Pedersen, W.W. Busse, W.C. Tan, Y.Z. Chen, S.V. Ohlsson, START Investigators Group, et al.
Early intervention with budesonide in mild persistent asthma: a randomised, double-blind trial.
Lancet., 361 (2003), pp. 1071-1076
[241]
R.S. Zeiger, J.W. Baker, M.S. Kaplan, D.S. Pearlman, M. Schatz, S. Bird, et al.
Variability of symptoms in mild persistent asthma: baseline data from the MIAMI study.
Respir Med., 98 (2004), pp. 898-905
[242]
R.A. Tan, S.L. Spector.
Exercise-induced asthma: diagnosis and management.
Ann Allergy Asthma Immunol., 89 (2002), pp. 226-235
[243]
N.P. Adams, J.C. Bestall, T.J. Lasserson, P.W. Jones, C. Cates.
Fluticasone versus placebo for chronic asthma in adults and children.
Cochrane Database Syst Rev., (2005), pp. CD003135
[244]
N.P. Adams, J.B. Bestall, R. Malouf, T.J. Lasserson, P.W. Jones.
Inhaled beclomethasone versus placebo for chronic asthma.
Cochrane Database Syst Rev., (2005), pp. CD002738
[245]
M.S. Koh, L.B. Irving.
Evidence-based pharmacologic treatment for mild asthma.
Int J ClinPract., 61 (2007), pp. 1375-1379
[246]
H.K. Reddel, E.G. Belousova, G.B. Marks, C.R. Jenkins.
Does continuous use of inhaled corticosteroids improve outcomes in mild asthma?. A double-blind randomised controlled trial.
Prim Care Respir J., 17 (2008), pp. 39-45
[247]
P.M. O’Byrne, H. Bisgaard, P.P. Godard, M. Pistolesi, M. Palmqvist, Y. Zhu, et al.
Budesonide/formoterol combination therapy as both maintenance and reliever medication in asthma.
Am J Respir Crit Care Med., 171 (2005), pp. 129-136
[248]
S. Suissa, P. Ernst.
Inhaled corticosteroids: impact on asthma morbidity and mortality.
J Allergy Clin Immunol., 107 (2001), pp. 937-944
[249]
P. Daley-Yates, N. Brealey, S. Thomas, D. Austin, S. Shabbir, T. Harrison, et al.
Therapeutic index of inhaled corticosteroids in asthma: A dose-response comparison on airway hyperresponsiveness and adrenal axis suppression.
Br J Clin Pharmacol., 87 (2021), pp. 483-493
[250]
V. Plaza, A. Gómez-Outes, S. Quirce, I. Alobid, C. Álvarez, M. Blanco, et al.
Discrepancies Between GEMA and GINA in the Classification of Inhaled Corticosteroids.
Arch Bronconeumol, 56 (2020), pp. 472-473
[251]
P.M. O’Byrne, J.M. FitzGerald, E.D. Bateman, P.J. Barnes, N. Zhong, C. Keen, et al.
Inhaled Combined Budesonide-Formoterol as Needed in Mild Asthma.
N Engl J Med, 378 (2018), pp. 1865-1876
[252]
E.D. Bateman, H.K. Reddel, P.M. O’Byrne, P.J. Barnes, N. Zhong, C. Keen, et al.
As-needed budesonide-formoterol versus maintenance budesonide in mild asthma.
N Engl J Med., 378 (2018), pp. 1877-1887
[253]
R.S. Zeiger, S.R. Bird, M.S. Kaplan, M. Schatz, D.S. Pearlman, E.J. Orav, et al.
Short-term and long-term asthma control in patients with mild persistent asthma receiving montelukast or fluticasone: a randomized controlled trial.
Am J Med., 118 (2005), pp. 649-657
[254]
B.F. Chauhan, F.M. Ducharme.
Anti-leukotriene agents compared to inhaled corticosteroids in the management of recurrent and/or chronic asthma in adults and children.
Cochrane Database Syst Rev., (2012), pp. CD002314
[255]
S.P. Peters, N. Anthonisen, M. Castro, J.T. Holbrook, C.G. Irvin, L.J. Smith, et al.
ALA. American Lung Association Asthma Clinical Research Centers. Randomized comparison of strategies for reducing treatment in mild persistent asthma.
N Engl J Med, 356 (2007), pp. 2027-2039
[256]
W.W. Busse, T.B. Casale, M.S. Dykewicz, E.O. Meltzer, S.R. Bird, C.M. Hustad, et al.
Efficacy of montelukast during the allergy season in patients with chronic asthma and seasonal aeroallergen sensitivity.
Ann Allergy Asthma Immunol., 96 (2006), pp. 60-68
[257]
D.B. Price, A. Swern, C.A. Tozzi, G. Philip, P. Polos.
Effect of montelukast on lung function in asthma patients with allergic rhinitis: analysis from the COMPACT trial.
[258]
M. Ni Chroinin, I. Greenstone, T.J. Lasserson, F.M. Ducharme.
Addition of inhaled long-acting beta2-agonists to inhaled steroids as first line therapy for persistent asthma in steroid-naive adults and children.
Cochrane Database Syst Rev., (2009), pp. CD005307
[259]
A. Woolcock, B. Lundback, N. Ringdal, L.A. Jacques.
Comparison of addition of salmeterol to inhaled steroids with doubling of the dose of inhaled steroids.
Am J RespirCrit Care Med., 153 (1996), pp. 1481-1488
[260]
R.A. Pauwels, C.G. Löfdahl, D.S. Postma, A.E. Tattersfield, P. O’Byrne, P.J. Barnes, et al.
Effect of inhaled formoterol and budesonide on exacerbations of asthma. Formoterol and Corticosteroids Establishing Therapy (FACET) International Study Group.
N Engl J Med., 337 (1997), pp. 1405-1411
[261]
S. Shrewsbury, S. Pyke, M. Britton.
Meta-analysis of increased dose of inhaled steroid or addition of salmeterol in symptomatic asthma (MIASMA).
BMJ., 320 (2000), pp. 1368-1373
[262]
E.D. Bateman, H.A. Boushey, J. Bousquet, W.W. Busse, T.J. Clark, R.A. Pauwels, GOAL Investigators Group, et al.
Can guideline-defined asthma control be achieved? The Gaining Optimal Asthma ControL study.
Am J Respir Crit Care Med., 170 (2004), pp. 836-844
[263]
I.R. Greenstone, M.N. Ni Chroinin, V. Masse, A. Danish, H. Magdalinos, X. Zhang, et al.
Combination of inhaled long-acting beta2-agonists and inhaled steroids versus higher dose of inhaled steroids in children and adults with persistent asthma.
Cochrane Database Syst Rev., (2005), pp. CD005533
[264]
M. Masoli, M. Weatherall, S. Holt, R. Beasley.
Moderate dose inhaled corticosteroids plus salmeterol versus higher doses of inhaled corticosteroids in symptomatic asthma.
Thorax., 60 (2005), pp. 730-734
[265]
S.R. Salpeter, N.S. Buckley, T.M. Ormiston, E.E. Salpeter.
Meta-analysis: effect of long-acting beta-agonists on severe asthma exacerbations and asthma-related deaths.
Ann Intern Med., 144 (2006), pp. 904-912
[266]
M. Weatherall, M. Wijesinghe, K. Perrin, M. Harwood, R. Beasley.
Meta-analysis of the risk of mortality with salmeterol and the effect of concomitant inhaled corticosteroid therapy.
Thorax., 65 (2010), pp. 39-43
[267]
J. Lötvall, E.D. Bateman, E.R. Bleecker, W.W. Busse, A. Woodcock, R. Follows, et al.
24-h duration of the novel LABA vilanterol trifenatate in asthma patients treated with inhaled corticosteroids.
Eur Respir J., 40 (2012), pp. 570-579
[268]
D.S. Pearlman, L. Greos, C. LaForce, C.J. Orevillo, R. Owen, M. Higgins.
Bronchodilator efficacy of indacaterol, a novel once-daily beta2-agonist, in patients with persistent asthma.
Ann Allergy Asthma Immunol., 101 (2008), pp. 90-95
[269]
K. Chapman, R. van Zyl-Smit, J. Maspero, H.A.M. Kerstjens, Y. Gon, M. Hosoe, et al.
One time a day mometasone/indacaterol fixed-dose combination versus two times a day fluticasone/salmeterol in patients with inadequately controlled asthma: pooled analysis from PALLADIUM and IRIDIUM studies.
BMJ Open Resp Res., 8 (2021), pp. e000819
[270]
K.F. Rabe, T. Atienza, P. Magyar, P. Larsson, C. Jorup, U.G. Lalloo.
Effect of budesonide in combination with formoterol for reliever therapy in asthma exacerbations: a randomised controlled, double-blind study.
Lancet., 368 (2006), pp. 744-753
[271]
K.F. Rabe, E. Pizzichini, B. Ställberg, S. Romero, A.M. Balanzat, T. Atienza, et al.
Budesonide/formoterol in a single inhaler for maintenance and relief in mild-to-moderate asthma: a randomized, double-blind trial.
Chest., 129 (2006), pp. 246-256
[272]
C. Vogelmeier, A. D’Urzo, R. Pauwels, J.M. Merino, M. Jaspal, S. Boutet, et al.
Budesonide/formoterol maintenance and reliever therapy: an effective asthma treatment option?.
EurRespir J., 26 (2005), pp. 819-828
[273]
J. Bousquet, L.P. Boulet, M.J. Peters, H. Magnussen, J. Quiralte, N.E. Martinez-Aguilar, et al.
Budesonide/formoterol for maintenance and relief in uncontrolled asthma vs. high-dose salmeterol/fluticasone.
Respir Med., 101 (2007), pp. 2437-2446
[274]
P. Kuna, M.J. Peters, A.I. Manjra, C. Jorup, I.P. Naya, N.E. Martínez-Jimenez, et al.
Effect of budesonide/formoterol maintenance and reliever therapy on asthma exacerbations.
Int J Clin Pract., 61 (2007), pp. 725-736
[275]
K.M. Kew, C. Karner, S.M. Mindus, G. Ferrara.
Combination formoterol and budesonide as maintenance and reliever therapy versus combination inhaler maintenance for chronic asthma in adults and children.
Cochrane Database Syst Rev., (2013), pp. CD009019
[276]
A. Papi, M. Corradi, C. Pigeon-Francisco, R. Baronio, Z. Siergiejko, S. Petruzzelli, et al.
Beclometasone-formoterol as maintenance and reliever treatment in patients with asthma: a double-blind, randomised controlled trial.
Lancet Respir Med., 1 (2013), pp. 23-31
[277]
D.M. Sobieraj, E.R. Weeda, E. Nguyen, C.I. Coleman, C.M. White, S.C. Lazarus, et al.
Association of inhaled corticosteroids and long-acting ß-agonists as controller and quick relief therapy with exacerbations and symptom control in persistent asthma: A systematic review and reta-analysis.
JAMA., 319 (2018), pp. 1485-1496
[278]
S.J. Szefler, R.J. Martin, T.S. King, H.A. Boushey, R.M. Cherniack, V.M. Chinchilli, et al.
Significant variability in response to inhaled corticosteroids for persistent asthma.
J Allergy Clin Immunol., 109 (2002), pp. 410-418
[279]
H. Powell, P.G. Gibson.
Inhaled corticosteroid doses in asthma: an evidence-based approach.
Med J Aust., 178 (2003), pp. 223-225
[280]
F.M. Ducharme, M. Ni Chroinin, I. Greenstone, T.J. Lasserson.
Addition of long-acting beta2-agonists to inhaled steroids versus higher dose inhaled steroids in adults and children with persistent asthma.
Cochrane Database Syst Rev., (2010), pp. CD005533
[281]
W.R. Pieters, K.K. Wilson, H.C. Smith, J.J. Tamminga, S. Sondhi.
Salmeterol/fluticasone propionate versus fluticasone propionate plus montelukast: a cost-effective comparison for asthma.
Treat Respir Med., 4 (2005), pp. 129-138
[282]
S. Joos, A. Miksch, J. Szecsenyi, B. Wieseler, U. Grouven, T. Kaiser, et al.
Montelukast as add-on therapy to inhaled corticosteroids in the treatment of mild to moderate asthma: a systematic review.
Thorax., 63 (2008), pp. 453-462
[283]
F.S. Ram, C.J. Cates, F.M. Ducharme.
Long-acting beta2-agonists versus anti-leukotrienes as add-on therapy to inhaled corticosteroids for chronic asthma.
Cochrane Database Syst Rev., (2005), pp. CD003137
[284]
B.F. Chauhan, F.M. Ducharme.
Addition to inhaled corticosteroids of long-acting beta2-agonists versus antileukotrienes for chronic asthma.
Cochrane Database Syst Rev., (2014), pp. CD003137
[285]
B.F. Chauhan, M.M. Jeyaraman, A. Singh, J. Lys, A.M. Abou-Setta, R. Zarychanski, et al.
Addition of anti-leukotriene agents to inhaled corticosteroids for adults and adolescents with persistent asthma.
Cochrane Database Syst Rev., (2017), pp. CD010347
[286]
A. Woodcock, J. Vestbo, N.D. Bakerly, J. New, J.M. Gibson, S. McCorkindale, Salford Lung Study Investigators, et al.
Effectiveness of fluticasone furoate plus vilanterol on asthma control in clinical practice: an open-label, parallel group, randomised controlled trial.
Lancet., 390 (2017 18), pp. 2247-2255
[287]
R.N. Van Zyl-Smit, M. Krüll, C. Gessner, Y. Gon, O. Noga, A. Richard, PALLADIUM trial investigators, et al.
Once-daily mometasone plus indacaterol versus mometasone or twice-daily fluticasone plus salmeterol in patients with inadequately controlled asthma (PALLADIUM): a randomised, double-blind, triple-dummy, controlled phase 3 study.
Lancet Respir Med., 8 (2020), pp. 987-999
[288]
J.C. Virchow, P. Kuna, P. Paggiaro, A. Papi, D. Singh, S. Corre, et al.
Single inhaler extrafine triple therapy in uncontrolled asthma (TRIMARAN and TRIGGER): two double-blind, parallel-group, randomised, controlled phase 3 trials.
Lancet., 394 (2019), pp. 1737-1749
[289]
H.A. Kerstjens, M. Engel, R. Dahl, P. Paggiaro, E. Beck, M. Vandewalker, et al.
Tiotropium in asthma poorly controlled with standard combination therapy.
N Engl J Med., 367 (2012), pp. 1198-1207
[290]
P. Rogliani, B.L. Ritondo, L. Calzetta.
Triple therapy in uncontrolled asthma: a network meta-analysis of Phase III studies.
Eur Respir J., (2021), pp. 2004233
[291]
M. Ni Chroinin, I.R. Greenstone, A. Danish, H. Magdolinos, V. Masse, X. Zhang, et al.
Long-acting beta2-agonists versus placebo in addition to inhaled corticosteroids in children and adults with chronic asthma.
Cochrane Database Syst Rev., (2005), pp. CD005535
[292]
J.H. Toogood, J.C. Baskerville, B. Jennings, N.M. Lefcoe, S.A. Johansson.
Influence of dosing frequency and schedule on the response of chronic asthmatics to the aerosol steroid, budesonide.
J Allergy ClinImmunol., 70 (1982), pp. 288-298
[293]
M. Tonelli, M. Zingoni, E. Bacci, F.L. Dente, A. Di Franco, D. Giannini, et al.
Short-term effect of the addition of leukotriene receptor antagonists to the current therapy in severe asthmatics.
Pulm Pharmacol Ther., 16 (2003), pp. 237-240
[294]
J.C. Virchow Jr., A. Prasse, I. Naya, L. Summerton, A. Harris.
Zafirlukast improves asthma control in patients receiving high-dose inhaled corticosteroids.
Am J RespirCrit Care Med., 162 (2000), pp. 578-585
[295]
D. Singh, J.C. Virchow, G.W. Canonica, A. Vele, M. Kots, G. Georges, A. Papi.
Extrafine triple therapy in patients with asthma and persistent airflow limitation.
Eur Respir J., 56 (2020), pp. 2000476
[296]
H.A.M. Kerstjens, J. Maspero, K.R. Chapman, R.N. van Zyl-Smit, M. Hosoe, A.M. Tanase, IRIDIUM trial investigators, et al.
Once-daily, single-inhaler mometasone-indacaterol-glycopyrronium versus mometasone-indacaterol or twice-daily fluticasone-salmeterol in patients with inadequately controlled asthma (IRIDIUM): a randomised, double-blind, controlled phase 3 study.
Lancet Respir Med., 8 (2020), pp. 1000-1012
[297]
C. Gessner, O. Kornmann, J. Maspero, R. van Zyl-Smit, M. Krüll, A. Salina, et al.
Fixed-dose combination of indacaterol/glycopyrronium/mometasone furoate once-daily versus salmeterol/fluticasone twice-daily plus tiotropium once-daily in patients with uncontrolled asthma: A randomised. Phase IIIb, non-inferiority study (ARGON).
Respir Med., 170 (2020), pp. 106021
[298]
L.H.Y. Kim, C. Saleh, A. Whalen-Browne, P.M. O’Byrne, D.K. Chu.
Triple vs Dual Inhaler Therapy and Asthma Outcomes in Moderate to Severe Asthma: A Systematic Review and Meta-analysis.
JAMA., 325 (2021 Jun 22), pp. 2466-2479
[299]
E. Befekadu, C. Onofrei, G.L. Colice.
Tiotropium in asthma: a systematic review.
J Asthma Allergy., 7 (2014), pp. 11-21
[300]
L.A. Lee, Z. Bailes, N. Barnes, L.P. Boulet, D. Edwards, A. Fowler, et al.
Efficacy and safety of once-daily single-inhaler triple therapy (FF/UMEC/VI) versus FF/VI in patients with inadequately controlled asthma (CAPTAIN): a double-blind, randomised, phase 3A trial.
Lancet Respir Med., 9 (2021 Jan), pp. 69-84
[301]
G.G. Brusselle, C. Vanderstichele, P. Jordens, R. Deman, H. Slabbynck, V. Ringoet, et al.
Azithromycin for prevention of exacerbations in severe asthma (AZISAST): a multicentre randomised double-blind placebo-controlled trial.
Thorax., 68 (2013), pp. 322-329
[302]
E.H. Wong, J.D. Porter, M.R. Edwards, S.L. Johnston.
The role of macrolides in asthma: current evidence and future directions.
Lancet Respir Med., 2 (2014), pp. 657-670
[303]
S.A. Hiles, V.M. McDonald, M. Guilhermino, G.G. Brusselle, P.G. Gibson.
Does maintenance azithromycin reduce asthma exacerbations? An individual participant data meta-analysis.
Eur Respir J, (2019 Sep 12.),
[304]
M. Humbert, R. Beasley, J. Ayres, R. Slavin, J. Hébert, J. Bousquet, et al.
Benefits of omalizumab as add-on therapy in patients with severe persistent asthma who are inadequately controlled despite best available therapy (GINA 2002 step 4 treatment): INNOVATE.
[305]
M. Humbert, W. Berger, G. Rapatz, F. Turk.
Add-on omalizumab improves day-to-day symptoms in inadequately controlled severe persistent allergic asthma.
[306]
W.W. Busse, M. Massanari, F. Kianifard, G.P. Geba.
Effect of omalizumab on the need for rescue systemic corticosteroid treatment in patients with moderate-to-severe persistent IgE-mediated allergic asthma: a pooled analysis.
Curr Med Res Opin., 23 (2007), pp. 2379-2386
[307]
R. Normansell, S. Walker, S.J. Milan, E.H. Walters, P. Nair.
Omalizumab for asthma in adults and children.
Cochrane database of systematc reviews., (2014), pp. CD003559
[308]
I.D. Pavord, S. Korn, P. Howarth, E.R. Bleecker, R. Buhl, O.N. Keene, et al.
Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial.
Lancet., 380 (2012), pp. 651-659
[309]
H. Ortega, G. Chupp, P. Bardin, A. Bourdin, G. Garcia, B. Hartley, et al.
The role of mepolizumab in atopic and nonatopic severe asthma with persistent eosinophilia.
Eur Respir J., 44 (2014), pp. 239-241
[310]
H.G. Ortega, M.C. Liu, I.D. Pavord, G.G. Brusselle, J.M. FitzGerald, A. Chetta, et al.
Mepolizumab treatment in patients with severe eosinophilic asthma.
N Engl J Med., 371 (2014), pp. 1198-1207
[311]
H.G. Ortega, S.W. Yancey, B. Mayer, N.B. Gunsoy, O.N. Keene, E.R. Bleecker, 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., 4 (2016), pp. 549-556
[312]
Y. Liu, S. Zhang, D.W. Li, S.J. Jiang.
Efficacy of anti-interleukin-5 therapy with mepolizumab in patients with asthma: a meta-analysis of randomized placebo-controlled trials.
[313]
E.H. Bel, S.E. Wenzel, P.J. Thompson, C.M. Prazma, O.N. Keene, S.W. Yancey, et al.
Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma.
N Engl J Med., 371 (2014), pp. 1189-1197
[314]
N. Lugogo, C. Domingo, P. Chanez, R. Leigh, M.J. Gilson, R.G. Price, et al.
Long-term efficacy and safety of mepolizumab in patients with severe eosinophilic asthma: A multi-center, open-label, phase IIIb study.
Clin Ther., 38 (2016), pp. 2058-2070.e1
[315]
A. Menzies-Gow, G. Colice, J.M. Griffiths, G. Almqvist, S. Ponnarambil, P. Kaur, G. Ruberto, K. Bowen, A. Hellqvist, M. Mo, E. Garcia Gil.
NAVIGATOR: a phase 3 multicentre, randomized, double-blind, placebo-controlled, parallel-group trial to evaluate the efficacy and safety of tezepelumab in adults and adolescents with severe, uncontrolled asthma.
Respir Res., 21 (2020 Oct 13), pp. 266
[316]
M.E. Wechsler, G. Colice, J.M. Griffiths, G. Almqvist, T. Skarby, T. Piechowiak, et al.
SOURCE: a phase 3, multicentre, randomized, double-blind, placebo-controlled, parallel group trial to evaluate the efficacy and safety of tezepelumab in reducing oral corticosteroid use in adults with oral corticosteroid dependent asthma.
Lancet Respir Med., (2022 Mar 29:),
[317]
A. Torrego, I. Solá, A.M. Muñoz, I. Roqué, M. Figuls, J.J. Yepes-Nuñez, P. Alonso-Coello, et al.
Bronchial thermoplasty for moderate or severe persistent asthma in adults.
Cochrane Database of Systematic Reviews., (2014), pp. D009910
[318]
B. Mash, A. Bheekie, P.W. Jones.
Inhaled vs oral steroids for adults with chronic asthma.
Cochrane Database Syst Rev., (2000), pp. CD002160
[319]
S.W. Clarke, S.P. Newman.
Therapeutic aerosols 2–Drugs available by the inhaled route.
Thorax., 39 (1984), pp. 1-7
[320]
M.B. Dolovich, R. Dhand.
Aerosol drug delivery: developments in device design and clinical use.
Lancet., 377 (2011), pp. 1032-1045
[321]
B.L. Laube, H.M. Janssens, F.H. de Jongh, S.G. Devadason, R. Dhand, P. Diot, et al.
European Respiratory Society; International Society for Aerosols in Medicine. What the pulmonary specialist should know about the new inhalation therapies.
Eur Respir J., 37 (2011), pp. 1308-1331
[322]
Consenso SEPAR-ALAT sobre terapia inhalada..
Arch Bronconeumol, 49 (2013), pp. 1-14
[323]
GEMA Inhaladores. Madrid: Luzán 5; 2018. Disponible en www.gemasma.com.
[324]
J. Sanchis, C. Corrigan, M.L. Levy, J.L. Viejo, ADMIT Group.
Inhaler devices-from theory to practice.
Respir Med., 107 (2013), pp. 495-502
[325]
J. Sanchis, I. Gich, S. Pedersen.
Aerosol Drug Management Improvement Team (ADMIT). Systematic Review of Errors in Inhaler Use: Has Patient Technique Improved Over Time?.
Chest., 150 (2016), pp. 394-406
[326]
V. Plaza, J. Giner, J.G. Rodrigo, M. Dolovich, J. Sanchis.
Errors in the use of inhalers by health care professionals: A Systematic Review.
J Allergy Clin Immunol Pract., 6 (2018), pp. 987-995
[327]
P.H. Brown, A.P. Greening, G.K. Crompton.
Large volume spacer devices and the influence of high dose beclomethasone dipropionate on hypothalamo-pituitary-adrenal axis function.
Thorax., 48 (1993), pp. 233-238
[328]
S.P. Newman, M.T. Newhouse.
Effect of add-on devices for aerosol drug delivery: deposition studies and clinical aspects.
J Aerosol Med., 9 (1996), pp. 55-70
[329]
S.P. Newman.
Spacer devices for metered dose inhalers.
Clin Pharmacokinet., 43 (2004), pp. 349-360
[330]
W. Vincken, M.L. Levy, J. Scullion, O.S. Usmani, P.N.R. Dekhuijzen, C.J. Corrigan.
Spacer devices for inhaled therapy: why use them, and how?.
ERJ Open Res [Internet]., 4 (2018),
[331]
V. Plaza, J. Sanchis.
Medical personnel and patient skill in the use of metered dose inhalers: a multicentric study. CESEA Group.
Respiration., 65 (1998), pp. 195-198
[332]
V. Plaza, J. Giner, M. Calle, P. Rytilä, C. Campo, P. Ribó, et al.
Impact of patient satisfaction with his or her inhaler on adherence and asthma control.
Allergy Asthma Proc., 39 (2018), pp. 437-444
[333]
C. Armour, S. Bosnic-Anticevich, M. Brillant, D. Burton, L. Emmerton, I. Krass, et al.
Pharmacy Asthma Care Program (PACP) improves outcomes for patients in the community.
Thorax., 62 (2007), pp. 496-592
[334]
C.L. Armour, H.K. Reddel, K.S. Lemay, B. Saini, L.D. Smith, S.Z. Bosnic-Anticevich, et al.
Feasibility and Effectiveness of an Evidence-Based Asthma Service in Australian Community Pharmacies: A Pragmatic Cluster Randomized Trial.
Journal of Asthma., 50 (2013), pp. 302-309
[335]
I.A. Basheti, H.K. Reddel, C.L. Armour, S.Z. Bosnic-Anticevich.
Improved asthma outcomes with a simple inhaler technique intervention by community pharmacists.
J Allergy Clin Immunol., 119 (2007), pp. 1537-1538
[336]
V. García-Cárdenas, D. Sabater-Hernández, P. Kenny, F. Martínez-Martínez, M.J. Faus, S.I. Benrimoj.
Effect of a pharmacist intervention on asthma control. A cluster randomised trial.
Respiratory Medicine., 107 (2013), pp. 1346-1355
[337]
V. Giraud, F.-A. Allaert, N. Roche.
Inhaler technique and asthma: Feasability and acceptability of training by pharmacists.
Respir Med., 105 (2011), pp. 1815-1822
[338]
A. Hämmerlein, U. Müller, M. Schulz.
Pharmacist-led intervention study to improve inhalation technique in asthma and COPD patients: Improvement of inhalation technique.
J Evaluation Clinical Practice., 17 (2011), pp. 61-70
[339]
E. Mehuys, L. van Bortel, L. de Bolle, I. van Tongelen, L. Annemans, J.P. Remon, et al.
Effectiveness of pharmacist intervention for asthma control improvement.
Eur Respir J., 31 (2008), pp. 790-799
[340]
L.-Y. Wong, S.-S. Chua, A.-R. Husin, H. Arshad.
A pharmacy management service for adults with asthma: a cluster randomised controlled trial.
Family Practice., 34 (2017), pp. 564-573
[341]
J. Giner, V. Macián, C. Hernández, E.D.E.N. Grupo.
Multicenter prospective study of respiratory patient education and instruction in the use of inhalers (EDEN study).
Arch Bronconeumol., 38 (2002), pp. 300-305
[342]
Y. Takaku, K. Kurashima, C. Ohta, T. Ishiguro, N. Kagiyama, T. Yanagisawa, et al.
How many instructions are required to correct inhalation errors in patients with asthma and chronic obstructive pulmonary disease?.
Respir Med., 123 (2017), pp. 110-115
[343]
I.A. Basheti, N.M. Obeidat, H.K. Reddel.
Effect of novel inhaler technique reminder labels on the retention of inhaler technique skills in asthma: a single-blind randomized controlled trial.
NPJ Prim Care Respir Med., 27 (2017), pp. 9
[344]
J. Van del Palen, J.J. Klein, C.L. van Hervaarden, G.A. Zielhuis, E.R. Seydel.
Multiple inhalers confuse asthma patients.
Eur Respir J., 14 (1999), pp. 1034-1037
[345]
P.N.R. Dekhuijzen, W. Vincken, J.C. Virchow, N. Roche, A. Agusti, F. Lavorini, et al.
Prescription of inhalers in asthma and COPD: Towards a rational, rapid and effective approach.
Respir Med., 107 (2013), pp. 1817-1821
[346]
C. Cabrera López, I. Urrutia-Landa, C.A. Jiménez-Ruiz.
Año SEPAR por la calidad del aire. Papel de la SEPAR en favor del control del cambio climático.
Arch Bronconeumol, 57 (2021), pp. 313-314
[347]
British Thoracic Society. Environment and Lung Health Position Statement 2020. Disponible en: https://www.brit-thoracic.org.uk/about-us/governance-documents-and-policies/position-statements/.
[348]
S.C. Lazarus, V.M. Chinchilli, N.J. Rollings, H.A. Boushey, R. Cherniack, T.J. Craig, et al.
Smoking affects response to inhaled corticosteroids or leukotriene receptor antagonists in asthma.
Am J Respir Crit Care Med., 175 (2007), pp. 783-790
[349]
P. Lange, J. Parner, J. Vestbo, P. Schnohr, G. Jensen.
A 15-year follow-up study of ventilatory function in adults with asthma.
N Engl J Med., 339 (1998), pp. 1194-1200
[350]
A.L. James, L.J. Palmer, E. Kicic, P.S. Maxwell, S.E. Lagan, G.F. Ryan, et al.
Decline in lung function in the Busselton Health Study: the effects of asthma and cigarette smoking.
Am J Respir Crit Care Med., 171 (2005), pp. 109-114
[351]
K.L. Clearie, L. McKinlay, P.A. Williamson, B.J. Lipworth.
Fluticasone/Salmeterol Combination Confers Benefits in People With Asthma Who Smoke.
Chest., 141 (2012), pp. 330-338
[352]
L. Hedman, A. Bjerg, S. Sundberg, B. Forsberg, E. Rönmark.
Both environmental tobacco smoke and personal smoking is related to asthma and wheeze in teenagers.
Thorax., 66 (2011), pp. 20-25
[353]
C.A. Jiménez, M. Barrueco, S. Solano, M. Torrecilla, M. Domínguez, J.L. Díaz-Maroto, et al.
Recomendaciones en el abordaje diagnóstico y terapéutico del tabaquismo. Documento de consenso.
Arch Bronconeumol., 39 (2003), pp. 35-41
[354]
F.D. Martinez, A.L. Wright, L.M. Taussig, C.J. Holberg, M. Halonen, W.J. Morgan.
Asthma and wheezing in the first six years of life. The Group Health Medical Associates.
N Engl J Med., 332 (1995), pp. 133-138
[355]
D. Mackay, S. Haw, J.G. Ayres, C. Fischbacher, J.P. Pell.
Smoke-free legislation and hospitalizations for childhood asthma.
N Engl J Med., 363 (2010), pp. 1139-1145
[356]
M. Sims, R. Maxwell, A. Gilmore.
Short-term impact of the smokefree legislation in England on emergency hospital admissions for asthma among adults: a population-based study.
Thorax., 68 (2013), pp. 619-624
[357]
J.E. Bayly, D. Bernat, L. Porter, K. Choi.
Secondhand Exposure to Aerosols From Electronic nicotine delivery systems and asthma exacerbations among youth with asthma.
Chest., 155 (2018), pp. 88-93
[358]
R. Bals, J. Boyd, S. Esposito, R. Foronjy, P.S. Hiemstra, C.A. Jiménez-Ruiz, et al.
Electronic cigarettes: a task force report from the European Respiratory Society.
[359]
D.C. Christiani.
Vaping-Induced Lung Injury.
N Engl J Med., (2019 Sep 6),
[360]
V. Plaza, J. Serrano, C. Picado, J. Sanchis, High Risk Asthma Research Group.
Frequency and clinical characteristics of rapid-onset fatal and near-fatal asthma.
Eur Respir J., 19 (2002), pp. 846-852
[361]
E. Nizankowska-Mogilnicka, G. Bochenek, L. Mastalerz, M. Swierczynska, C. Picado, G. Scadding, et al.
EAACI/GA2LEN guideline: aspirin provocation tests for diagnosis of aspirin hypersensitivity.
Allergy., 62 (2007), pp. 1111-1118
[362]
A.D. Izquierdo, I. Bobolea, I. Doña, P. Campo, C. Segura, N. Ortega, et al.
Position statement of the Spanish Society of Allergology and Clinical Immunology on provocation tests with aspirin/nonsteroidal anti-inflammatory drugs.
J Investig Allergol Clin Immunol., 30 (2020), pp. 1-13
[363]
W.J. Morgan, E.F. Crain, R.S. Gruchalla, G.T. O’Connor, M. Kattan, R. Evans, et al.
Results of a home-based environmental intervention among urban children with asthma.
N Engl J Med., 351 (2004), pp. 1068-1080
[364]
W. Phipatanakul, B. Cronin, R.A. Wood, P.A. Eggleston, M.C. Shih, L. Song, et al.
Effect of environmental intervention on mouse allergen levels in homes of inner-city Boston children with asthma.
Ann Allergy Asthma Immunol., 92 (2004), pp. 420-425
[365]
T. Shirai, T. Matsui, K. Suzuki, K. Chida.
Effect of pet removal on pet allergic asthma.
Chest., 127 (2005), pp. 1565-1571
[366]
R. Orriols, K. Abu, E. Alday, M.J. Cruz, J.B. Gáldiz, I. Isidro, et al.
Normativa del asma ocupacional.
Arch Bronconeumol., 42 (2006), pp. 457-474
[367]
J. Portnoy, G.L. Chew, W. Phipatanakul, P.B. Williams, C. Grimes, K. Kennedy, et al.
Environmental assessment and exposure reduction of cockroaches: a practice parameter.
J Allergy Clin Immunol., 132 (2013), pp. 802-808
[368]
F.A. Rabito, J.C. Carlson, H. He, D. Werthmann, C. Schal.
A single intervention for cockroach control reduces cockroach exposure and asthma morbidity in children.
Journal of Allergy and Clinical Immunology., 140 (2017), pp. 565-570
[369]
C. Luczynska, E. Tredwell, N. Smeeton, P. Burney.
A randomized controlled trial of mite allergen-impermeable bed covers in adult mite-sensitized asthmatics.
Clin Exp Allergy., 33 (2003), pp. 1648-1653
[370]
A. Woodcock, L. Forster, E. Matthews, J. Martin, L. Letley, M. Vickers, et al.
Medical Research Council General Practice Research Framework. Control of exposure to mite allergen and allergen-impermeable bed covers for adults with asthma.
N Engl J Med, 349 (2003), pp. 225-236
[371]
P.C. Gotzsche, H.K. Johansen.
House dust mite control measures for asthma: systematic review.
Allergy., 63 (2008), pp. 646-659
[372]
C.S. Murray, P. Foden, H. Sumner, E. Shepley, A. Custovic, A. Simpson.
Preventing severe asthma exacerbations in children. A randomized trial of mite-impermeable bed covers.
Am J Respir Crit Care Med., 196 (2017), pp. 150-158
[373]
T. Htut, T.W. Higenbottam, G.W. Gill, R. Darwin, P.B. Anderson, N. Syed.
Eradication of house dust mite from homes of atopic asthmatic subjects: a double-blind trial.
J Allergy Clin Immunol., 107 (2001), pp. 55-60
[374]
S. Halken, A. Host, U. Niklassen, L.G. Hansen, F. Nielsen, S. Pedersen, et al.
Effect of mattress and pillow encasings on children with asthma and house dust mite allergy.
J Allergy ClinImmunol., 111 (2003), pp. 169-176
[375]
B.F. Leas, K.E. D’Anci, A.J. Apter, T. Bryant-Stephens, M.P. Lynch, J.L. Kaczmarek, et al.
Effectiveness of indoor allergen reduction in asthma management: A systematic review.
Journal of Allergy and Clinical Immunology., 141 (2018), pp. 1854-1869
[376]
M.J. Abramson, R.M. Puy, J.M. Weiner.
Injection allergen immunotherapy for asthma.
Cochrane Database of Systematic Reviews., (2010), pp. CD001186
[377]
S. Dhami, A. Kakourou, F. Asamoah, I. Agache, S. Lau, M. Jutel, et al.
Allergen immunotherapy for allergic asthma: a systematic review and meta-analysis.
Allergy., 72 (2017), pp. 1825-1848
[378]
N.F. Adkinson Jr., P.A. Eggleston, D. Eney, E.O. Goldstein, K.C. Schuberth, J.R. Bacon, et al.
A controlled trial of immunotherapy for asthma in allergic children.
N Engl J Med., 336 (1997), pp. 324-331
[379]
C. Pitsios, P. Demoly, M.B. Bilo, R. Gerth van Wijk, O. Pfaar, G.J. Sturm, et al.
Clinical contraindications to allergen immunotherapy: an EAACI position paper.
Allergy., 70 (2015), pp. 897-909
[380]
I. Agache, S. Lau, C.A. Akdis, S. Smolinska, M. Bonini, O. Cavkaytar, et al.
EAACI Guidelines on Allergen Immunotherapy: House dust mite-driven allergic asthma.
Allergy., 74 (2019), pp. 855-873
[381]
D.I. Bernstein, M. Wanner, L. Borish, G.M. Liss, Immunotherapy Committee, American Academy of Allergy, Asthma and Immunology.
Twelve-year survey of fatal reactions to allergen injections and skin testing: 1990-2001.
J Allergy Clin Immunol., 113 (2004), pp. 1129-1136
[382]
J.M. Olaguibel, M.J. Álvarez.
Efficacy of sublingual allergen vaccination for respiratory allergy in children. Conclusions from one meta-analysis.
J Investig Allergol Clin Immunol., 15 (2005), pp. 9-16
[383]
M. Penagos, E. Compalati, F. Tarantini, C.E. Baena-Cagnani, G. Passalacqua, G.W. Canonica.
Efficacy of mometasone furoate nasal spray in the treatment of allergic rhinitis. Meta-analysis of randomized, double-blind, placebo-controlled, clinical trials.
Allergy., 63 (2008), pp. 1280-1291
[384]
S.Y. Lin, N. Erekosima, J.M. Kim, M. Ramanathan, C. Suarez-Cuervo, Y. Chelladurai, et al.
Sublingual immunotherapy for the treatment of allergic rhinoconjunctivitis and asthma: A systematic review.
JAMA., 309 (2013), pp. 1278-1288
[385]
J.C. Virchow, V. Backer, P. Kuna, L. Prieto, H. Nolte, H.H. Villesen, et al.
Efficacy of a house dust mite sublingual allergen immunotherapy tablet in adults with allergic asthma: A randomized clinical trial.
JAMA., 315 (2016), pp. 1715-1725
[386]
S.R. Durham, S.M. Walker, E.M. Varga, M.R. Jacobson, F. O’Brien, W. Noble, et al.
Long-term clinical efficacy of grass-pollen immunotherapy.
N Engl J Med., 341 (1999), pp. 468-475
[387]
L. Jacobsen, B. Niggemann, S. Dreborg, H.A. Ferdousi, S. Halken, A. Høst, The PAT investigator group, et al.
Specific immunotherapy has long-term preventive effect of seasonal and perennial asthma: 10-year follow-up on the PAT study.
[388]
M. Kristiansen, S. Dhami, G. Netuveli, S. Halken, A. Muraro, G. Roberts, et al.
Allergen immunotherapy for the prevention of allergy: A systematic review and meta-analysis.
Pediatr Allergy Immunol., 28 (2017), pp. 18-29
[389]
G.B. Pajno, G. Barberio, F. de Luca, L. Morabito, S. Parmiani.
Prevention of new sensitizations in asthmatic children monosensitized to house dust mite by specific immunotherapy. A six-year follow-up study.
Clin Exp Allergy., 31 (2001), pp. 1392-1397
[390]
S. Nasser, U. Vestenbæk, A. Beriot-Mathiot, P. Poulsen.
Cost-effectiveness of specific immunotherapy with Grazax in allergic rhinitis co-existing with asthma.
Allergy., 63 (2008), pp. 1624-1629
[391]
M. Asaria, S. Dhami, R. van Ree, R. Gerth van Wijk, A. Muraro, G. Roberts, et al.
Health economic analysis of allergen immunotherapy for the management of allergic rhinitis, asthma, food allergy and venom allergy: A systematic overview.
Allergy., 73 (2018), pp. 269-283
[392]
O. Abadoglu, D. Mungan, G. Pasaoglu, G. Celik, Z. Misirligil.
Influenza vaccination in patients with asthma: effect on the frequency of upper respiratory tract infections and exacerbations.
J Asthma., 41 (2004), pp. 279-283
[393]
C. Christy, C.A. Aligne, P. Auinger, T. Pulcino, M. Weitzman.
Effectiveness of influenza vaccine for the prevention of asthma exacerbations.
Arch Dis Child., 89 (2004), pp. 734-735
[394]
A. Sheikh, A. Alves, S. Dhami.
Pneumococcal vaccine for asthma.
Cochrane Database Syst. Rev., (2002), pp. CD002165
[395]
J.A. Castro, K. Abarca, E. Forno.
Asthma and the risk of invasive pneumococcal disease: a meta-analysis.
Pediatrics, 145 (2020), pp. e20191200
[396]
H.S. Izurieta, W.W. Thompson, P. Kramarz, D.K. Shay, R.L. Davis, F. DeStefano, et al.
Influenza and the rates of hospitalization for respiratory disease among infants and young children.
N Eng J Med., 342 (2000), pp. 232-239
[397]
V.K. Jain, L. Rivera, K. Zaman, R.A. Espos Jr., C. Sirivichayakul, B.P. Quiambao, et al.
Vaccine for Prevention of Mild and Moderate-to-Severe Influenza in Children.
N Eng J Med., 369 (2013), pp. 2481-2491
[398]
J. Merckx, F.M. Ducharme, C. Martineau, R. Zemek, J. Gravel, D. Chalut, et al.
Respiratory viruses and treatment failure in children with asthma exacerbation.
Pediattrics., 142 (2018), pp. e20174105
[399]
T.R. Talbot, T.V. Hartert, E. Mitchel, N.B. Halasa, P.G. Arbogast, K.A. Poehling, et al.
Asthma as a risk factor for invasive pneumococcal disease.
N Engl J Med., 352 (2005), pp. 2082-2090
[400]
P. Klemets, O. Lyytikainen, P. Ruutu, J. Ollgren, T. Kaijalainen, M. Leinonen, et al.
Risk of invasive pneumococcal infections among working age adults with asthma.
Thorax., 65 (2010), pp. 698-702
[401]
D.K. Kim, C.B. Bridges, K.H. Harriman, Advisory Committee on Immunization Practices.
Advisory Committee on Immunization Practices Recommended Immunization Schedule for Adults Aged 19 Years or Older: United States, 2016.
Ann Intern Med, 164 (2016), pp. 184-194
[402]
J.J. Picazo, F. González-Romo, A. García, E. Peréz-Trallero, P. Gil, R. de la Cámara, et al.
Consenso sobre la vacunación anti-neumocócica en el adulto con patología de base.
Rev Esp Quimioter., 26 (2013), pp. 232-252
[403]
Grupo de trabajo vacunación frente a neumococo en grupos de riesgo 2015 de la Ponencia de Programas y Registro de Vacunaciones. Utilización de la vacuna frente a neumococo en grupos de riesgo. Comisión de Salud Pública del Consejo Interterritorial del Sistema Nacional de Salud. Ministerio de Sanidad, Servicios Sociales e Igualdad. 2015. Disponible en: http://www.msssi.gob.es/profesionales/saludPublica/prevPro- mocion/vacunaciones/docs/Neumococo_Gruposriesgo.pdf.
[404]
N.W. Johnston, M.R. Sears.
Asthma exacerbations. 1: epidemiology.
Thorax, 61 (2006), pp. 722-728
[405]
D.M. Hughes, M. McLeod, B. Garner, R.B. Goldbloom.
Controlled trial of a home and ambulatory program for asthmatic children.
Pediatrics., 87 (1991), pp. 54-61
[406]
V.T. Colland.
Learning to cope with asthma: a behavioural self-management program for children.
Patient Educ Couns., 22 (1993), pp. 141-152
[407]
J. Van Der Palen, J.J. Klein, G.A. Zielhuis, C.L.A. Van Herwaarden, E.R. Seydel.
Behavioural effect of self-treatment guidelines in a self-management program for adults with asthma.
Patient Educ Couns., 43 (2001), pp. 161-169
[408]
Gibson PG, Powell H, Coughlan J, Wilson AJ, Abramson M, Haywood Bauman A, et al. Educación para el autocuidado y examen médico regular para adultos con asma (Revisión Cochrane traducida). En: La Biblioteca Cochrane Plus, 2008 Número 1. Oxford: Update Software Ltd. Disponible en: http://www.update-software.com.
[409]
Powell H, Gibson PG. Opciones para la educación sobre el autocuidado para los adultos con asma (Revisión Cochrane traducida). En: La Biblioteca Cochrane Plus, 2008 Número 1. Oxford: Update Software Ltd. Disponible en: http://www.update-software.com.
[410]
British Thoracic Society, Scottish Intercollegiate Guidelines Network.
British guideline on the management of asthma.
Thorax., 69 (2014), pp. 1-192
[411]
H. Pinnock, H.L. Parke, M. Panagioti, L. Daines, G. Pearce, E. Epiphaniou, et al.
Systematic meta-review of supported self-management for asthma: A healthcare perspective.
[412]
L. Kuhn, K. Reeves, Y. Taylor, H. Tapp, A. McWilliams, A. Gunter, et al.
Planning for Action: The impact of an asthma action plan decision support tool integrated into an electronic health record (EHR) at a large health care system.
J Am Board Fam Med., 28 (2015), pp. 382-393
[413]
M.R. Partridge.
Patient education.
pp. 378-392
[414]
S. Ahmed, L. Steed, K. Harris, S.J.C. Taylor, H. Pinnock.
Interventions to enhance the adoption of asthma self-management behaviour in the South Asian and African American population: A systematic review.
NPJ Prim Care Respir Med., 28 (2018), pp. 5
[415]
P.G. Gibson, H. Powell, J. Coughlan, A.J. Wilson, M.J. Hensley, M. Abramson, et al.
Limited (information only) patient education programs for adults with asthma.
Cochrane Database Syst Rev., (2002), pp. CD001005
[416]
P.G. Gibson, H. Powell, J. Coughlan, A.J. Wilson, M. Abramson, P. Haywood, et al.
Self-management education and regular practitioner review for adults with asthma.
Cochrane Database Syst Rev., (2003), pp. CD001117
[417]
A.S. Melani, M. Bonavia, V. Cilenti, C. Cinti, M. Lodi, P. Martucci, et al.
Inhaler mishandling remains common in real life and is associated with reduced disease control.
Respir Med., 105 (2011), pp. 930-938
[418]
R.B. Haynes, H. McDonald, A.X. Garg, P. Montague.
Interventions for helping patients to follow prescriptions for medications.
Cochrane Database Syst Rev., (2002), pp. CD000011
[419]
T.L. Creer.
Medication compliance and childhood asthma.
pp. 303-333
[420]
V. Plaza, M. Peiró, M. Torrejón, M. Fletcher, A. López-Viña, J.M. Ignacio, J.A. Quintano, S. Bardagí, I. Gich, PROMETHEUS Study Group.
A repeated short educational intervention improves asthma control and quality of life.
Eur Respir J., 46 (2015), pp. 1298-1307
[421]
M.J. Abramson, M.J. Bailey, F.J. Couper, J.S. Driver, O.H. Drummer, A.B. Forbes, et al.
Are asthma medications and management related to deaths from asthma?.
Am J Respir Crit Care Med., 163 (2001), pp. 12-18
[422]
J. Douglass, R. Aroni, D. Goeman, K. Stewart, S. Sawyer, F. Thien, et al.
A qualitative study of action plans for asthma.
[423]
H.K. Reddel, G.B. Marks, C.R. Jenkins.
When can personal best peak flow be determined for asthma action plans?.
Thorax., 59 (2004), pp. 922-924
[424]
A. Lahdensuo.
Guided self management of asthma-how to do it.
[425]
J. Côté, D.M. Bowie, P. Robichaud, J.G. Parent, L. Battisti, L.P. Boulet.
Evaluation of two different educational interventions for adult patients consulting with an acute asthma exacerbation.
Am J Respir Crit Care Med., 163 (2001), pp. 1415-1419
[426]
P.G. Gibson, H. Powell.
Written action plans for asthma: An evidence-based review of the key components.
[427]
T.L. Gatheral, A. Rushton, D.J. Evans, C.A. Mulvaney, N.R. Halcovitch, G. Whiteley, et al.
Personalised asthma action plans for adults with asthma.
Cochrane Database Syst Rev., (2017), pp. CD011859
[428]
N.A. Gibson, A.E. Ferguson, T.C. Aitchison, J.Y. Paton.
Compliance with inhaled asthma medication in preschool children.
Thorax., 50 (1995), pp. 1274-1279
[429]
A. Bozek, J. Jarzab.
Adherence to asthma therapy in elderly patients.
J Asthma., 47 (2010), pp. 162-165
[430]
Y. Bingham, N. Sanghani, J. Cook, P. HAll, A. Jamalzadeh, Moore, R. Crouch, et al.
Electronic adherence monitoring identifies severe preschool wheezers who are steroid responsive.
Pediatric Pulmonology., 55 (2020), pp. 2254-2260
[431]
C.R. Horn, T.J.H. Clark, G.M. Cochrane.
Compliance with inhaled therapy and morbidity from asthma.
Respir Med., 84 (1990), pp. 67-70
[432]
N.S. Jentzsch, P. Camargos, E.S.C. Sarinho, J. Bousquet.
Adherence rate to beclomethasone dipropionate and the level of asthma control.
Respir Med., 106 (2012), pp. 338-343
[433]
C.S. Rand.
Adherence to asthma therapy in the preschool child.
Allergy., Supplement57 (2002), pp. 48-57
[434]
M. Hyland.
Types of noncompliance.
Eur Respir Rev., 8 (1998), pp. 255-259
[435]
V. Plaza, C. Fernández-Rodríguez, C. Melero, B.G. Cosío, L.M. Entrenas, L.P. de Llano, et al.
Validation of “Test of the Adherence to Inhalers” (TAI) for asthma and COPD patients.
J Aerosol Med Pulm Dug Deliv., 29 (2016), pp. 142-152
[436]
L.P. De Lano, A. Pallares, F.J. González-Barcala, M. Mosteiro-Añón, D. Corbacho, R. Dacal, et al.
Assessing adherence to inhaled medication in asthma: impact of once-daily versus twice-daily dosing frequency.
The ATAUD study. J Asthma., 55 (2018), pp. 933-938
[437]
V. Plaza, J. Giner, E. Curto, M.B. Alonso-Ortiz, M.I. Orue, J.M. Vega, the group of investigators of the RE-TAI study, et al.
Assessing adherence by combining the Test of Adherence to Inhalers with pharmacy prescription records.
J Investig Allergol Clin Immunol., (2019 Oct 10:),
[438]
Y.J. Taylor, H. Tapp, L.E. Shade, T.L. Liu, J.L. Mowrer, M.F. Dulin.
Impact of shared decision making on asthma quality of life and asthma control among children.
J Asthma., 55 (2018), pp. 675-683
[439]
E.L. Toy, N.U. Beaulieu, J.M. McHale, T.R. Welland, C.A. Plauschinat, A. Swensen, M.S. Duh.
Treatment of COPD: Relationships between daily dosing frequency, adherence, resource use, and costs.
Respir Med., 105 (2011), pp. 435-441
[440]
D. Price, A. Robertson, K. Bullen, C. Rand, R. Horne, H. Staudinger.
Improved adherence with once-daily versus twice-daily dosing of mometasona furoate administered via a dry powder inhaler: a randomized open-label study.
BMC Pulm Med., 510 (2010), pp. 1
[441]
L.M. Osman.
Patient preferences and inhaler use in chronic obstructive pulmonary disease.
Int J Resp Care., 2 (2006), pp. 95-99
[442]
L.G. Heaney, J. Busby, P. Bradding, R. Chaudhuri, A.H. Mansur, R. Niven, et al.
Remotely monitored therapy and nitric oxide suppression identifies nonadherence in severe asthma.
Am J Respir Crit Care Med., 199 (2019), pp. 454-464
[443]
I. Urrutia, V. Plaza, S. Pascual, C. Cisneros, L.M. Entrenas, M.T. Luengo, et al.
Asthma control and concordance of opinions between patients and pulmonologists.
J Asthma., 50 (2013), pp. 877-883
[444]
M. Castro, N.A. Zimmermann, S. Crocker, J. Bradley, C. Leven, K.B. Schechtman.
Asthma Intervention Program Prevents Readmissions in High Healthcare Users.
Am J Respir Crit Care Med., 168 (2003), pp. 1095-1099
[445]
A. Borgmeyer, P.M. Gyr, P.A. Jamerson, L.D. Henry.
Evaluation of the role of the pediatric nurse practitioner in an inpatient asthma program.
J Pediatr Health Care., 22 (2008), pp. 273-281
[446]
M.C. Kuethe, A.A. Vaessen-Verberne, R.G. Elbers, W.M. van Aalderen.
Nurse versus physician-led care for the management of asthma.
Cochrane Database Syst Rev., (2013), pp. CD009296
[447]
K.K. Hall, H.L. Petsky, A.B. Chang, K.F. O’Grady.
Caseworker-assigned discharge plans to prevent hospital readmission for acute exacerbations in children with chronic respiratory illness.
Cochrane Database Syst Rev., (2018), pp. CD012315
[448]
L.P. Boulet, M.È. Boulay, G. Gauthier, L. Battisti, V. Chabot, M.F. Beauchesne, et al.
Benefits of an asthma education program provided at primary care sites on asthma outcomes.
Respir Med., 109 (2015), pp. 991-1000
[449]
K.M. Kew, R. Carr, I. Crossingham.
Lay-led and peer support interventions for adolescents with asthma.
Cochrane Database Syst Rev., (2017), pp. CD012331
[450]
K. Harris, D. Kneale, T.J. Lasserson, V.M. McDonald, J. Grigg, J. Thomas.
School-based self-management interventions for asthma in children and adolescents: a mixed methods systematic review.
Cochrane Database Syst Rev., (2019), pp. CD011651
[451]
A.H.Y. Chan, J. Harrison, P.N. Black, E.A. Mitchell, J.M. Foster.
Using electronic monitoring devices to measure inhaler adherence: A practical guide for clinicians.
J Allergy Clin Immunol Pract., 3 (2015), pp. 335-349.e5
[452]
R.W. Morton, H.E. Elphick, A.S. Rigby, W.J. Daw, D.A. King, L.J. Smith, et al.
STAAR: A randomised controlled trial of electronic adherence monitoring with reminder alarms and feedback to improve clinical outcomes for children with asthma.
[453]
J. Halterman, M. Fagnano, R. Tajon, P. Tremblay, H. Wang, A. Butz, et al.
Effect of the School-Based Telemedicine Enhanced Asthma Management (SB-TEAM) Program on Asthma Morbidity: A Randomized Clinical Trial.
JAMA Pediatrics, 172 (2018), pp. e174938
[454]
B. Chongmelaxme, S. Lee, T. Dhippayom, S. Saokaew, N. Chaiyakunapruk, P. Dilokthornsakul.
The Effects of Telemedicine on Asthma Control and Patients’ Quality of Life in Adults: A Systematic Review and Meta-analysis.
J Allergy Clin Immunol Pract, 7 (2019), pp. 199-216.e11
[455]
H. Pinnock, E. Epiphaniou, G. Pearce, H. Parke, T. Greenhalgh, A. Sheikh, et al.
Implementing supported self-management for asthma: a systematic review and suggested hierarchy of evidence of implementation studies.
[456]
G. Cano Fuentes, C. Dastis, I. Morales, M.L. Manzanares, A. Fernández, L. Martín.
Ensayo clínico aleatorio para evaluar la eficacia de una intervención educativa desarrollada en atención primaria sobre asmáticos adultos.
Atencion Primaria., 46 (2014), pp. 117-139
[457]
S. Dokbua, P. Dilokthornsakul, N. Chaiyakunapruk, B. Saini, I. Krass, T. Dhippayom.
Effects of an Asthma Self-Management Support Service Provided by Community Pharmacists: A Systematic Review and Meta-Analysis.
J Manag Care Spec Pharm., 24 (2018), pp. 1184-1196
[458]
A. Manfrin, M. Tinelli, T. Thomas, J. Krska.
A cluster randomised control trial to evaluate the effectiveness and cost-effectiveness of the Italian medicines use review (I-MUR) for asthma patients.
BMC Health Services Research [Internet]., 17 (2017 Dec),
[459]
K.S. LeMay, C.L. Armour, H.K. Reddel.
Performance of a brief asthma control screening tool in community pharmacy: a cross-sectional and prospective longitudinal analysis.
Primary Care Respiratory Journal., 23 (2014), pp. 79-84
[460]
J.F. Van Boven, E.G. Hiddink, A.G. Stuurman-Bieze, C.C. Schuiling-Veninga, M.J. Postma, S. Vegter.
The pharmacists’ potential to provide targets for interventions to optimize pharmacotherapy in patients with asthma.
Int J Clin Pharm., 35 (2013), pp. 1075-1082
[461]
H.K. Reddel, D.R. Taylor, E.D. Bateman, L.-P. Boulet, H.A. Boushey, W.W. Busse, on behalf of the American Thoracic Society/European Respiratory Society Task Force on Asthma Control, Exacerbations, et al.
An Official American Thoracic Society/European Respiratory Society Statement: Asthma Control and Exacerbations. Standardizing Endpoints for Clinical Asthma Trials and Clinical Practice.
Am J Respir Crit Care Med., 180 (2009), pp. 59-99
[462]
P.G. Woodruff, S.D. Emond, A.K. Singh, C.A. Camargo Jr..
Sudden-onset severe acute asthma: clinical features and response to therapy.
Acad Emerg Med., 5 (1998), pp. 695-701
[463]
V. Plaza, J. Serrano, C. Picado, J. Sanchis, High Risk Asthma Research Group.
Frequency and clinical characteristics of rapid-onset fatal and near-fatal asthma.
Eur Respir J., 19 (2002), pp. 846-852
[464]
M.O. Turner, K. Noertjojo, S. Vedal, T. Bai, S. Crump, J.M. Fitzgerald.
Risk factors for near-fatal asthma: a case-control study in hospitalized patients with asthma.
Am J Respir Crit Care Med., 157 (1998), pp. 1804-1809
[465]
I. Mitchell, S.C. Tough, L.K. Semple, F.H. Green, P.A. Hessel.
Near-fatal asthma: a population-based study of risk factors.
Chest., 121 (2002), pp. 1407-1413
[466]
J. Serrano, V. Plaza, B. Sureda, J. de Pablo, C. Picado, S. Bardagi, et al.
Alexithymia: a relevant psychological variable in near-fatal asthma.
Eur Respir J., 28 (2006), pp. 296-302
[467]
G.J. Rodrigo, V. Plaza, S. Bardagí, J.A. Castro-Rodríguez, A. de Diego, S. Liñán, et al.
Guía ALERTA 2. América Latina y España: Recomendaciones para la prevención y el Tratamiento de la exacerbación Asmática.
Arch Bronconeumol, 46 (2010), pp. s2-s20
[468]
E.R. McFadden, R. Kisser, W.J. De Groot.
Acute bronchial Asthma: relations between clinical and physiological manifestations.
N Engl J Med, 288 (1973), pp. 221-225
[469]
D.H. Arnold, T. Gebretsadik, P.A. Minton, S. Higgins, T.V. Hartert.
Clinical measures associated with FEV1 in persons with asthma requiring hospital admission.
Am J Emerg Med., 25 (2007), pp. 425-429
[470]
Scottish Intercollegiate Guidelines Network, British Thoracic Society. British guideline on the management of asthma. A national clinical guideline. 2019. Disponible en: www.sign.ac.uk.
[471]
E. Neville, H. Gribbin, B.D. Harrison.
Acute severe asthma.
Respir Med, 85 (1991), pp. 463-474
[472]
B. Brenner, M.S. Kohn.
The acute asthmatic patient in the ED: to admit or discharge.
Am J Emerg Med., 16 (1998), pp. 69-75
[473]
D. Carruthers, B.D. Harrison.
Arterial blood gas analysis or oxygen saturation in the assessment of acute asthma?.
Thorax., 50 (1995), pp. 186-188
[474]
C.S. White, R.P. Cole, H.W. Lubetsky, J.H. Austin.
Acute asthma. Admission chest radiography in hospitalized adult patients.
Chest., 100 (1991), pp. 14-16
[475]
M.G. Roback, D.A. Dreitlein.
Chest radiograph in the evaluation of first time wheezing episodes: review of current clinical practice and efficacy.
Pediatr Emerg Care., 14 (1998), pp. 181-184
[476]
G.J. Rodrigo, C. Rodrigo, J.B. Hall.
Acute Asthma in adults. A review.
Chest., 125 (2004), pp. 1081-1082
[477]
P.J. Honkoop, D.R. Taylor, A.D. Smith, J.B. Snoeck-Stroband, J.K. Sont.
Early detection of asthma exacerbations by using action points in self-management plans.
Eur Respir J., 41 (2013), pp. 53-59
[478]
C. Reisner, A. Kotch, G. Dworkin.
Continuous versus frequent intermittent nebulization of albuterol in acute asthma: a randomized, prospective study.
Ann Allergy Asthma Immunol., 75 (1995), pp. 41-47
[479]
C.J. Cates, J.A. Crilly, B.H. Rowe.
Holding chambers (spacers) versus nebulisers for beta-agonist treatment of acute asthma.
Cochrane Database of Systematic Reviews., (2006), pp. CD000052
[480]
B.H. Rowe, C. Spooner, F.M. Ducharme, J.A. Bretzlaff, G.W. Bota.
Early emergency department treatment of acute asthma with systemic corticosteroids.
Cochrane Database of Systematic Reviews., (2001), pp. CD002178
[481]
R. Manser, D. Reid, M. Abramson.
Corticosteroids for acute severe asthma in hospitalised patients.
Cochrane Database Syst Rev., (2000), pp. CD001740
[482]
B.H. Rowe, C.H. Spooner, F.M. Ducharme, G.W. Bota.
Corticosteroids for preventing relapse following acute exacerbations of asthma.
Cochrane Database of Systematic Reviews., (2007), pp. CD000195
[483]
T. Hasegawa, K. Ishihara, S. Takakura, H. Fujii, T. Nishimura, M. Okazaki, et al.
Duration of systemic corticosteroids in the treatment exacerbation; a randomized study.
Intern Med., 39 (2000), pp. 794-797
[484]
B.D. Harrison, T.C. Stokes, G.J. Hart, D.A. Vaughan, N.J. Ali, A.A. Robinson.
Need for intravenous hydrocortisone in addition to oral prednisolone in patients admitted to hospital with severe asthma without ventilatory failure.
[485]
D. Ratto, C. Alfaro, J. Sipsey, M.M. Glovsky, O.P. Sharma.
Are intravenous corticosteroids required in status asthmaticus?.
JAMA., 260 (1988), pp. 527-529
[486]
M. Lahn, P. Bijur, E.J. Gallagher.
Randomized clinical trial of intramuscular vs oral methylprednisolone in the treatment of asthma exacerbations following discharge from an emergency department.
Chest., 126 (2004), pp. 362-368
[487]
L.M. Osman, C. Calder, D.J. Godden, J.A. Friend, L. McKenzie, J.S. Legge, et al.
A randomised trial of self-management planning for adult patients admitted to hospital with acute asthma.
Thorax., 57 (2002), pp. 869-874
[488]
S. Tapp, T.J. Lasserson, B.H. Rowe.
Education interventions for adults who attend the emergency room for acute asthma.
Cochrane Database of Systematic Reviews., (2007), pp. CD003000
[489]
G.J. Rodrigo, M. Rodriguez-Verde, V. Peregalli, C. Rodrigo.
Effects of short-term 28% and 100% oxygen in paCO2 and peak expiratory flow rate in acute Asthma. A randomized trial.
Chest., 124 (2003), pp. 1312-1317
[490]
K. Perrin, M. Wijesinghe, B. Healy, K. Wadsworth, R. Bowditch, S. Bibby, et al.
Randomised controlled trial of high concentration versus titrated oxygen therapy in severe exacerbations of asthma.
[491]
J. Corbo, P. Bijur, M. Lahn, E.J. Gallagher.
Concordance between capnography and arterial blood gas measurements of carbon dioxide in acute asthma.
Ann Emerg Med., 46 (2005), pp. 323-327
[492]
C.J. Cates, E.J. Welsh, B.H. Rowe.
Holding chambers (spacers) versus nebulisers for beta-agonist treatment of acute asthma.
Cochrane Database of Systematic Reviews., (2013), pp. CD000052
[493]
J.R. Turner, K.J. Corkery, D. Eckman, A.M. Gelb, A. Lipavsky, D. Sheppard.
Equivalence of continuous flow nebulizer and metered-dose inhaler with reservoir bag for treatment of acute airflow obstruction.
[494]
G.A. Salzman, M.T. Steele, J.P. Pribble, R.M. Elenbaas, D.R. Pyszczynski.
Aerosolized metaproterenol in the treatment of asthmatics with severe airflow obstruction. Comparison of two delivery methods.
Chest., 95 (1989), pp. 1017-1020
[495]
A.H. Idris, M.F. McDermott, J.C. Raucci, A. Morrabel, S. McGorray, L. Hendeles.
Emergency department treatment of severe asthma. Metered-dose inhaler plus holding chamber is equivalent in effectiveness to nebulizer.
[496]
K.B. Newman, S. Milne, C. Hamilton, K. Hall.
A comparison of albuterol administered by metered-dose inhaler and spacer with albuterol by nebulizer in adults presenting to an urban emergency department with acute asthma.
[497]
G.J. Rodrigo, C. Rodrigo.
Continuous vs intermittent beta-agonists in the treatment of acute adult asthma: a systematic review with meta-analysis.
Chest., 122 (2002), pp. 160-165
[498]
C.A. Camargo, C.H. Spooner, B.H. Rowe.
Continuous versus intermittent beta-agonists for acute asthma.
Cochrane Database of Systematic Reviews., (2003), pp. CD001115
[499]
A.H. Travers, S.J. Milan, A.P. Jones, C.A. Camargo Jr., B.H. Rowe.
Addition of intravenous beta2-agonists to inhaled beta2-agonists for acute asthma.
Cochrane Database of Systematic Reviews., (2012), pp. CD010179
[500]
F.E. Simons, X. Gu, K.J. Simons.
Epinephrine absorption in adults: intramuscular versus subcutaneous injection.
J Allergy Clin Immunol., 108 (2001), pp. 871-873
[501]
F.E.R. Simons, J.R. Roberts, X. Gu, K.J. Simons.
Epinephrine absorption in children with a history of anaphylaxis.
J Allergy Clin Immunol., 101 (1998), pp. 33-37
[502]
S.G.A. Brown.
The pathophysiology of shock in anaphylaxis.
Immunol Allergy Clin North Am., 27 (2007), pp. 165-175
[503]
G.J. Rodrigo, L.J. Nannini.
Comparison between nebulized adrenaline and beta2 agonists for the treatment of acute asthma. A meta-analysis of randomized trials.
Am J Emerg Med., 24 (2006), pp. 217-222
[504]
J. Soar, G.D. Perkins, G. Abbas, A. Alfonzo, A. Barelli, J.J.L.M. Bierens, et al.
European Resuscitation Council Guidelines for Resuscitation 2010 Section 8. Cardiac arrest in special circumstances: electrolyte abnormalities, poisoning, drowning, accidental hypothermia, hyperthermia, asthma, anaphylaxis, cardiac surgery, trauma, pregnancy, electrocution.
Resuscitation., 81 (2010), pp. 1400-1433
[505]
T. Vanden Hoek, L. Morrison, M. Shuster, M. Donnino, E. Sinz, E. Lavonas, et al.
Part 12: Cardiac arrest in special situations: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care.
Circ J Am Hear Assoc., 122 (2010), pp. S829-S861
[506]
R. Manser, D. Reid, M.J. Abramson.
Corticosteroids for acute severe asthma in hospitalised patients.
Cochrane Database of Systematic Reviews., (2001), pp. CD001740
[507]
G.J. Rodrigo, J.A. Castro-Rodriguez.
Anticholinergics in the treatment of children and adults with acute asthma: a systematic review with meta-analysis.
Thorax, 60 (2005), pp. 740-746
[508]
M.L. Edmonds, S.J. Milan, C.A. Camargo Jr., C.V. Pollack, B.H. Rowe.
Early use of inhaled corticosteroids in the emergency department treatment of acute asthma.
Cochrane Database Syst Rev., (2012), pp. CD002308
[509]
R. Nowak, C.H. Emerman, J.P. Hanrahanc, M.V. Parsey, N.A. Hanania, R. Claus, et al.
A comparison of levalbuterol with racemic albuterol in the treatment of acute severe asthma exacerbations in adults.
Am J Emerg Med., 24 (2006), pp. 259-267
[510]
F.A. Lederle, R.E. Pluhar, A.M. Joseph, D.E. Niewoehner.
Tapering of corticosteroid therapy following exacerbation of asthma. A randomized, double-blind, placebo-controlled trial.
Arch Intern Med., 147 (1987), pp. 2201-2203
[511]
M.Q. Hatton, A.S. Vathenen, M.J. Allen, S. Davies, N.J. Cooke.
A comparison of “abruptly stopping” with “tailing off” oral corticosteroids in acute asthma.
Respir Med., 89 (1995), pp. 101-104
[512]
P. Nair, S.J. Milan, B.H. Rowe.
Addition of intravenous aminophylline to inhaled beta2-agonists in adults with acute asthma.
Cochrane Database of Systematic Reviews., (2012), pp. CD002742
[513]
B.H. Rowe, J. Bretzlaff, C. Bourdon, G. Bota, S. Blitz, C.A. Camargo.
Magnesium sulfate for treating exacerbations of acute asthma in the emergency department.
Cochrane Database of Systematic Reviews., (2000), pp. CD001490
[514]
J.M. FitzGerald.
Magnesium sulfate is effective for severe acute asthma treated in the emergency department.
West J Med., 172 (2000), pp. 96
[515]
M.C. Gallegos-Solórzano, R. Pérez-Padilla, R.J. Hernández-Zenteno.
Usefulness of inhaled magnesium sulfate in the coadjuvant management of severe asthma crisis in an emergency department.
Pulm Pharmacol Ther., 23 (2010), pp. 432-437
[516]
K.M. Kew, L. Kirtchuk, C.I. Michell.
Intravenous magnesium sulfate for treating adults with acute asthma in the emergency department.
Cochrane Database Syst Rev., (2014 May 28), pp. CD010909
[517]
R. Knightly, S.J. Milan, R. Hughes, J.A. Knopp-Sihota, B.H. Rowe, R. Normansell, et al.
Inhaled magnesium sulfate in the treatment of acute asthma.
Cochrane Database Syst Rev., 11 (2017 Nov 28), pp. CD003898
[518]
G. Rodrigo, C. Pollack, C. Rodrigo, B.H. Rowe.
Heliox for nonintubated acute asthma patients.
Cochrane Database of Systematic Reviews., (2006), pp. CD002884
[519]
C.L. Colebourn, V. Barber, J.D. Young.
Use of helium-oxygen mixture in adult patients presenting with exacerbations of asthma and chronic obstructive pulmonary disease: a systematic review.
Anaesthesia., 62 (2007), pp. 34-42
[520]
G.J. Rodrigo, J.A. Castro-Rodriguez.
Heliox-driven b2-agonists nebulization for children and adults with acute asthma: a systematic review with meta-analysis.
Ann Allergy Asthma Immunol., 112 (2014), pp. 29-34
[521]
M. Pallin, M.T. Naughton.
Noninvasive ventilation in acute asthma.
J Crit Care., 29 (2014), pp. 586-593
[522]
N. Arrotta, J. Hill, C. Villa-Roel, E. Dennett, M. Harries, B.H. Rowe.
Factors associated with hospital admission in adult patients with asthma exacerbations: A systematic review.
[523]
K. Hasegawa, A.F. Sullivan, Y. Tsugawa, S.J. Turner, S. Massaro, S. Clark, et al.
Comparison of US emergency department acute asthma care quality: 1997-2001 and 2011-2012.
J Allergy Clin Immunol., 135 (2015), pp. 73-80
[524]
B.H. Rowe, C. Villa-Roel, R.B. Abu-Laban, R. Stenstrom, D. Mackey, I.G. Stiell, et al.
Admissions to Canadian hospitals for acute asthma: a prospective, multicentre study.
Can Respir J., 17 (2010), pp. 25-30
[525]
G. Rodrigo, C. Rodrigo.
Early prediction of poor response in acute asthma patients in the emergency department.
Chest., 114 (1998), pp. 1016-1021
[526]
M.M. Wilson, R.S. Irwin, A.E. Connolly, C. Linden, M.M. Manno.
A prospective evaluation of the 1-hour decision point for admission versus discharge in acute asthma.
J Intensive Care Med., 18 (2003), pp. 275-285
[527]
A.M. Kelly, D. Kerr, C. Powell.
Is severity assessment after one hour of treatment better for predicting the need for admission in acute asthma?.
Respir Med., 98 (2004), pp. 777-781
[528]
P. Piñera-Salmerón, F.J. Álvarez-Gutiérrez, J. Domínguez-Ortega, C. Álvarez, M. Blanco-Aparicio, I. Dávila, et al.
Recomendaciones de derivación del paciente adulto con crisis de asma desde el servicio de Urgencias.
Emergencias., 32 (2020), pp. 258-268
[529]
G.J. Rodrigo, V. Plaza, S.B. Forns, J.A. Castro-Rodriguez, A. de Diego, S.L. Cortes, et al.
[ALERTA 2 guidelines. Latin America and Spain: recommendations for the prevention and treatment of asmatic exacerbations. Spanish Pulmonology and Thoracic Surgery Society (SEPAR). Asthma Department of the Latinamerican Thoracic Association (ALAT)].
Arch Bronconeumol., 46 (2010), pp. 2-20
[530]
M.G. Pearson, I. Ryland, B.D. Harrison.
National audit of acute severe asthma in adults admitted to hospital. Standards of Care Committee, British Thoracic Society.
Qual Health Care., 4 (1995), pp. 24-30
[531]
P. Piñera, J. Delgado, J. Dominguez, M. Labrador, F.J. Alvarez, E. Martinez, et al.
Management of asthma in the emergency department: a consensus statement.
Emergencias., 30 (2018), pp. 268-277
[532]
C.A. Camargo Jr., G. Rachelefsky, M. Schatz.
Managing asthma exacerbations in the emergency department: summary of the National Asthma Education And Prevention Program Expert Panel Report 3 guidelines for the management of asthma exacerbations.
Proc Am Thorac Soc, 6 (2009), pp. 357-366
[533]
H. Reddel, S. Ware, G. Marks, C. Salome, C. Jenkins, A. Woolcock.
Differences between asthma exacerbations and poor asthma control.
Lancet., 353 (1999), pp. 364-369
[534]
R. Brisk, L.G. Heaney.
Asthma control and exacerbations: two different sides of the same coin.
Curr Opin Pulm Med., 22 (2016), pp. 32-37
[535]
J.A. Castillo Laita, J. de Benito, A. Escribano, M. Fernández, S. García, Grupo de trabajo para el Consenso sobre Tratamiento del Asma Infantil.
Consenso sobre tratamiento del asma en pediatría.
An Pediatr (Barc)., 67 (2007), pp. 253-273
[536]
K. Harris, D. Kneale, T.J. Lasserson, V.M. McDonald, J. Grigg, J. Thomas.
School-based self-management interventions for asthma in children and adolescents: a mixed methods systematic review.
Cochrane Database Syst Rev., 28 (2019), pp. CD011651
[537]
H. Pinnock, H.L. Parke, M. Panagioti, L. Daines, G. Pearce, E. Epiphaniou, et al.
Systematic meta-review of supported self-management for asthma: a health care perspective.
[538]
L.M. Osman, C. Calder.
Implementing asthma education programmes in paediatric respiratory care: setting, timing, people and evaluation.
Paed Respir Rev., 5 (2004), pp. 140-146
[539]
J. Korta, J. Valverde, M. Praena, J. Figuerola, C.R. Rodríguez, S. Rueda, et al.
La educación terapéutica en el asma.
An Pediatr (Barc)., 66 (2007), pp. 496-517
[540]
J.K. Giese.
Evidence-based pediatric asthma interventions and outcome measures in a healthy homes program: An integrative review.
J Asthma., 56 (2019), pp. 662-673
[541]
N. Saxby, S. Beggs, M. Battersby, S. Lawn.
What are the components of effective chronic condition self-management education interventions for children with asthma, cystic fibrosis, and diabetes?. A systematic review.
Patient Educ Couns., 102 (2019), pp. 607-622
[542]
R.L. Zemek, S.K. Bhogal, F.M. Ducharme.
Systematic review of randomized controlled trials examining written action plans in children: what is the plan?.
Arch Pediatr Adolesc Med., 162 (2008), pp. 157-163
[543]
C. Gillette, N. Rockich-Winston, M. Shepherd, S. Flesher.
Children with asthma and their caregivers help improve written asthma action plans: A pilot mixed-method study.
J Asthma., 55 (2018), pp. 609-614
[544]
GINA 2019. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention NHL-BI/WHO Workshop Report. Disponible en http://www.ginasthma.com.
[545]
Cortés Rico O, Rodríguez C, Castillo JA; Grupo de Vías Respiratorias. Normas de Calidad para el tratamiento de la Crisis de Asma en el niño y adolescente. Documentos técnicos del GVR (publicación DT-GVR-1; 2015). Disponible en: http://www.respirar.org/index.php/grupo-vias-respiratorias/protocolos.
[546]
S. Bhogal, R.L. Zemek, F. Ducharme.
Written action plans for asthma in children (Cochrane Review).
Cochrane Database Syst Rev., (2006), pp. CD005306
[547]
A. Ohlmann.
Peak Flow versus Symptom Monitoring to Manage Childhood Asthma.
Kaleidoscope., 5 (2015), pp. 7
[548]
H.L. Yoos, H. Kitzman, A. McMullen, C. Henderson, K. Sidora.
Symptom monitoring in childhood asthma: a randomized clinical trial comparing peak expiratory flow rate with symptom monitoring.
Ann Allergy Asthma Immunol., 88 (2002), pp. 283-291
[549]
N.P. Adams, J.C. Bestall, T.J. Lasserson, P.W. Jones, C. Cates.
Fluticasone versus placebo for chronic asthma in adults and children.
Cochrane Database Syst Rev., (2005), pp. CD003135
[550]
S. Kaiser, T. Huynh, L.B. Bacharier, J.L. Rosenthal, L.A. Bakel, P.C. Parkin, et al.
Preventing exacerbations in preschoolers with recurrent wheeze: A meta-analysis.
Pediatrics., 137 (2016), pp. e20154496
[551]
J.A. Castro-Rodríguez, S. Pedersen.
The role of inhaled corticosteroids in management of asthma in infants and preschoolers.
Curr Opin Pulm Med., 19 (2013), pp. 54-59
[552]
A.M. Fitzpatrick, D.J. Jackson, D.T. Mauger, S.J. Boehmer, W. Phipatanakul, W.J. Sheehan, et al.
Individualized therapy for persistent ashtma in young children.
J Allergy Clin Immunol., 138 (2016), pp. 1608-1618.e12
[553]
A.M. Teper, C.D. Kofman, G.A. Szulman, S.M. Vidaurreta, A.F. Maffey.
Fluticasone improves pulmonary function in children under 2 years old with risk factors for asthma.
Am J Respir Crit Care Med., 171 (2005), pp. 587-590
[554]
M. McKean, F. Ducharme.
Inhaled steroids for episodic viral wheeze of childhood.
Cochrane Database Syst Rev., (2000), pp. CD001107
[555]
P. Brand, D. Caudri, E. Eber, E.A. Gaillard, L. García-Marcos, G. Hedlin, et al.
Classification and pharmacological treatment of preschool wheezing: changes since 2008.
Eur Respir J., 43 (2014), pp. 1172-1177
[556]
T.W. Guilbert, W.J. Morgan, R.S. Zeiger, D.T. Mauger, S.J. Boehmer, S.J. Szefler, et al.
Long-term inhaled corticosteroids in preschool children at high risk for asthma.
N Engl J Med., 354 (2006), pp. 1985-1997
[557]
H. Bisgaard, M.N. Hermansen, L. Loland, L.B. Halkjaer, F. Buchvald.
Intermitent inhaled corticosteroids in infants with epidodic wheezing.
N Engl J Med., 354 (2006), pp. 1998-2005
[558]
L.B. Bacharier, B.R. Phillips, R.S. Zeiger, S.J. Szefler, F.D. Martínez, R.E. Lemanske Jr., CARE Network.
Episodic use of an inhaled corticosteroid or leukotriene receptor antagonist in preeschool children with moderate-to-severe intermittent wheezing.
J Allergy Clin Immunol., 122 (2008), pp. 1127-1135
[559]
F.M. Ducharme, S.D. Dell, D. Radhakrishnan, R.M. Grad, W.T. Watson, C.L. Yang, et al.
Diagnosis and management of asthma in preschoolers: A Canadian Thoracic Society and Canadian Paediatric Society position paper.
Paediatric Child Health., 20 (2015), pp. 353-371
[560]
B.F. Chauhan, C. Chartrand, F.M. Ducharme.
Intermittent versus daily inhaled corticosteroids for persistent asthma in children and adults.
Cochrane Database of Systematic Reviews., (2013), pp. CD009611
[561]
F.M. Ducharme, C. Lemire, F.J. Noya, G.M. Davis, N. Alos, H. Leblond, et al.
Preemptive use of high-dose fluticasone for virus induced wheezing in young children.
N Engl J Med., 360 (2009), pp. 339-353
[562]
R.S. Zeiger, D. Mauger, L.B. Bacharier, T.W. Guilbert, F.D. Martínez, R.F. Lemanske Jr., et al.
CARE Network of the National Heart. Lung and Blood Institute. Daily or intermittent budesonide in preschool children with recurrent wheezing.
N Engl J Med., 365 (2011), pp. 1990-2001
[563]
R.M. Fernandes, A. Wingert, B. Vandermeer, R. Featherstone, S. Ali, A.C. Plint, et al.
Safety of corticosteroids in young children with acute respiratory conditions: a systematic review and meta-analysis.
BMJ Open., 9 (2019), pp. e028511
[564]
Expert Panel Working Group of the National Heart, Lung, Blood Institute(NHLBI) administered, coordinated National Asthma Education, Prevention Program Coordinating Committee(NAEPPCC).
2020 Focused Updates to the Asthma Management Guidelines: A report from the National Asthma Education and Prevention Program Coordinating Committee Expert Panel Working Group.
J Allergy Clin Immunol., 146 (2020), pp. 1217-1270
[565]
H.W. Kelly, A.L. Sternberg, R. Lescher, A.L. Fuhlbrigge, P. Williams, R.S. Zeiger, CAMP Research Group, et al.
Effect of inhaled glucocorticosteroids in childhood on adult height.
N Engl J Med., 367 (2012), pp. 904-912
[566]
L. Zhang, S.O. Prietsch, F.M. Ducharme.
Inhaled corticosteroids in children with persistent ashtma effects of growth.
Cochrane Database Syst Rev, (2014), pp. CD009471
[567]
P.T. Daley-Yates.
Inhaled corticosteroids: potency, dose equivalence and therapeutic index.
Br J Clin Pharmacol., 80 (2015), pp. 372-380
[568]
M. Brodlie, A. Gupta, C.E. Rodríguez-Martínez, J.A. Castro-Rodríguez, F.M. Ducharme, M.C. McKean.
Leukotriene receptor antagonists as maintenance and intermittent therapy for episodic viral wheeze in children.
Paediatr Respir Rev., 17 (2016), pp. 57-59
[569]
J.A. Castro-Rodriguez, A. Beckhaus, E. Forno.
Efficacy of oral corticosteroids in the treatment of acute wheezing episodis in asthmatic preschoolers: systematic review with meta-analysis.
Pediatr Pulmonol, 51 (2016), pp. 868-876
[570]
H.R. Hussein, A. Gupta, S. Broughton, G. Ruiz, N. Brathwaite, C.J. Bossley.
A meta-analysis of montelukast for recurrent wheeze in preschool children.
Eur J Pediatr., 176 (2017), pp. 963-969
[571]
B.F. Chauhan, R. Ben Salah, F.M. Ducharme.
Addition of anti-leukotriene agents to inhaled corticosteroids in children with persistent asthma.
Cochrane Database of Systematic Reviews., (2013), pp. CD009585
[572]
M. Ni Chroinin, I.R. Greenstone, A. Danish, H. Magdolinos, V. Masse, X. Zhang, et al.
Long-acting beta-2-agonists versus placebo in addition to inhaled corticosteroids in children and adults with chronic asthma.
Cochrane Database Syst Rev., (2005), pp. CD005535
[573]
G.J. Rodrigo, V.P. Moral, L.G. Marcos, J.A. Castro-Rodríguez.
Safety of regular use of long-acting beta agonists as monotherapy or added to inhaled corticosteroids in asthma. A systematic review.
Pulm Pharmacol Ther., 22 (2009), pp. 9-19
[574]
H. Bisgaard, P. Le Roux, D. Bjâmer, A. Dymek, J.H. Vermeulen, C. Hultquist.
Budesonide/Formoterol Maintenance Plus Reliever Therapy. A new strategy in pediatric asthma.
Chest., 130 (2006), pp. 1733-1743
[575]
D.M. Soberaj, E.R. Weeda, E. Nguyen, C.I. Coleman, C.M. White, S.C. Lazarus, et al.
Association of inhaled corticosteroids and long-acting beta-agonists as controller and quick refief therapy with exacerbations and symptom control in persistent asthma: A systematic review and meta-analysis.
JAMA., 319 (2018), pp. 1485-1496
[576]
A.A. Vaessen-Verbene, N.J. van der Berg, J.c. van Nierop, H.J. Brackel, G.P. Gerrits, W.C. Hop, et al.
Combination therapy salmeterol/fluticasone versus doubling dose of fluticasone children with asthma.
Am J Respir Crit Care Med., 182 (2010), pp. 1221-1227
[577]
B.F. Chauhan, F.M. Ducharme.
Addition to inhaled corticosteroids of long-acting beta-2-agonists versus anti-leukotrienes for chronic asthma.
Cochrane Database Syst Rev, (2014), pp. CD003137
[578]
G.J. Rodrigo, H. Neffen.
Efficacy and safety of tiotropium in school-age children with moderate-to-severe symptomatic asthma: A systematic review.
Pediatr Allergy Immunol., 28 (2017), pp. 573-578
[579]
E.L.J.E. Vrijandt, G. El Azzi, M. Vandewalker, N. Rupp, T. Harper, L. Graham, et al.
Safety and efficacy of tiotropium in children aged 1-5 years with persistent asthmatic symptoms: a randomised, double-blind, placebo-controlled trial.
Lancet Respiratory Medicine., 6 (2018), pp. 127-137
[580]
A. Bush, A.M. Fitzpatrick, S. Saglani, W.C. Anderson, S.J. Szefler.
Difficult to treat asthma management in school age children.
J Allergy Clin Immunol Pract., 10 (2022), pp. 359-375
[581]
H. Ahmed, S. Turner.
Severe Asthma in children-a review of definitions, epidemiology and treatment options in 2019.
Pediatr Pulmonol, 54 (2019), pp. 778-787
[582]
M.J. Abramson, R.M. Puy, J.M. Weiner.
Injection allergen immunotherapy for asthma.
Cochrane Database Syst Rev., (2010), pp. CD001186
[583]
A. Fiocchi, A.T. Fox.
Preventing progression of allergic rhinitis: the role of specific immunotherapy.
Arch Dis Child Educ Pract Ed., 96 (2011), pp. 91-100
[584]
M. Kristiansen, S. Dhami, G. Netuveli.
Allergen immunotherapy for the prevention of allergy: A systematic review and meta-analysis.
Pediatr Allergy Immunol., 28 (2017), pp. 18-29
[585]
F. Ducharme, F. di Salvio.
Antileukotriene agents compared to inhaled corticosteroids in the management of recurrent and or chronic asthma in adults and children.
Cochrane Database Syst Rev., 2012 (2012 May 16), pp. CD002314
[586]
J. Bekhof, R. Reimink, P.L. Brand.
Systematic review: insufficient validation of clinical scores for the assessment of acute dyspnoea in wheezing children.
Paediatr Respir Rev., 15 (2014), pp. 98-112
[587]
H. Eggink, P. Brand, R. Reimink, J. Bekhof.
Clinical Scores for Dyspnoea Severity in Children: A Prospective Validation Study.
PLoS One., 11 (2016 6), pp. e0157724
[588]
S.R. Smith, J.D. Baty, D. Hodge.
Validation of the pulmonary score. An Asthma severity score for children.
Acad Emerg Med., 9 (2002), pp. 99-104
[589]
C.F. Robertson, F. Smith, R. Beck, H. Levison.
Response to frequent low doses of nebulized salbutamol in acute asthma.
J Pediatr., 106 (1985), pp. 672-674
[590]
S. Tapp, T.J. Lasserson, B.H. Rowe.
Education interventions for adults who attend the emergency room for acute asthma.
Cochrane Database Syst Rev., (2007), pp. CD003000
[591]
J.A. Castro-Rodríguez, G.J. Rodrigo.
Beta-agonist through metered-dose inhaler with valved holding chamber versus nebulizer for acute exacerbation of wheezing or asthma in children under 5 years of age. A systematic review with meta-analysis.
J Ped., 145 (2004), pp. 172-177
[592]
J. Deerojanawong, W. Manuyakorn, N. Prapphal, C. Harnruthakorn, S. Sritippayawan, R. Samransamruajkit.
Randomized controllled trial of salbutamol aerosol therapy via metered dose inhaler-spacer vs jet nebulizer in young children with wheezing.
Pediatr Pulmonol., 39 (2005), pp. 466-472
[593]
N. Mitselou, G. Hedlin.
Spacers versus nebulizers in treatment of acute asthma-a prospective randomized study in preschool children.
J Asthma., 53 (2016), pp. 1059-1062
[594]
G.J. Rodrigo, V. Plaza, S. Bardagí, J.A. Castro-Rodríguez, A. de Diego, S. Liñán, et al.
Guía ALERTA 2. América Latina y España: Recomendaciones para la prevencióny el Tratamiento de la exacerbación Asmática.
Arch Bronconeumol, 46 (2010), pp. s2-s20
[595]
R. Iramain, J.A. Castro-Rodriguez, A. Jara, M. Cardozo, N. Bogado, R. Morinigo, et al.
Salbutamol and ipratropium by inhalar is superior to nebulizer in children with severe acute asthma exacerbation: Randomized clinical trial.
Pediatr Pulmonol., 54 (2019), pp. 372-377
[596]
C.J. Cates, E.J. Welsh, B.H. Rowe.
Holding chambers (spacers) versus nebulisers for beta-agonist treatment of acute asthma.
Cochrane Database Syst Rev., (2013), pp. CD000052
[597]
H. Khine, S.M. Fuchs, A.L. Saville.
Continuous vs intermittent nebulized albuterol for emergency management of asthma.
Acad Emerg Med., 3 (1996), pp. 1019-1024
[598]
B. Griffits, F.M. Ducharme.
Combined inhaled anticholinergics and short-acting beta2-agonists for initial treatment of acute asthma in children.
Cochrane Database Syst Rev., (2013), pp. CD000060
[599]
K. Vézina, B.F. Chauhan, F.M. Ducharme.
Inhaled anticholinergics and short-acting beta2-agonists versus short-acting beta2-agonists alone for children with acute asthma in hospital.
Cochrane Database of Syst Rev., (2014), pp. CD010283
[600]
M.L. Everard, A. Bara, M. Kurian, T.M. Elliot, F. Ducharme, V. Mayowe.
Anticholinergic drugs for wheeze in children under the age of two years.
Cochrane Database Syst Rev, (2005), pp. CD001279
[601]
P. Brand, D. Caudri, E. Eber, E.A. Gaillard, L. García-Marcos, G. Hedlin, et al.
Classification and pharmacological treatment of preschool wheezing: changes since 2008.
Eur Respir J., 43 (2014), pp. 1172-1177
[602]
J. Panickar, M. Lakhanpaul, P.C. Lambert, P. Kenia, T. Stephenson, A. Smyth, et al.
Oral Prednisolone for Preschool Children with Acute Virus-induced wheezing.
N Engl J Med, 360 (2009), pp. 329-338
[603]
B.H. Rowe, C.H. Spooner, F.M. Ducharme, G.W. Bota.
Corticosteroids for preventing relapse following acute exacerbations of asthma.
Cochrane Database Syst Rev., (2007), pp. CD000195
[604]
J.M. Becker, A. Arora, R.J. Scarfone, N.D. Spector, M.E. Fontana-Penn, E. Gracely, et al.
Oral versus intravenous corticosteroids in children hospitalised with asthma.
J Allergy Clin Immunol., 103 (1999), pp. 586-590
[605]
P.L. Barnett, G.L. Caputo, M. Bassin, N. Kuppermann.
Intravenous versus oral corticosteroids in the management of acute asthma in children.
Ann Emerg Med., 29 (1997), pp. 212-217
[606]
T. Hasegawa, K. Ishihara, S. Takakura, H. Fujii, T. Nishimura, M. Okazaki, et al.
Duration of systemic corticosteroids in the treatment exacerbation; a randomized study.
Intern Med., 39 (2000), pp. 794-797
[607]
S. Kayani, D.C. Shannon.
Adverse behavioral effects of treatment for acute exacerbation of asthma in children: a comparison of two doses of oral steroids.
Chest., 122 (2002), pp. 624-628
[608]
J.L. Mathew.
Oral Dexamethasone versus Oral Prednisolone in Acute Asthma: A New Randomized Controlled Trial and Updated Meta-analysis: Evidence-based Medicine Viewpoint.
Indian Pediatr, 55 (2018), pp. 155-159
[609]
N. Paniagua, R. Lopez, N. Muñoz, M. Tames, E. Mojica, E. Arana-Arri, et al.
Randomized trial of dexamethasone versus prednisone for children with acute asthma exacerbations.
J Pediatr., 191 (2017), pp. 190-196
[610]
J.J. Cronin, S. McCoy, U. Kennedy, S.N. An Fhailí, A. Wakai, Hayden, et al.
A Randomized Trial of Single-Dose Oral Dexamethasone Versus Multidose Prednisolone for Acute Exacerbations of Asthma in Children Who Attend the Emergency Department.
Ann Emerg Med, 67 (2016), pp. 593-601
[611]
G.E. Keeney, M.P. Gray, A.K. Morrison, M.N. Levas, E.A. Kessler, G.D. HILL, et al.
Dexamethasone for Acute Asthma Exacerbations inChildren: A Meta-analysis.
Pediatrics., 133 (2014), pp. 493-499
[612]
D.J. Jackson, L.B. Bacharier, D.T. Mauger, S. Boehmer, A. Beigelman, J.F. Chmiel, et al.
National Heart, Lung, and Blood Institute AsthmaNet. Quintupling inhaled glucocorticoids to prevent childhood asthma exacerbations.
NEJM., 378 (2018), pp. 891-901
[613]
A.A. Beckhaus, M.C. Riutor, J.A. Castro Rodriguez.
Inhaled versus systemic corticosteroids for acute ashma in children. A systematic review.
Pediatr Pulmonol., 49 (2014), pp. 326-334
[614]
N. Kearns, I. Maijers, J. Harper, R. Beasley, M. Weatherall.
Inhaled corticosteroids in acute asthma: systemic review and meta-analysis.
J Allergy Clin Immunol Pract., 8 (2020), pp. 605-617
[615]
C.E. Rodriguez-Martinez, M.P. Sossa-Briceño, J.A. Castro-Rodriguez.
Advantage of inhaled corticosteroids as additional therapyto systemic corticosteroids for pediatric acute asthma exacerbations:a cost-effectiveness analysis.
J Asthma., 17 (2019), pp. 1-10
[616]
D.K. Cheuk, T.C. Chau, S.L. Lee.
A meta-analysis on intravenous magnesium sulphate for treating acute asthma.
Arch Dis Child., 90 (2005), pp. 74-77
[617]
B. Griffiths, K.M. Kew.
Intravenous magnesium sulfate for treating children with acute asthma in the emergency department.
Cochrane Database Syst Rev., (2016), pp. CD011050
[618]
S. Schuh, J. Sweeney, M. Rumantir, A.L. Coates, A.R. Willan, D. Stephens, et al.
Effect of Nebulized Magnesium vs Placebo Added to Albuterol on Hospitalization Among Children With Refractory Acute Asthma Treated in the Emergency Department: A Randomized Clinical Trial.
JAMA., 324 (2020 Nov 24), pp. 2038-2047
[619]
R. Knightly, S.J. Milan, R. Hughes, J.A. Knopp-Sihota, B.H. Rowe, R. Normansell, et al.
Inhaled magnesium sulfate in the treatment of acute asthma (Review).
Cochrane Database Syst Rev., (2017), pp. CD003898
[620]
S. Schuh, J. Sweeney, S.B. Freedman, A.L. Coates, D.W. Johnson, G. Thompson, et al.
Magnesium nebulization utilization in management of pediatric asthma (MagNUM PA) trial: study protocol for a randomized controlled trial.
Trials., 17 (2016), pp. 261
[621]
G.C. Geelhoed, L.I. Landau, P.N. Le Souef.
Evaluation of SaO2 as a predictor of outcome in 280 children presenting with acute asthma.
Ann Emerg Med., 23 (1994), pp. 1236-1241
[622]
K. Perrin, M. Wijesinghe, B. Healy, K. Wadsworth, R. Bowditch, S. Bibby, et al.
Randomised controlled trial of high concentration versus tritated oxygen therapy in severe exacerbations of asthma.
Thorax., 66 (2011), pp. 937-941
[623]
Y. Ballestero, J. de Pedro, N. Portillo, O. Martínez-Múgica, E. Arana-Arri, J. Benito.
Pilot Clinical Trial of High-Flow Oxygen Therapy in Children with Asthma in the Emergency Service.
J Pediatr., 194 (2018), pp. 204-210
[624]
F. González Martínez, M.I. González, B. Toledo, J. Pérez, M. Medina, C. Rodríguez, et al.
Treatment with high-flow oxygen therapy in asthma exacerbations in a paediatric hospital ward: Experience from 2012 to 2016.
An Pediatr (Barc)., 90 (2019), pp. 72-78
[625]
J. Pilar, V. Modesto i Alapont, Y.M. López-Fernández, López-Macías, D. García-Urabayen, I. Amores-Hernández.
High-flow nasal cannula therapy versus non-invasive ventilation in children with severe acute asthma exacerbation: An observational cohort study.
Med Intensiva., 41 (2017), pp. 418-424
[626]
S.K. Korang, J. Feinberg, J. Wetterslev, J.C. Jakobsen.
Non-invasive positive pressure ventilation for acute asthma in children (Review).
Cochrane Database Syst Rev., (2016), pp. CD012067
[627]
J. Bousquet, N. Khaltaev, A.A. Cruz, J. Denburg, W.J. Fokkens, A. Togias, et al.
Allergic Rhinitis and Its Impact on Asthma (ARIA) 2008 Update (in Collaboration with the World Health Organization, GA(2)LEN and AllerGen).
[628]
A.M. Navarro, C. Colás, E. Antón, J. Conde, I. Dávila, M.T. Dordal, et al.
Epidemiology of allergic rhinitis in allergy consultations in Spain: Alergológica-2005.
J Investig Allergol Clin Immunol., 19 (2009), pp. 7-13
[629]
A. Valero, M. Ferrer, E. Baró, J. Sastre, A.M. Navarro, E. Martí-Guadaño, et al.
Discrimination between Moderate and Severe Disease May Be Used in Patients with Either Treated or Untreated Allergic Rhinitis.
Allergy., 65 (2010), pp. 1609-1613
[630]
R.R. Orlandi, T.T. Kingdom, P.H. Hwang, T.L. Smith, J.A. Alt, P.M. Baroody, et al.
International Consensus Statement on Allergy and Rhinology: Rhinosinusitis.
Int Forum Allergy Rhinol, 6 (2016), pp. S22-S209
[631]
D. Jarvis, R. Newson, J. Lotvall, D. Hastan, T. Tomassen, T. Keil, et al.
Asthma in Adults and Its Association with Chronic Rhinosinusitis: The GA2LEN Survey in Europe.
Allergy., 67 (2012), pp. 91-98
[632]
D.H. Pols, J.B. Wartna, E.I. van Alphen, H. Moed, N. Rasenberg, P.J.E. Bindels, et al.
Interrelationships between Atopic Disorders in Children: A Meta-Analysis Based on ISAAC Questionnaires.
PLoS One., 10 (2015), pp. e0131869
[633]
P.W. Hellings, L. Klimek, C. Cingi, I. Agache, C. Akdis, C. Bachert, et al.
Non-Allergic Rhinitis: Position Paper of the European Academy of Allergy and Clinical Immunology.
Allergy., 72 (2017), pp. 1657-1665
[634]
P. Ojeda, M.D. Ibáñez, J.M. Olaguibel, J. Sastre, T. Chivato, investigators participating in the National Survey of the Spanish Society of Allergology and Clinical Immunology Alergológica 2015.
Alergólogica 2015: A National Survey on Allergic Diseases in the Spanish Pediatric Population.
J Investig Allergol Clin Immunol., 28 (2018), pp. 321-329
[635]
P. Ojeda, J. Sastre, J.M. Olaguibel, T. Chivato, and investigators participating in the National Survey of the Spanish Society of Allergology and Clinical Immunology Alergológica 2015.
“Alergólogica 2015: A National Survey on Allergic Diseases in the Adult Spanish Population.
J Investig Allergol Clin Immunol., 28 (2018), pp. 151-164
[636]
B. Björkstén, C. Tadd, P. Ellwood, A. Stewart, D. Strachan, ISAAC Phase, III, Study Group.
Worldwide Time Trends for Symptoms of Rhinitis and Conjunctivitis: Phase III of the International Study of Asthma and Allergies in Childhood.
Pediatr Allergy Immunol, 19 (2008), pp. 110-124
[637]
C. Colás, M. Brosa, E. Antón, J. Montoro, A. Navarro, T. Dordal, et al.
Estimate of the Total Costs of Allergic Rhinitis in Specialized Care Based on Real-World Data: The FERIN Study.
Allergy., 72 (2017), pp. 959-966
[638]
S.K. Wise, S.Y. Lin, E. Toskala, R.R. Orlandi, C.A. Akdis, J.A. Alt, et al.
International Consensus Statement on Allergy and Rhinology: Allergic Rhinitis.
Inte Forum of Allergy Rhinol, 8 (2018), pp. 108-352
[639]
K. Gendo, E.B. Larson.
Evidence-Based Diagnostic Strategies for Evaluating Suspected Allergic Rhinitis.
Ann Intern Med, 140 (2004), pp. 278-289
[640]
P. Supakthanasiri, J. Klaewsongkram, H. Chantaphakul.
Reactivity of Allergy Skin Test in Healthy Volunteers.
Singapore Medical Journal., 55 (2014), pp. 34-36
[641]
M.T. Dordal, M. Lluch-Bernal, M.C. Sánchez, C. Rondón, A. Navarro, J. Montoro, et al.
Allergen-specific nasal provocation testing: review by the rhinoconjunctivitis committee of the Spanish Society of Allergy and Clinical Immunology.
J Investig Allergol Clin Immunol., 21 (2011), pp. 1-12
[642]
I. Agache, M. Bilò, G.J. Braunstahl, L. Delgado, P. Demoly, P. Eigenmann, et al.
In Vivo Diagnosis of Allergic Diseases-Allergen Provocation Tests.
Allergy, 70 (2015), pp. 355-365
[643]
C. Rondon, P. Campo, I. Eguiluz-Gracia, C. Plaza, G. Bogas, P.C. Galindo, et al.
Local Allergic Rhinitis Is an Independent Rhinitis Phenotype: The Results of a 10-Year Follow-up Study.
Allergy., 73 (2018), pp. 470-478
[644]
A. Valero, A.M. Navarro, A. del Cuvillo, I. Alobid, J.R. Benito, C. Colás, et al.
Position Paper on Nasal Obstruction: Evaluation and Treatment.
J Investig Allergol Clin Immunol, 28 (2018), pp. 67-90
[645]
E. Heffler, M. Landi, C. Caruso, S. Fichera, F. Gani, G. Guida, et al.
Nasal Cytology: Methodology with Application to Clinical Practice and Research.
Clin Exp Allergy, 48 (2018), pp. 1092-1106
[646]
T. Hummel, K.L. Whitcroft, K. Andrews, A. Altundag, C. Cinghi, R.M. Costanzo, et al.
Position Paper on Olfactory Dysfunction.
Rhinology, Supp54 (2017), pp. 1-30
[647]
International Rhinitis Management Group.
International Consensus Report on the Diagnosis and Management of Rhinitis.
Allergy, 49 (1994), pp. 1-34
[648]
V. Bauchau, S.R. Durham.
Epidemiological Characterization of the Intermittent and Persistent Types of Allergic Rhinitis.
[649]
A. Valero, S. Quirce, I. Dávila, J. Delgado, J. Domínguez-Ortega.
Allergic respiratory disease: Different allergens, different symptoms.
Allergy., 72 (2017), pp. 1306-1316
[650]
J. Montoro, A. del Cuvillo, J. Mullol, X. Molina, J. Bartra, I. Dávila, et al.
Validation of the Modified Allergic Rhinitis and Its Impact on Asthma (ARIA) Severity Classification in Allergic Rhinitis Children: The PEDRIAL Study.
Allergy., 11 (2012), pp. 1437-1442
[651]
A. Valero, M. Ferrer, J. Sastre, A.M. Navarro, L. Monclús, E. Martí-Guadaño, et al.
A new criterion by which to discriminate between patients with moderate allergic rhinitis and patients with severe allergic rhinitis based on the Allergic Rhinitis and its Impact on Asthma severity items.
J Allergy Clin Immunol., 120 (2007), pp. 359-365
[652]
A. Del Cuvillo, V. Santos, J. Montoro, J. Bartra, I. Davila, M. Ferrer, et al.
Allergic rhinitis severity can be assessed using a visual analogue scale in mild, moderate and severe.
Rhinology., 55 (2017), pp. 34-38
[653]
A. Del Cuvillo, J. Sastre, C. Colás, A.M. Navarro, J. Mullol, A. Valero.
Adaptation to Spanish and Validation of the Rhinitis Control Assessment Test (RCAT) Questionnaire.
J Investig Allergol Clin Immunol., (2019 May 28),
[654]
D. Caimmi, N. Baiz, L.K. Tanno, P. Demoly, S. Arnavielhe, R. Murray, et al.
Validation of the MASK-Rhinitis Visual Analogue Scale on Smartphone Screens to Assess Allergic Rhinitis Control.
Clin Exp Allergy., 47 (2017), pp. 1526-1533
[655]
R. Shaaban, M. Zureik, D. Soussan, C. Neukirch, J. Heinrich, J. Sunyer, et al.
Rhinitis and onset of asthma: a longitudinal population-based study.
Lancet., 372 (2008), pp. 1049-1057
[656]
C. Rondon, G. Bogas, E. Barrionuevo, M. Blanca, M.J. Torres, P. Campo.
Nonallergic rhinitis and lower airway disease.
Allergy., 72 (2017), pp. 24-34
[657]
B.I. Nwaru, S. Suzuki, L. Ekerljung, S. Sjölander, R. Mincheva, E.P. Rönmark, et al.
Furry Animal Allergen Component Sensitization and Clinical Outcomes in Adult Asthma and Rhinitis.
J Allergy Clin Immunol Pract, 7 (2019), pp. 1230-1238
[658]
A. Valero, C. Pereira, C. Loureiro, C. Martínez-Cócera, C. Murio, P. Rico, et al.
Interrelationship between skin sensitization, rhinitis, and asthma in patients with allergic rhinitis: a study of Spain and Portugal.
J Investig Allergol Clin Immunol., 19 (2009), pp. 167-172
[659]
A.M. Navarro, A. Valero, B. Julia, S. Quirze.
Coexistence of asthma and allergic rhinitis in adult patients attending clínics: ONEAIR Study.
J Investig Allergol Clin Immunol., 18 (2008), pp. 233-238
[660]
F. Amat, A. Vial, B. Pereira, I. Petit, A. Labbe, J. Just.
Predicting the long-term course of asthma in wheezing infants is still a challenge.
ISRN Allergy., 27 (2011), pp. 493624
[661]
V. Siroux, N. Ballardini, M. Soler, C. Lupinek, A. Boudier, I. Pin, et al.
The asthma-rhinitis multimorbidity is associated with IgE polysensitization in adolescents and adults.
Allergy., 73 (2018), pp. 1447-1458
[662]
L. Cui, J. Yin.
Association of serum specific IgE levels with asthma in autumn pollen-induced allergic rhinitis: A retrospective analysis.
J Asthma., 56 (2019), pp. 505-511
[663]
S. Toppila-Salmi, S. Chanoine, J. Karjalainen, J. Pekkanen, J. Bousquet, V. Siroux.
Risk of adult-onset asthma increases with the number of allergic multimorbidities and decreases with age.
Allergy., 74 (2019), pp. 2406-2416
[664]
V. Siroux, A. Boudier, R. Nadif, C. Lupinek, R. Valenta, J. Bousquet.
Association between asthma, rhinitis, and conjunctivitis multimorbidities with molecular IgE sensitization in adults.
Allergy., 74 (2019), pp. 824-827
[665]
B. Leynaert, C. Neukirch, S. Kony, A. Guénégou, J. Bousquet, M. Aubier, et al.
Association between asthma and rhinitis according to atopic sensitization in a population-based study.
J Allergy Clin Immunol., 113 (2004), pp. 86-93
[666]
J.A. Castillo, J. Mullol.
Comorbilidad de rinitis y asma en España (estudio RINAIR).
Arch Bronconeumol., 44 (2008), pp. 597-603
[667]
A. Arnedo-Pena, L. García-Marcos, G. García, I. Aguinagua, C. González, M. Morales, et al.
Time trends and geographical variations in the prevalence of symptoms of allergic rhinitis in 6-7-year-old children from eight areas of Spain according to the ISAAC.
An Pediatría Barc Spain., 62 (2005), pp. 229-236
[668]
A. Magnan, J.P. Meunier, C. Saugnac, J. Gasteau, F. Neukirch.
Frequency and impact of allergic rhinitis in asthma patients in everyday general medical practice: a French observational cross-sectional study.
[669]
E.P. De Groot, A. Nijkamp, E.J. Duiverman, P.L.P. Brand.
Allergic rhinitis is associated with poor asthma control in children with asthma.
[670]
E. Valovirta, R. Pawankar.
Survey on the impact of comorbid allergic rhinitis in patients with asthma.
BMC Pulm Med., 6 (2006), pp. S3
[671]
S. Gaugris, V. Sazonov-Kocevar, M. Thomas.
Burden of concomitant allergic rhinitis in adults with asthma.
[672]
L. Calus, N. van Bruaene, C. Bosteels, S. Dejonckheere, T. van Zele, G. Holtappels, et al.
Twelve-year follow-up study after endoscopic sinus surgery in patients with chronic rhinosinusitis with nasal polyposis.
Clin Transl Allergy., 9 (2019), pp. 30
[673]
M.E. Boulay, L.P. Boulet.
Lower airway inflammatory responses to repeated very-low-dose allergen challenge in allergic rhinitis and asthma.
Clin Exp Allergy., 32 (2002), pp. 1441-1447
[674]
M. Gaga, P. Lambrou, N. Papageorgiou, N.G. Koulouris, E. Kosmas, S. Fragakis, et al.
Eosinophils are a feature of upper and lower airway pathology in non-atopic asthma, irrespective of the presence of rhinitis.
Clin Exp Allergy., 30 (2000), pp. 663-669
[675]
A. Kessel.
The impact of intranasal corticosteroids on lung function in children with allergic rhinitis.
Pediatr Pulmonol., 49 (2014), pp. 932-937
[676]
S. Lohia, R.J. Schlosser, Z.M. Soler.
Impact of intranasal corticosteroids on asthma outcomes in allergic rhinitis: a meta-analysis.
Allergy., 68 (2013), pp. 569-579
[677]
A. Oka, K. Matsunaga, T. Kamei, Y. Sakamoto, T. Hirano, A. Hayata, et al.
Ongoing allergic rhinitis impairs asthma control by enhancing the lower airway inflammation.
J Allergy Clin Immunol Pract., 2 (2014), pp. 172-178
[678]
M. Deliu, D. Belgrave, A. Simpson, C.S. Murray, G. Kerry, A. Custovic.
Impact of rhinitis on asthma severity in school-age children.
Allergy., 69 (2014), pp. 1515-1521
[679]
C.L. Yu, W.T. Huang, C.M. Wang.
Treatment of allergic rhinitis reduces acute asthma exacerbation risk among asthmatic children aged 2-18 years.
J Microbiol Immunol Infect., (2018 Oct 25),
[680]
F.C. Hampel, A.J. Pedinoff, R.L. Jacobs, C.F. Caracta, S.K. Tantry.
Olopatadine-mometasone combination nasal spray: Evaluation of efficacy and safety in patients with seasonal allergic rhinitis.
Allergy Asthma Proc., 40 (2019 Jul 3), pp. 261-272
[681]
N. Segall, B. Prenner, W. Lumry, C.F. Caracta, S.K. Tantry.
Long-term safety and efficacy of olopatadine-mometasone combination nasal spray in patients with perennial allergic rhinitis.
Allergy Asthma Proc., 40 (2019 Sep 1), pp. 301-310
[682]
C.P. Andrews, D. Mohar, Y. Salhi, S.K. Tantry.
Efficacy and safety of twice-daily and once-daily olopatadine-mometasone combination nasal spray for seasonal allergic rhinitis.
Ann Allergy Asthma Immunol., 124 (2020), pp. 171-178.e2
[683]
G. Roberts, M. Xatzipsalti, L.M. Borrego, A. Custovic, S. Halken, P.W. Hellings, et al.
Paediatric Rhinitis: Position Paper of the European Academy of Allergy and Clinical Immunology.
Allergy., 68 (2013), pp. 1102-1116
[684]
A. Izquierdo-Domínguez, A.L. Valero, J. Mullol.
Comparative Analysis of Allergic Rhinitis in Children and Adults.
Curr Allergy Asthma Rep, 13 (2013), pp. 142-151
[685]
A. Izquierdo-Dominguez, I. Jauregui, A. del Cuvillo, J. Montoro, I. Davila, J. Sastre, et al.
“Allergy Rhinitis: Similarities and Differences between Children and Adults.
Rhinology., 55 (2017), pp. 326-331
[686]
W.J. Fokkens, V.J. Lund, J. Mullol, C. Bachert, I. Alobid, F. Baroody, et al.
EPOS 2012: European position paper on rhinosinusitis and nasal polyps 2012. A summary for otorhinolaryngologists.
Rhinology., 50 (2012), pp. 1-12
[687]
G. De Greve, P.W. Hellings, W.J. Fokkens, B. Pugin, B. Steelant, S.F. Seys.
Endotype-driven treatment in chronic upper airway diseases.
Clin Transl Allergy., 7 (2017), pp. 22
[688]
D. Hastan, W.J. Fokkens, C. Bachert, R.B. Newson, J. Bislimovska, A. Bockelbrink, et al.
Chronic rhinosinusitis in Europe--an underestimated disease. A GA2LEN study.
Allergy., 66 (2011), pp. 1216-1223
[689]
J. Staikuniene, S. Vaitkus, L.M. Japertiene, S. Ryskiene.
Association of chronic rhinosinusitis with nasal polyps and asthma: clinical and radiological features, allergy and inflammation markers.
Med Kaunas Lith., 44 (2008), pp. 257-265
[690]
J.P. Rajan, N.E. Wineinger, D.D. Stevenson, A.A. White.
Prevalence of aspirin-exacerbated respiratory disease among asthmatic patients: A meta-analysis of the literature.
J. Allergy Clin Immunol., 135 (2015), pp. 676-681
[691]
I. Alobid, E. Antón, M. Armengot, J. Chao, C. Colás, A. del Cuvillo, et al.
Rhinoconjunctivitis Committee, Spanish Society of Allergy and Clinical Immunology, Rhinology and Allergy Commission, Spanish Society of Otorhinolaryngology. SEAIC-SEORL. Consensus Document on Nasal Polyposis. POLINA Project.
J Investig Allergol Clin Immunol, 21 (2011), pp. 1-58
[692]
A.M.H. Wuister, N.A. Goto, E.J. Oostveen, W.U. de Jong, E.S. van der Valk, N.M. Kaper, et al.
Nasal endoscopy is recommended for diagnosing adults with chronic rhinosinusitis.
Otolaryngol Head Neck Surg., 150 (2014), pp. 359-364
[693]
A.M.M. Shahizon, A. Suraya, Z. Rozmnan, A.A. Aini, B.S. Gendeh.
Correlation of computed tomography and nasal endoscopic findings in chronic rhinosinusitis.
Med J Malaysia., 63 (2008), pp. 211-215
[694]
Alobid Alobid I, Castillo Vizuete JA, Colás Sanz C (coords.). Guía POLINA. Documento de consenso sobre rinosinusitis crónica con poliposis nasal. Madrid: Sociedad Española de Otorrinolaringología y Cirugía de Cabeza y Cuello; 2023.
[695]
C.B. Small, J. Hernandez, A. Reyes, E. Schenkel, A. Damiano, P. Stryszak, H. Staudinger, M. Danzig.
Efficacy and safety of mometasone furoate nasal spray in nasal polyposis.
J Allergy Clin Immunol., 116 (2005), pp. 1275-1281
[696]
P. Stjärne, K. Blomgren, P. Cayé-Thomasen, S. Salo, T. Søderstrøm.
The efficacy and safety of once-daily mometasone furoate nasal spray in nasal polyposis: a randomized, double-blind, placebo-controlled study.
Acta Otolaryngol., 126 (2006), pp. 606-612
[697]
L.Y. Chong, K. Head, C. Hopkins, C. Philpott, M.J. Burton, A.G.M. Schilder.
Different types of intranasal steroids for chronic rhinosinusitis.
Cochr Database Syst Rev., (2016), pp. CD011993
[698]
Y. Zhang, C. Wang, Y. Huang, H. Lou, L. Zhang.
Efficacy of Short-Term Systemic Corticosteroid Therapy in Chronic Rhinosinusitis With Nasal Polyps: A Meta-Analysis of Randomized Controlled Trials and Systematic Review.
Am J Rhinol Allergy., 33 (2019), pp. 567-576
[699]
R.M. Leung, K. Dinnie, T.L. Smith.
When do the risks of repeated courses of corticosteroids exceed the risks of surgery?.
Int. Forum Allergy Rhinol., 4 (2014), pp. 871-876
[700]
J. Rimmer, W. Fokkens, L.Y. Chong, C. Hopkins.
Surgical versus medical interventions for chronic rhinosinusitis with nasal polyps.
Cochr Database Syst. Rev., (2014), pp. CD006991
[701]
R. Vashishta, Z.M. Soler, S.A. Nguyen, R.J. Schlosser.
A systematic review and meta-analysis of asthma outcomes following endoscopic sinus surgery for chronic rhinosinusitis.
Int Forum Allergy Rhinol., 3 (2013), pp. 788-794
[702]
C. Philpott, C. Hopkins, S. Erskine, N. Kumar, A. Robertson, A. Farboud, et al.
The burden of revision sinonasal surgery in the UK-data from the Chronic Rhinosinusitis Epidemiology Study (CRES): a cross-sectional study.
BMJ Open, 5 (2015), pp. e006680
[703]
A. Ehnhage, P. Olsson, K.G. Kölbeck, M. Skedinger, P. Stjärne, NAFS Study Group.
One year after endoscopic sinus surgery in polyposis: asthma, olfaction, and quality-of-life outcomes.
Otolaryngol Head Neck Surg., 146 (2012), pp. 834-841
[704]
Y. Cao, H. Hong, Y. Sun, Y. Lai, R. Xu, J. Shi, et al.
The effects of endoscopic sinus surgery on pulmonary function in chronic rhinosinusitis patients with asthma: a systematic review and meta-analysis.
Eur Arch Otorhinolaryngol., 276 (2019), pp. 1405-1411
[705]
J.L. Wentzel, Z.M. Soler, K. de Young, S.A. Nguyen, S. Lohia, R.J. Schlosser.
Leukotriene antagonists in nasal polyposis: A meta-analysis and systematic review.
Am J Rhinol Allergy., 27 (2013), pp. 482-489
[706]
Z. Huang, B. Zhou.
Clarithromycin for the treatment of adult chronic rhinosinusitis: a systematic review and meta-analysis.
Int. Forum Allergy Rhinol., 9 (2019), pp. 545-555
[707]
J. Van der Veen, S.F. Seys, M. Timmermans, P. Levie, M. Jorissen, W.J. Fokkens, et al.
Real-life study showing uncontrolled rhinosinusitis after sinus surgery in a tertiary referral centre.
Allergy., 72 (2017), pp. 282-290
[708]
P. Gevaert, L. Calus, T. van Zele, K. Blomme, N. de Ruyck, W. Bauters, et al.
Omalizumab is effective in allergic and nonallergic patients with nasal polyps and asthma. J. Allergy Clin.
Immunol., 131 (2013), pp. 110-116.e1
[709]
P. Gevaert, T.A. Omachi, J. Corren, J. Mullol, J. Han, S.E. Lee, D. Kaufman, M. Ligueros-Saylan, M. Howard, R. Zhu, R. Owen, K. Wong, L. Islam, C. Bachert.
Efficacy and safety of omalizumab in nasal polyposis: 2 randomized phase 3 trials.
J Allergy Clin Immunol., 146 (2020 Sep), pp. 595-605
[710]
C. Bachert, A.R. Sousa, V.J. Lund, G.K. Scadding, P. Gevaert, S. Nasser, et al.
Reduced need for surgery in severe nasal polyposis with mepolizumab: Randomized trial.
J Allergy Clin Immunol., 140 (2017), pp. 1024-1031.e14
[711]
P. Gevaert, N. van Bruaene, T. Cattaert, K. van Steen, T. van Zele, F. Acke, et al.
Mepolizumab, a humanized anti-IL-5 mAb, as a treatment option for severe nasal polyposis.
J. Allergy Clin Immunol., 128 (2011), pp. 989-995
[712]
C. Bachert, L. Mannent, R.M. Naclerio, J. Mullol, B.J. Ferguson, P. Gevaert, et al.
Effect of Subcutaneous Dupilumab on Nasal Polyp Burden in Patients With Chronic Sinusitis and Nasal Polyposis: A Randomized Clinical Trial.
JAMA., 142 (2016), pp. 698-699
[713]
C. Bachert, P.W. Hellings, J. Mullol, D.L. Hamilos, P. Gevaert, R.M. Naclerio, et al.
Dupilumab improves health-related quality of life in patients with chronic rhinosinusitis with nasal polyposis.
Allergy., 75 (2020), pp. 148-157
[714]
C. Bachert, J.K. Han, M. Desrosiers, P.W. Hellings, N. Amin, S.E. Lee, et al.
Efficacy and safety of dupilumab in patients with severe chronic rhinosinusitis with nasal polyps (LIBERTY NP SINUS-24 and LIBERTY NP SINUS-52): results from two multicentre, randomised, double-blind, placebo-controlled, parallel-group phase 3 trials.
Lancet., 394 (2019), pp. 1638-1650
[715]
J.K. Han, C. Bachert, W. Fokkens, M. Desrosiers, M. Wagenmann, S.E. Lee, et al.
Mepolizumab for chronic rhinosinusitis with nasal polyps (SYNAPSE): a randomised, double-blind, placebo-controlled, phase 3 trial.
Lancet Respir Med., 9 (2021 Oct), pp. 1141-1153
[716]
W.J. Fokkens, V.J. Lund, C. Hopkins, P.W. Hellings, R. Kern, S. Reitsma, et al.
European Position Paper on Rhinosinusitis and Nasal Polyps.
2020. Rhinology, 58 (2020), pp. 1-464
[717]
R.R. Orlandi, T.T. Kingdom, T.L. Smith, B. Bleier, A. DeConde, A.U. Luong, et al.
International consensus statement on allergy and rhinology: rhinosinusitis 2021.
Int Forum Allergy Rhinol., 11 (2021 Mar), pp. 213-739
[718]
L.Y. Chong, P. Piromchai, S. Sharp, K. Snidvongs, K.E. Webster, C. Philpott, C. Hopkins, M.J. Burton.
Biologics for chronic rhinosinusitis.
Cochrane Database Syst Rev., 3 (2021 Mar 12), pp. CD013513
[719]
R. Naclerio, F. Baroody, C. Bachert, B. Bleier, L. Borish, E. Brittain, et al.
Clinical Research Needs for the Management of Chronic Rhinosinusitis with Nasal Polyps in the New Era of Biologics: A National Institute of Allergy and Infectious Diseases Workshop.
J Allergy Clin Immunol Pract., 8 (2020 May), pp. 1532-1549.e1
[720]
H. Nagase, S. Ueki, S. Fujieda.
The roles of IL-5 and anti-IL-5 treatment in eosinophilic diseases: Asthma, eosinophilic granulomatosis with polyangiitis, and eosinophilic chronic rhinosinusitis.
Allergol Int., 69 (2020 Apr), pp. 178-186
[721]
D.R. Taylor, E.D. Bateman, L.P. Boulet, H.A. Boushey, W.W. Busse, T.B. Casale, et al.
A new perspective on concepts of asthma severity and control.
Eur Respir J., 32 (2008), pp. 545-554
[722]
E. Martínez-Moragón, J. Serra-Batllés, A. de Diego, M. Palop, P. Casan, C. Rubio-Terrés, et al.
por el grupo de Investigadores del estudio AsmaCost. Economic cost of treating the patient with asthma in Spain: the AsmaCost study.
Arch Bronconeumol., 45 (2009), pp. 481-486
[723]
J. Serra Batlles, V. Plaza, A. Comella.
Changes in clinical, pulmonary function, quality of life and costs in a cohort of asthmatic patients followed for 10 years.
Arch Bronconeumol., 47 (2011), pp. 482-487
[724]
M. Kerkhof, T.N. Tran, J.B. Soriano, S. Golam, D. Gibson, E.V. Hillyer, et al.
Healthcare resource use and costs of severe, uncontrolled eosinophilic asthma in the UK general population.
[725]
F.J. Álvarez-Gutiérrez, M. Blanco-Aparicio, F. Casas-Maldonado, V. Plaza, F.J. Gonzalez-Barcala, J.A. Carretero-Gracia, et al.
Documento de consenso de asma grave en adultos. Actualización 2022.
Open Respiratory Archives., 4 (2022), pp. 100192
[726]
K.F. Chung, S.E. Wenzel, J.L. Brozek, A. Bush, M. Castro, J. Sterk, et al.
International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma.
Eur Respir J, 43 (2014), pp. 343-373
[727]
J. Bousquet, E. Mantzouranis, A.A. Cruz, N. Aït-Khaled, C.E. Baena-Cagnani, E.R. Bleecker, et al.
Uniform definition of asthma severity, control, and exacerbations: Document presented for the World Health Organization Consultation on Severe Asthma.
J Allergy Clin Immunol., 126 (2010), pp. 926-938
[728]
E.H. Bel, A. Sousa, L. Fleming, A. Bush, K.F. Chung, J. Verrsnel, on behalf of the members of the Unbiased Biomarkers for the Prediction of Respiratory Disease Outcome (U-BIOPRED) Consortium, et al.
Consensus Generation.
Thorax., 66 (2011), pp. 910-917
[729]
P. Barranco, C. Pérez-Francés, S. Quirce, E. Gómez-Torrijos, R. Cárdenas, S. Sánchez-García, Severe Asthma Working Group of the SEAIC Asthma Committee, et al.
Consensus Document on the Diagnosis of Severe Uncontrolled Asthma.
J Investig Allergol Clin Immunol, 22 (2012), pp. 460-475
[730]
P.P. Hekking, M. Amelink, R.R. Wener, M.L. Bouvy, E.H. Bel.
Comorbibities in difficult-to-control asthma.
J Allergy Clin Immunol Pract., 6 (2018), pp. 108-113
[731]
S. Quirce, V. Plaza, C. Picado, M. Vennera, J. Casafont.
Prevalence of uncontrolled severe persistent asthma in pneumology and allergy hospital units in Spain.
J Investig Allergol Clin Immunol., 21 (2011), pp. 466-471
[732]
P.P. Hekking, R.R. Werner, M. Amelink, A.H. Zwinderman, M.L. Bouvy, E.H. Bel.
The prevalence of severe refractory asthma.
J Allergy Clin Immunol., 135 (2015), pp. 896-902
[733]
A.J. Woolcock.
Corticosteroid-resistant asthma. Definitions.
Ann J Respir Crit Care Med., 154 (1996), pp. S45-S48
[734]
C.R. Keenan-a, S. Salem, E.R. Fietz, R.C. Gualano, A.G. Stewart.
Glucocorticoid-resistant asthma and novel an-tiinflammatory drugs.
Drug Discovery Today., 17 (2012), pp. 1031-1038
[735]
C.R. Keenan, D. Radojicic, M. Li, A. Radwan, A.G. Stewart.
Heterogeneity inmechanisms influencing glucocorticoid sensitivity: the need for a systems biology approach to treatment of glucocorticoid-resistant inflammation.
Pharmacol Ther., 150 (2015), pp. 81-93
[736]
P.J. Barnes, A.P. Greening, G.K. Crompton.
Glucocorticoid resistance in asthma.
Am J Respir Crit Care Med., 152 (1995), pp. S125-S140
[737]
D. Reddy, F. Little.
Glucocorticoid-resistant asthma: more than meets the eye.
J Asthma., 50 (2013), pp. 1036-1044
[738]
C. Cisneros, C. Melero, C. Almonacid, M. Perpiñá, C. Picado, E. Martínez-Moragón, et al.
Guidelines for severe uncontrolled asthma.
Arch Bronconeumol., 51 (2015), pp. 235-246
[739]
E.I. Israel, H.K. Reddel.
Severe and difficult to trate asthma in adults.
NEMJ., 377 (2017), pp. 965-976
[740]
Guía de bolsillo para el manejo y la prevención del asma (GINA) 2019. Disponible en https://ginasthma.org/wp-content/uploads/2019/07/GINA-Spanish-2019-wms.pdf.
[741]
F.J. Álvarez-Gutiérrez, M. Blanco, V. Plaza, C. Cisneros, J.L. García-Rivero, A. Padilla, et al.
en representación del grupo de consenso Foro-SEPAR. Documento de consenso en asma grave en adultos: actualización 2020.
Open Respiratory Archives., 2 (2020), pp. 158-174
[742]
L.A. Pérez de Llano, R. Villoro, M. Merino, M.C. Gómez-Neira, C. Muñiz, A. Hidalgo.
Cost effectiveness of outpatient asthma clinic.
Arch Bronconeumol., 52 (2016), pp. 660-661
[743]
V.L. Clark, P.G. Gibson, G. Genn, S.A. Hiles, I.D. Pavord, V.M. McDonald, et al.
Multidimensional assesment of severe asthma: A systematic review and meta-analysis.
Respirology., 22 (2017), pp. 1262-1275
[744]
T.R. Tay, J. Lee, N. Radhakrishna, F. Hore-Lacy, R. Stirling, R. Hoy, et al.
A structured aprroach to specialist-referred difficult asthma patients improves control of comorbiditiesand enhances asthma outcomes.
J Allergy Clin Imunol Pract., 5 (2017), pp. 956-964
[745]
L.G. Heaney, E. Conway, C. Kelly, B.T. Johnson, C. English, M. Stevenson, et al.
Predictors of therapy asthma: outcomes of a systematic evaluation protocol.
Thorax., 58 (2003), pp. 561-566
[746]
S.D. Aaron, K.L. Vademheen, L.P. Boulet, R.A. Mclvor, J.M. Fitgeralt, P. Hernández, et al.
Canadian respiratory clinical research consortium. Overdiagnosis of asthma in obeses and nonobese adults.
CMAJ., 179 (2008), pp. 1121-1131
[747]
S.D. Aaron, L.P. Boulet, H.K. Reddel, A.S. Gershon.
Underdiagnosis and overdiagnosis of asthma.
Am J Respir Crit Care Med., 198 (2018), pp. 1012-1020
[748]
J. Gamble, M. Stevenson, L.G. Heaney.
A study of a multi-level intervention improve non-adherence in difficult to control asthma.
Respir Med., 105 (2011), pp. 1308-1315
[749]
A. Gherasim, D. Ahn, A. Bernstein.
Confounders of severe asthma: diagnoses to consider when asthma symptoms persist despite optimal therapy.
World Allergy organ J., 11 (2018), pp. 29
[750]
K.F. Chung.
Diagnosis and management of severe asthma.
Semin Respir Crit Care Med., 39 (2018), pp. 91-99
[751]
R.J. Hancox.
Bronchodilator tolerance and rebound bronchoconstriction during regular inhaled beta-agonist treatment.
Respir Med., 94 (2000), pp. 767-771
[752]
J.K. Gerald, T.F. Carr, C.Y. Wel, J.T. Holbrook, L.B. Gerald.
Albuterol overuse: a marker of psychological distress?.
J Allergy Clin Immunol Pract., 3 (2015), pp. 957-962
[753]
T.F. Carr, M. Kraft.
Use of biomarkers to identify phenotypes and endotypes of severe asthma.
Ann Allergy Asthma Immunol., 121 (2018), pp. 414-420
[754]
Z. Diamant, S. Vijverberg, K. Alving, A. Bakirtas, L. Bjermer, A. Custovic, et al.
Towards clinically applicable biomarkers for asthma- An EAACI position paper.
Allergy., 74 (2019), pp. 1835-1851
[755]
P. Haldar, I.D. Pavord, D.E. Shaw, M.A. Berry, M. Thomas, C.E. Brightling, et al.
Cluster analysis and clinical asthma phenotypes.
Am J Respir Crit Care Med., 178 (2008), pp. 218-224
[756]
G.P. Anderson.
Endotyping asthma: new insights into key pathogenic mechanisms in a complex, heterogeneous disease.
Lancet., 372 (2008), pp. 1107-1119
[757]
W.C. Moore, D.A. Meyers, S.E. Wenzel, W.G. Teague, H. Li, X. Li, et al.
National Heart, Lung, and Blood Institute's Severe Asthma Research Program. Identification of asthma phenotypes using a clustering analysis in the Severe Asthma Research Program.
Am J Respir Crit Care Med, 181 (2010), pp. 315-323
[758]
N.R. Bhakta, P.G. Woodruff.
Human asthma phenotypes: from the clinic, to cytokines, and back again.
Immunol Rev., 242 (2011), pp. 220-232
[759]
J. Lotvall, A. Akdis, L.B. Bacharier, L. Bjermer, T.B. Casale, A. Custovic, et al.
Asthma enditypes: a new approach to classification of disease entlities within the asthma syndrome.
J Allergy Clin Immunol., 127 (2011), pp. 533-560
[760]
S. Wenzel.
Asthma phenotypes: the evolution from clinical to molecular approaches.
Nat Med., 18 (2012), pp. 716-725
[761]
I.O. Agache.
From phenotypes to endotypes to asthma treatment.
Curr Opin Allergy Clin Immunol., 13 (2013), pp. 249-256
[762]
A.M. Fitzpatrick, W.C. Moore.
Severe asthma phenotypes - how should they guide evaluation and treatment?.
J Allergy Clin Immunol Practice., 5 (2017), pp. 901-908
[763]
W.W. Busse.
Definition and impact.
pp. 1-15
[764]
G. Brusselle, M. Germinaro, S. Weiss, J. Zangrilli.
Reslizumab in patients with inadequately controlled late-onset asthma and elevated blood eosinophils.
Pulm Pharmacol Ther., 43 (2017), pp. 39-45
[765]
P. Ilmarinen, L.E. Tuomisto, O. Niemelä, M. Tommola, J. Haanpaa, H. Kankaanranta.
Cluster analysis on longitudinal data of patients with adult-onset asthma.
J Allergy Clin Immunol Pract., 5 (2017), pp. 967-978
[766]
J. Darveaux, W.W. Busse.
Biologics in asthma - the next step toward personalized treatment.
J Allergy Clin Immunol Pract., 3 (2015), pp. 152-160
[767]
P.G. Woodruff, B. Modrek, D.F. Choy, G. Jia, A.R. Abbas, A. Ellwanger, et al.
T-helper type 2-driven inflammation defines major subphenotypes of asthma.
Am J Respir Crit Care Med., 180 (2009), pp. 388-395
[768]
G.G. Brusselle, C. Vanderstichele, P. Jordens, R. Deman, H. Slabbynck, V. Ringoet, et al.
Azithromycin for prevention of exacerbations in severe asthma (AZISAST): a multicentre randomized double-blind placebo-controlled trial.
[769]
M.C. Peters, Z.K. Mekonnen, S. Yuan, N.R. Bhakta, P.G. Woodruff, J.V. Fahy.
Measures of gene expression in sputum cells can identify TH2-high and TH2-low subtypes of asthma.
J Allergy Clin Immunol., 133 (2014), pp. 388-394
[770]
T.N. Tran, R.S. Zeiger, S.P. Peters, G. Colice, P. Newbold, M. Goldman, et al.
Overlap of atopic, eosinophilic, and TH2-high asthma phenotypes in a general population with current asthma.
Annals of Allergy, Asthma & Immunology., 116 (2016), pp. 37-42
[771]
S.C. Erzurum, B.M. Gaston.
Biomarkers in asthma: a real hope to better manage asthma.
Clin Chest Med., 33 (2012), pp. 459-471
[772]
W.W. Busse, S.T. Holgate, S.W. Wenzel, P. Klekotka, Y. Chon, J. Feng, et al.
Biomarker profiles in asthma with high vs low airway reversibility and poor disease control.
Chest., 148 (2015), pp. 1489-1496
[773]
R. Buhl, M. Humbert, L. Bjermer, P. Chanez, L.G. Heaney, I. Pavord, expert group of the European Consensus Meeting for Severe Eosinophilic Asthma, et al.
Severe eosinophilic asthma: a roadmap to consensus.
Eur Respir J., 49 (2017), pp. 49
[774]
W.C. Moore, A.T. Hastie, X. Li, H. Li, W.W. Busse, N.N. Jarjour, et al.
National Heart Lung, and Blood Institute's Severe Asthma Research Program Sputum neutrophil counts are associated with more severe asthma phenotypes using cluster analysis.
J Allergy Clin Immunol, 133 (2014), pp. 1557-1563
[775]
G.G. Brusselle, T. Maes, K.R. Bracke.
Eosinophils in the spotlight: eosinophilic airway inflammation in non-allergic asthma.
Nat Med., 19 (2013), pp. 977-979
[776]
A. Ray, J. Kolls.
Neutrophilic Inflammation in Asthma and Association with Disease Severity.
Trends Immunol, 38 (2017), pp. 942-954
[777]
D. Robinson, M. Humbert, R. Buhl, A.A. Cruz, H. Inoue, S. Korom, et al.
Revisiting type 2-high and type 2-low airway inflammation in asthma: current knowledge and therapeutic implications.
Clin Exp Allergy., 47 (2017), pp. 161-175
[778]
H.S. Chang, T.H. Lee, J.A. Jun, A.R. Baek, J.S. Park, S.M. Koo, et al.
Neutrophilic inflammation in asthma: mechanisms and therapeutic considerations.
Expert Rev Respir Med., 11 (2017), pp. 29-40
[779]
D.E. Shaw, A.R. Sousa, S.J. Fowler, L.J. Fleming, G. Roberts, J. Corfield, U-BIOPRED Study Group, et al.
Clinical and inflammatory characteristics of the European U-BIOPRED adult severe asthma cohort.
Eur Respir J., 46 (2015), pp. 1308-1321
[780]
C.S. Kuo, S. Pavlidis, M. Loza, F. Baribaud, A. Rowe, I. Pandis, U-BIOPRED Study Group, et al.
T-helper cell type 2 (Th2) and non-Th2 molecular phenotypes of asthma using sputum transcriptomics in U-IOPRED.
Eur Respir J., 49 (2017),
[781]
I. Sulaiman, G. Greene, E. MacHale, J. Seheult, M. Mokoka, S. D’Arcy, et al.
A randomized clinical trial of feed-back on inhaler adherence and technique in patients with severe uncontrolled asthma.
Eur Respir J., 51 (2018),
[782]
L.G. Heaney, J. Busby, P. Bradding, R. Chaudhuri, A.H. Mansur, R. Niven, et al.
Remotely monitored therapy and nitric oxide suppression identifies non-adherence in severe asthma.
Am J Crit Care Med., 4 (2019), pp. 454-464
[783]
H.A. Kerstjens, M. Engel, R. Dahl, P. Paggiaro, E. Beck, M. Vandewalker, et al.
Tiotropium in asthma poorly controlled with standard combination therapy.
N Engl J Med., 367 (2012), pp. 1198-1207
[784]
K.M. Kew, K. Dahri.
Long-acting muscarinic antagonists (LAMA) added to combination long-acting beta2-agonists and inhaled corticosteroids (LABA/ICS) versus LABA/ICS for adults with asthma.
Cochrane Database Syst Rev., (2016), pp. CD011721
[785]
M. Solèr, J. Matz, R. Townley, R. Buhl, J. O’Brien, H. Fox, et al.
The anti-IgE antibody omalizumab reduces exacerbations and steroid requirement in allergic asthmatics.
Eur Respir J., 18 (2001), pp. 254-261
[786]
W. Busse, J. Corren, B.Q. Lanier, M. McAlary, A. Fowler-Taylor, G.D. Cioppa, et al.
Omalizumab, anti-IgE recombinant humanized monoclonal antibody, for the treatment of severe allergic asthma.
J Allergy Clin Immunol., 108 (2001), pp. 184-190
[787]
N.A. Hanania, O. Alpan, D.L. Hamilos, J.J. Condemi, I. Reyes-Rivera, J. Zhu, et al.
Omalizumab in severe allergic asthma inadequately controlled with standard therapy: A randomized trial.
Ann Internal Med., 154 (2011), pp. 573-582
[788]
R. Normansell, S. Walker, S.J. Milan, E.H. Walters, P. Nair.
Omalizumab for asthma in adults and children.
Cochrane database of systematc reviews., (2014), pp. CD003559
[789]
F. Holguin, J.C. Cardet, K.F. Chung, S. Diver, D.S. Ferreira, A. Fitzpatrick, et al.
Management of severe asthma: a European Respiratory Society/American Thoracic Society guideline.
Eur Respir J., 55 (2020),
[790]
M. Humbert, C. Taillé, L. Mala, V. Le Gros, J. Just, M. Molimard, STELLAIR investigators.
Omalizumab effectiveness in patients with severe allergic asthma according to blood eosinophilic count: The STELLAIR study.
Eur Respir J., 51 (2018), pp. 1702523
[791]
T.B. Casale, A.T. Luskin, W. Busse, R.S. Zeiger, B. Trzaskoma, M. Yang, et al.
Omalizumab effectiveness by biomarker status in patients with asthma: Evidence from PROSPERO, a prospective rea-world study.
J Allergy Clin Immunol Pract., 7 (2019), pp. 156-164
[792]
D. Ledford, W. Busse, B. Trzaskoma, T.A. Omachi, K. Rosen, B.E. Chipps, et al.
A randomized multicenter study evaluating Xolair persistence of response after long-term therapy.
J allergy Clin Immunol., 140 (2017), pp. 162-169
[793]
M.D.C. Vennera, C. Sabadell, C. Picado, Spanish Omalizumab Registry.
Duration of the efficacy of omalizumab after treatment discontinuation in “real life” severe asthma.
[794]
C. Domingo, X. Pomares, A. Navarro, M.J. Amengual, C. Monton, A. Sogo, et al.
A step-down protocol for omalizumab treatment in oral corticosteroid-dependent allergic asthma patients.
Br J Clin Pharmacol., 84 (2018), pp. 339-348
[795]
A.L. Voskamp, A. Gillman, K. Symons, A. Sandrini, J.M. Rolland, R.E. O’Hehir, et al.
Clinical efficacy and immunologic effects of omalizumab in allergic bronchopulmonary aspergillosis.
J Allergy Clin Immunol Pract., 3 (2015), pp. 192-199
[796]
J.X. Li, L.C. Fan, M.H. Li, W.J. Cao, J.F. Xu.
Beneficial effects of Omalizumab therapy in allergic bronchopulmonary aspergillosis: A synthesis review of published literature.
Respir Med., 122 (2017), pp. 33-42
[797]
I.D. Pavord, S. Korn, P. Howarth, E.R. Bleecker, R. Buhl, N. Keene, et al.
Mepolizumab for severe eosinophilic asthma (DREAM): a multicenter, double-blind, placebo-controlled trial.
Lancet., 380 (2012), pp. 651-659
[798]
H.G. Ortega, M.C. Liu, I.D. Pavord, G.G. Brusselle, J.M. Fitzgerald, A. Chetta, et al.
Mepolizumab treatment in patients with severe eosinophilic asthma.
N Engl J Med., 371 (2014), pp. 1198-1208
[799]
H.G. Ortega, S.W. Yancey, B. Mayer, N.B. Gunsoy, O.N. Keene, E.R. Bleecker, 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., 4 (2016), pp. 549-556
[800]
P. Nair, M.M. Pizzichini, M. Kjarsgaard, M.D. Inman, A. Efthimiadis, E. Pizzichini, et al.
Mepolizumab for prednisone-dependent asthma with sputum eosinophilia.
N Engl J Med., 360 (2009), pp. 985-993
[801]
E.H. Bel, S.E. Wenzel, P.J. Thompson, C.M. Prazma, O.N. Keene, S.W. Yancey, et al.
Oral glucocorticoid-sparing effect of mepoliumab in eosinofphilic asthma.
N Engl J Med., 371 (2014), pp. 1189-1198
[802]
C. Domingo Ribas, T. Carrillo Díaz, M. Blanco Aparicio, E. Martínez Moragón, D. Banas Conejero, M.G. Sánchez Herrero, REDES Study Group.
Correction to: REal worlD Effectiveness and Safety of Mepolizumab in a Multicentric Spanish Cohort of Asthma Patients Stratified by Eosinophils: The REDES Study.
Drugs., 81 (2021 Nov), pp. 1949-1951
[803]
N. Lugogo, C. Domingo, P. Chanez, R. Leigh, M.J. Gilson, R.G. Price, et al.
Long-term Efficacy and Safety of Mepolizumab in Patients With Severe Eosinophilic Asthma: A Multi-center, Open-label.
Phase IIIb Study. Clin Ther., 38 (2016), pp. 2058-2070
[804]
S. Khatri, W. Moore, P.G. Gibson, R. Leigh, A. Bourdin, J. Maspero, et al.
Assessment of the long-term safety of mepolizumab and durability of clinical response in patients with severe eosinophilic asthma.
J Allergy Clin Immunol., 143 (2019), pp. 1742-1751
[805]
F. Roufosse, J.E. Kahn, M.E. Rothenberg, A.J. Wardlaw, A.D. Klion, S.Y. Kirby, et al.
Efficacy and safety of mepolizumab in hypereosinophilic syndrome: A phase III, randomized, placebo-controlled trial.
J Allergy Clin Immunol., 146 (2020), pp. 1397-1405
[806]
G.J. Gleich, F. Roufosse, G. Chupp, S. Faguer, B. Walz, A. Reiter, et al.
Safety and Efficacy of Mepolizumab in Hypereosinophilic Syndrome: An Open-Label Extension Study.
J Allergy Clin Immunol Pract., 9 (2021), pp. 4431-4440.e1
[807]
M. Castro, S. Mathur, F. Hargreave, L.P. Boulet, F. Xie, J. Young, Res-5-0010 Study Group, et al.
Reslizumab for poorly controlled, eosinophilic asthma: a randomized, placebo-controlled study.
Am J Respir Crit Care Med., 184 (2011), pp. 1125-1132
[808]
M. Castro, J. Zangrilli, M.E. Wechsler, E.D. Bateman, G.G. Brusselle, P. Bardin, et al.
Reslizumab for inadequately controlled asthma with elevated blood eosinophil counts: result from two multicenter, parallel, double-blind, randomized, placebo-controlled, phase 3 trials.
Lancet Respir Med., 3 (2015), pp. 355-366
[809]
J. Corren, S. Weinstein, L. Janka, J. Zangrilli, M. Garin.
Phase 3 Study of Reslizumab in Patients with Poorly Controlled Asthma: Effects Across a Broad Range of Eosinophil Counts.
Chest., 150 (2016), pp. 799-810
[810]
A. Bourdin, M. McDonald, R. Vanlandingham.
Reslizumab is effective in asthma patients with or without allergen specific IgE.
Allergy: european journal of allergy and clinical immunology., 73 (2018), pp. 202-203
[811]
P. Nair, P. Bardin, M. Humbert, K.R. Murphy, L. Hickey, M. Garin, et al.
Efficacy of Intravenous Reslizumab in Oral Corticosteroid Dependent Asthma.
J Allergy Clin Immunol Pract., 8 (2020), pp. 555-564
[812]
M. Mukherjee, F. Aleman, M. Kjarsgaard, B. Salter, G. Nair, N. La Vigne, et al.
Weight-adjusted intravenous reslizumab in severe asthma with inadequate response to fixed-dose subcutaneous mepolizumab.
Am J Respir Crit Care Med., 197 (2018), pp. 38-46
[813]
L.A. Pérez de Llano, B.G. Cosío, C. Domingo, I. Urrutia, I. Bobolea, A. Valero, et al.
Efficacy and safety of reslizumab in patients with severe asthma with inadequate response to omalizumab: A multicenter, open-label pilot study.
J Allergy Clin Immunol Pract., 7 (2019), pp. 2277-2283
[814]
K. Murphy, J. Jacobs, L. Bjermer, J.M. Fahrenholz, Y. Shalit, M. Garin, et al.
Long-term safety and efficacy of reslizumab in patients with eosinophilic asthma.
J Allergy Clin Immunol Pract., 5 (2017), pp. 1572-1581
[815]
E.R. Bleecker, J.M. Fitzgerald, P. Chanez, A. Papi, S.F. Weinstein, P. Braker, et al.
Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting beta-2agonists (SIROCCO): a randomized, multicenter, placebo-controlled, phase 3 trial.
Lancet., 88 (2016), pp. 2115-2127
[816]
J.M. FitzGerald, E.R. Bleecker, P. Nair, S. Korn, K. Ohta, M. Lommatzsch, et al.
Benralizumab, an anti-interleukin-5 receptor alpha monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomized, double-blind, placebo-controlled phase 3 trial.
Lancet., 388 (2016), pp. 2128-2141
[817]
P. Nair, S. Wenzel, K.F. Rabe, A. Bourdin, N.L. Lugogo, P. Kuna, et al.
Oral Glucocorticoid-Sparing Effect of Benralizumab in Severe Asthma.
N Engl J Med., 376 (2017), pp. 2448-2458
[818]
A. Menzies-Gow, J. Corren, E.H. Bel, J. Maspero, L.G. Heaney, M. Gurnell, et al.
Corticosteroid tapering with benralizumab treatment for eosi-nophilic asthma: PONENTE Trial.
ERJ Open Res., 5 (2019), pp. 00009-2019
[819]
M. Goldman, I. Hirsch, J.G. Zangrilli, P. Newbold, X. Xu.
The association between blood eosinophil count and benralizumab efficacy for patients with severe, uncontrolled asthma: subanalyses of the Phase III SIROCCO and CALIMA studies.
Current medical research and opinion., 33 (2017), pp. 1605-1613
[820]
J.M. FitzGerald, E.R. Bleecker, A. Menzies-Gow, J.G. Zangrilli, I. Hirsch, P. Metcalfe, et al.
Predictors of enhanced response with benralizumab for patients with severe asthma: pooled analysis of the SIROCCO and CALIMA studies.
Lancet respiratory medicine., 6 (2018), pp. 51-64
[821]
T.W. Harrison, P. Chanez, F. Menzella, G.W. Canonica, R. Louis, B.G. Cosio, 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.
Lancet Respir Med., 9 (2021 Mar), pp. 260-274
[822]
G.W. Canonica, T.W. Harrison, P. Chanez, F. Menzella, R. Louis, B.G. Cosio, et al.
Benralizumab improves symptoms of patients with severe, eosinophilic asthma with a diagnosis of nasal polyposis.
Allergy., 77 (2022 Jan), pp. 150-161
[823]
W.W. Busse, E.R. Bleecker, J.M. FitzGerald, G.T. Ferguson, P. Barker, S. Sproule, et al.
Long-term safety and efficacy of benralizumab in patients with severe, uncontrolled asthma: 1-year results from the BORA phase 3 extension trial.
Lancet Respir Med., 7 (2019), pp. 46-59
[824]
S. Korn, A. Bourdin, G. Chupp, B.G. Cosio, D. Arbetter, M. Shah, et al.
Integrated Safety and Efficacy Among Patients Receiving Benralizumab for Up to 5 Years.
J Allergy Clin Immunol Pract., 9 (2021 Dec), pp. 4381-4392.e4
[825]
E. Martínez-Moragón, I. García-Moguel, J. Nuevo, G. Resler, ORBE study investigators.
Real-world study in severe eosinophilic asthma patients refractory to anti-IL5 biological agents treated with benralizumab in Spain (ORBE study).
BMC Pulm Med., 21 (2021 Dec 18), pp. 417
[826]
S. Wenzel, L. Ford, D. Pearlman, S. Spector, L. Sher, F. Skobieranda, et al.
Dupilumab in persistent asthma with elevated eosinophil levels.
N Engl J Med., 368 (2013), pp. 2455-2466
[827]
S. Wenzel, M. Castro, J. Corren, J. Maspero, L. Wang, B. Zhang, et al.
Dupilumab efficacy and safety in adults with uncontrolled persistent asthma despite use of medium-to-high-dose inhaled corticosteroids plus a long-acting B2 agonist: a randomised double-blind placebo-controlled pivotal phase 2b dose-ranging trial.
[828]
M. Castro, J. Corren, I.D. Pavord, J. Maspero, S. Wenzel, K.F. Rabe, et al.
Dupilumab Efficacy and Safety in Moderate-to-Severe Uncontrolled Asthma.
N Engl J Med., 378 (2018), pp. 2486-2496
[829]
L.B. Bacharier, J.F. Maspero, C.H. Kateralis, A.G. Fiocchi, R. Gagnon, I. de Mir, et al.
Dupilumab in children with uncontrolled moderate to severe asthma.
New Eng J Med., 385 (2021), pp. 2230-2240
[830]
L.D. Sher, M.E. Wechsler, K.F. Rabe, J.F. Maspero, N. Daizadeh, X. Mao, et al.
Dupilumab Reduces Oral Corticosteroid Use in Patients With Corticosteroid-Dependent Severe Asthma: An Analysis of the Phase 3, Open-Label Extension TRAVERSE Trial.
Chest, 162 (2022 Jul), pp. 46-55
[831]
K.F. Rabe, P. Nair, G. Brusselle, J.F. Maspero, M. Castro, L. Sher, et al.
Efficacy and Safety of Dupilumab in Glucocorticoid-Dependent Severe Asthma.
N Engl J Med., 378 (2018), pp. 2475-2485
[832]
S. Diver, L. Khalfaoui, C. Emson, S.E. Wenzel, A. Menzies-Gow, M.E. Wechsler, et al.
Effect of tezepelumab on airway inflammatory cells, remodelling, and hyperresponsiveness in patients with moderate-to-severe uncontrolled asthma (CASCADE): a double-blind, randomised, placebo-controlled, phase 2 trial.
Lancet Respir Med., 9 (2021 Nov), pp. 1299-1312
[833]
A. Menzies-Gow, G. Colice, J.M. Griffiths, G. Almqvist, S. Ponnarambil, P. Kaur, G. Ruberto, K. Bowen, Å. Hellqvist, M. Mo, E. Garcia Gil.
NAVIGATOR: a phase 3 multicentre, randomized, double-blind, placebo-controlled, parallel-group trial to evaluate the efficacy and safety of tezepelumab in adults and adolescents with severe, uncontrolled asthma.
Respir Res., 21 (2020 Oct 13), pp. 266
[834]
M.E. Wechsler, G. Colice, J.M. Griffiths, G. Almqvist, T. Skarby, T. Piechowiak, et al.
SOURCE: a phase 3, multicentre, randomized, double-blind, placebo-controlled, parallel group trial to evaluate the efficacy and safety of tezepelumab in reducing oral corticosteroid use in adults with oral corticosteroid dependent asthma.
Lancet Respir Med., (2022 Mar 29:),
[835]
A. Menzies-Gow, M.E. Wechsler, C.E. Brightling, S. Korn, J. Corren, E. Israel, et al.
Long-term safety and efficacy of tezepelumab in people with severe, uncontrolled asthma (DESTINATION): a randomised, placebo-controlled extension study.
Lancet Respir Med., (2023 Jan 23),
[836]
K.M. Kew, K. Undela, I. Kotortsi, G. Ferrara.
Macrolides for chronic asthma.
Cochrane Database Syst Rev., (2015), pp. CD002997
[837]
S.A. Hiles, V.M. McDonald, M. Guilhermino, G.G. Brusselle, P.G. Gibson.
Does maintenance azithromycin reduce asthma exacerbations? An individual participant data meta-analysis.
Eur Respir J., 54 (2019), pp. 1901381
[838]
P.G. Gibson, I.A. Yang, J.W. Upham, P.N. Reynolds, S. Hodge, A.L. James, et al.
Effect of azithromycin on asthma exacerbations and quality of life in adults with persistent uncontrolled asthma (AMAZES): a randomized, double-blind, placebo-controlled trial.
Lancet., 390 (2017), pp. 659-668
[839]
S.D. Shukla, S.L. Taylor, P.G. Gibson, D. Barker, J.W. Upham, I.A. Yang, et al.
Add-on azithromycin reduces sputum cytokines in non-eosinophilic asthma: an AMAZES substudy.
Thorax., 76 (2021 Jul), pp. 733-736
[840]
M.E. Wechsler, M. Laviolette, A.S. Rubin, J. Fiterman, E. Lapa, J.R. Silva, et al.
Bronchial thermoplasty: long-term safety and effectiveness in patients with severe persitent asthma.
J Allergy Clin Immunol., 132 (2013), pp. 1295-1302
[841]
A.W.M. Goorsenberg, J.N.S. d’Hooghe, K. Srikanthan, N.H.T. Ten Hacken, E.J.M. Weersink, J.J.T.H. Roelofs, et al.
Bronchial Thermoplasty Induced Airway Smooth Muscle Reduction and Clinical Response in Severe Asthma. The TASMA Randomized Trial.
Am J Respir Crit Care Med, 203 (2021 Jan 15), pp. 175-184
[842]
M. Castro, A.I. Musani, M.L. Mayse, N.S. Shargill.
Bronchial thermoplasty: a novel technique in the treatment of severe asthma.
Ther Adv Respir Dis., 4 (2010), pp. 101-116
[843]
I.D. Pavord, G. Cox, N.C. Thomson, A.S. Rubin, P.A. Corris, R.M. Niven, et al.
Safety and efficacy of bronchial thermoplasty in symptomatic, severe asthma.
Am J Resoir Crit care med., 176 (2007), pp. 1185-1191
[844]
G. Cox, N.C. Thomson, A.S. Rubin, R.M. Niven, P.A. Corris, H.C. Siersted, et al.
Asthma control during the year after bronchial thermoplasty.
N Engl J Med., 29 (2007), pp. 1327-1337
[845]
G. Chupp, M. Laviolette, L. Cohn, C. McEvoy, S. Bansal, A. Shifren, et al.
Long-term outcomes of brionchial thermoplasty in subjects with severe asthma: a comparison of 3-year follow-up results from two prospective multicentre studies.
Eur Respir J., 50 (2017),
[846]
A. Torrego, I. Solá, A.M. Muñoz, I. Roqué, M. Figuls, J.J. Yepes-Nuñez, et al.
Bronchial thermoplasty for moderate or severe persistent asthma in adults.
Cochrane Database of Systematic Reviews., (2014), pp. CD009910
[847]
N.C. Thomson, A.S. Rubin, R.M. Niven, P.A. Corris, H.C. Siersted, R. Olivenstein, et al.
Long-term (5 year) safety of bronchial thermoplasty: asthma intervention research (AIR) trial.
BMC Pulm Med., 8 (2011),
[848]
R. Chaudhuri, A. Rubin, K. Sumino, E. Lapa, J.R. Silva, R. Niven, S. Siddiqui, et al.
Safety and effectiveness of bronchial thermoplasty after 10 years in patients with persistent asthma (BT10+): a follow-up of three randomised controlled trials.
Lancet Respir Med., 9 (2021 May), pp. 457-466
[849]
J.L. Zazzali, M.S. Broder, T.A. Omachi, E. Chang, G.H. Sun, K. Raimundo.
Risk of corticosteroid-related adverse events in asthma patients with high oral corticosteroid use.
Allergy Asthma Proc., 36 (2015), pp. 268-274
[850]
P.W. Sullivan, V.H. Ghushchyan, G. Globe, M. Schatz.
Oral corticosteroid exposure and adverse effects in asthmatic patients.
J Allergy Clin Immunol., 141 (2018), pp. 110-116
[851]
A.A. Dalal, M.S. Duh, L. Gozalo, M.N. Robitaille, F. Albers, S. Yancey, et al.
Dose-Response Relationship Between Long-Term Systemic Corticosteroid Use and Related Complications in Patients with Severe Asthma.
J Manag Care Spec Pharm, 22 (2016), pp. 833-847
[852]
R.G. Ojirala, T.K. Aldrich, D.J. Prezant, M.J. Sinnett, J.B. Enden, M.H. Williams.
High-dose intramuscular triamcinolone in severe, chronic, life-threatening asthma.
N Engl J Med., 324 (1991), pp. 585-589
[853]
A. Ten Brinke, A.H. Zwinderman, P.J. Sterk, K.F. Rabe, E.H. Bel.
“Refractory” eosinophilic airway inflammation in severe asthma: effect of parenteral corticosteroids.
Am J Respir Cir Care Med., 170 (2004), pp. 601-605
[854]
J.L. Izquierdo, C. Almonacid, C. Campos, D. Morena, M. Benavent, D. González-de-Olano, et al.
Systemic Corticosteroids in Patients with Bronchial Asthma: A Real-Life Study.
J Investig Allergol Clin Immunol., (2021 Nov 11),
[855]
L.M. Entrenas Costa, F. Casas-Maldonado, J.G. Soto Campos, A. Padilla-Galo, A. Levy, F.J. Álvarez Gutiérrez, et al.
Economic Impact and Clinical Outcomes of Omalizumab Add-On Therapy for Patients with Severe Persistent Asthma: A Real-World Study.
Pharmacoecon Open, 3 (2019 Sep), pp. 333-342
[856]
D.B. Price, F. Trudo, J. Voorham, X. Xu, M. Kerkhof, J. Ling Zhi Jie, et al.
Adverse outcomes from initiation of systemic corticosteroids for asthma: long-term observational study.
J Asthma Allergy., 11 (2018), pp. 193-204
[857]
D. Liu, A. Ahmet, L. Ward, P. Krishnamoorthy, E.D. Mandelcom, R. Leigh, et al.
A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy.
Allergy Asthma Clin Immunol., 9 (2013), pp. 30
[858]
C.M. Suehs, A. Menzies-Gow, D. Price, E.R. Bleecker, G.W. Canonica, M. Gurnell, et al.
Oral Corticosteroids Tapering Delphi Expert Panel. Expert Consensus on the Tapering of Oral Corticosteroids for the Treatment of Asthma. A Delphi Study.
Am J Respir Crit Care Med, 203 (2021), pp. 871-881
[859]
A. Menzies-Gow, M. Gurnell, L.G. Heaney, J. Corren, E.H. Bel, J. Maspero, et al.
Oral corticosteroid elimination via a personalised reduction algorithm in adults with severe, eosinophilic asthma treated with benralizumab (PONENTE): a multicentre, open-label, single-arm study.
Lancet Respir Med., 10 (2022), pp. 47-58
[860]
L. Pérez de Llano, I. Dávila, E. Martínez-Moragón, J. Domínguez-Ortega, C. Almonacid, C. Colás, et al.
Development of a Tool to Measure the Clinical Response to Biologic Therapy in Uncontrolled Severe Asthma: The FEV1, Exacerbations, Oral Corticosteroids, Symptoms Score.
J Allergy Clin Immunol Pract., 9 (2021 Jul), pp. 2725-2731
[861]
S.G. Kelsen, I.O. Agache, W. Soong, E. Israel, G.L. Chupp, D.S. Cheung, et al.
Astegolimab (anti-ST2) efficacy and safety in adults with severe asthma: A randomized clinical trial.
J Allergy Clin Immunol., 148 (2021 Sep), pp. 790-798
[862]
M.E. Wechsler, M.K. Ruddy, I.D. Pavord, E. Israel, K.F. Rabe, L.B. Ford, et al.
Efficacy and Safety of Itepekimab in Patients with Moderate-to-Severe Asthma.
N Engl J Med., 385 (2021 Oct 28), pp. 1656-1668
[863]
W.G. Teague, B.R. Phillips, J.V. Fahy, S.E. Wenzel, A.M. Fitzpatrick, W.C. Moore, et al.
Baseline features of the severe asthma research program (SARP III) cohort: differences with age.
J Allergy Clin Immunol Pract., 6 (2018), pp. 545-554
[864]
L. Fleming, C. Murray, A.T. Bansal, S. Hashimoto, H. Bisgaard, A. Bush, et al.
The burden of severe asthma in childhood and adolescence: resuklts from the paediatric U-BIOPRED cohorts.
Eur Respir J., 46 (2015), pp. 1322-1333
[865]
A. Lang, K.H. Carlsen, G. Haaland, C.S. Devulapalli, M. Munthe-Kaas, P. Mowinckel, et al.
Severe asthma in childhood: assessed in 10 year olds in a birth cohort study.
Allergy., 63 (2008), pp. 1054-1060
[866]
B. Nordlund, E. Melen, E.S. Schultz, H. Gronlund, G. Hedlin, I. Kull.
Prevalence of severe childhood asthma according to the WHO.
Respir Med., 108 (2014), pp. 1234-1237
[867]
A.M. Plaza-Martin, M.C. Vennera, J. Galera, L. Herráez, on behalf of the PREX Study Group.
Prevalence and clinical profile of difficult-to-control severe asthma in children: Results from pneumology and allergy hospital units in Spain.
Allergol Immunopathol (Madr.)., 42 (2014), pp. 510-517
[868]
A.J. Blasco, E.G. Pérez-Yarza, Lázaro, P. de Mercado, A. Bonillo, C.A. Díaz, A. Moreno.
Coste del asma en Pediatría en España: un modelo de evaluación de costes basado en la prevalencia.
An Pediatr (Barc)., 74 (2011), pp. 145-153
[869]
A. Tai, H. Tran, M. Roberts, N. Clarke, J. Wilson, C.F. Robertson.
The association between childhood asthma and adult chronic obstructive pulmonary disease.
[870]
M.J. McGeachie, K.P. Yates, X. Zhou, F. Guo, A.L. Sternberg, M.L. Van Natta, et al.
Patterns of growth and decline in lung function in persistent childhood asthma.
N Engl J Med., 374 (2016), pp. 1842-1852
[871]
G. Hedlin, A. Bush, K. Lodrup-Carlsen, G. Wennergren, F.M. de Benedictis, E. Melen, et al.
Problematic severe asthma in children, not one problem but many: a GA2LEN initiative.
Eur Respir J., 36 (2010), pp. 196-201
[872]
H. Ahmed, S. Turner.
Severe asthma in children -a reviews of definitions, epidemiology, and treatments options in 2019.
Pediatr Pulmonol., 54 (2019), pp. 778-787
[873]
A. Bush, S. Saglani.
Management of severe asthma in children.
Lancet., 376 (2010), pp. 814-825
[874]
M. Verkleij, A. Beelen, B.E. van Ewijk, R. Geenen.
Multidisciplinary treatment in children with problematic severe asthma: a prospective evaluation.
Pediatr Pulmonol, 52 (2017), pp. 588-597
[875]
A. Bush, L. Fleming, S. Saglani.
Severe asthma in children.
Respirology., 22 (2017), pp. 886-897
[876]
M. Bracken, L. Fleming, P. Hall, N. van Stiphout, C. Bossley, E. Biggart, et al.
The importance de nurse-led home visits in the assessment of children with problematic asthma.
Arch dis Child., 94 (2009), pp. 780-784
[877]
A.M. Fitzpatrick, W.G. Teague, D.A. Meyers, S.P. Peters, X. Li, H. Li, et al.
Heterogeneity of severe asthma in childhood: confirmation by cluster analysis of children in the National Institutes of health/National heart, lung, and blood Institute severe ashtma Research Program.
J Allergy Clin Immunol., 127 (2011), pp. 382-389
[878]
L.P. Boulet, D. Vervloet, Y. Magar, J.M. Foster.
Adherence: the goal to control asthma.
Clin Chest Med., 33 (2012), pp. 785-795
[879]
G.R. Todd, C.L. Acerini, R. Ross-Russell, S. Zahra, J.T. Warner, D. McCance.
Survey of adrenal crisis associated with inhaled corticosteroids in the United Kingdom.
Arch Dis Child., 87 (2002), pp. 457-461
[880]
H.F. Rosenberg, K.D. Dyer, P.S. Foster.
Eosinophils: changing perspectives in health and disease.
Nat Rev Immunol., 13 (2013), pp. 9-22
[881]
R.G. Suárez, C. Galván, C. Oliva, A. Aguirre, C. Vázquez, Grupo de Trabajo sobre Tabaquismo de la Infancia y Adolescencia de la Sociedad Española de Neumología Pediátrica.
Exposición pasiva al humo del tabaco del niño asmático y su asociación con la gravedad del asma.
An Pediatr (Barc)., 78 (2013), pp. 35-42
[882]
C.J. Bossley, L. Flemming, N. Ullmann, A. Gupta, A. Adams, P. Nagakumar, et al.
Assesment of corticosteroid response in paediatric severe asthma using a multi-domain approach.
J Allergy Clin Immunol., 138 (2016), pp. 413-420
[883]
W. Phipatanakul, D.T. Mauger, E.R. Sorkness.
Effects of age and disease severity on systemic corticosteroid responses in asthma.
Am J Respir Crit Care Med, 195 (2017), pp. 1439-1448
[884]
G.J. Rodrigo, H. Neffen.
Efficacy and safety of tiotropium in school-age children with moderate-to-severe symptomatic asthma: A systematic review.
Pediatr Allergy Immunol., 28 (2017), pp. 573-578
[885]
S.J. Szefler, C. Vogelberg, J.A. Bernstein, S. Goldstein, L. Mansfield, L. Zaremba-Pechmann, et al.
Tiotropium Is Efficacious in 6-to 17-Year-Olds with Asthma, Independent of T2 Phenotype.
J Allergy Clin Immunol Pract., 7 (2019), pp. 2286-2295
[886]
R. Normansell, S. Walker, S.J. Milan, E.H. Walters, P. Nair.
Omalizumab for asthma in adults and children.
Cochrane Database Syst Rev., (2014), pp. CD003559
[887]
G.J. Rodrigo, H. Neffen.
Systematic review on the use of omalizumab for the treatment of asthmatic children and adolescents.
Pediatr Allergy Immunol., 26 (2015), pp. 551-556
[888]
B.E. Chipps, B. Lanier, H. Milgrom, A. Deschildre, G. Hedlin, S.J. Szefler, et al.
Omalizumab in children with uncontrolled allergic asthma: review of clinical trial and real-world experience.
J Allergy Clin Immunol., 139 (2017), pp. 1431-1444
[889]
J. Corren, A. Kavati, B. Ortiz, J.A. Colby, K. Ruiz, B.A. Maiese, et al.
Efficacy and safety of omalizumab in children and adolescents with moderate-to-severe ashtma: A systematic literature review.
Allergy Asthma Proc., 38 (2017), pp. 250-263
[890]
A. Gupta, I. Pouliquen, D. Austin, R.G. Price, R. Kempsford, J. Steinfeld, et al.
Subcutaneous mepolizumab in children aged 6 to 11 years with severe eosinophilic asthma.
Pediatr Pulmonol., 54 (2019), pp. 1957-1967
[891]
H.A. Farne, A. Wilson, C. Powell, L. Bax, S.J. Milan.
Anti-IL5 therapies for asthma (Review).
Cochrane Database Syst Rev., (2017), pp. CD010834
[892]
A. Gupta, M. Ikeda, B. Geng, J. Azmi, R.G. Price, E.S. Bradford, S.W. Yancey, J. Steinfeld.
Long-term safety and pharmacodynamics of mepolizumab in children with severe asthma with an eosinophilic phenotype.
J Allergy Clin Immunol, 144 (2019), pp. 1336-1342
[893]
X. Pan, Y. Liu, J. Luo, S. Li, S. Diao, H. Li, et al.
The efficacy and safety of azithromycin in treatment for childhood asthma: A systematic review and meta-analysis.
Pediatr Pulmonol., 57 (2022), pp. 631-639
[894]
J.J. Ghimire, K.R. Jat, J. Sankar, R. Lodha, V.K. Iyer, H. Gautam, et al.
Azithromycin for poorly controlled asthma in children: A randomized controlled trial.
Chest., 161 (2022), pp. 1456-1464
[895]
S. Yosihara, H. Fukuda, M. Tamura, O. Arisaka, M. Ikeda, N. Fukuda, et al.
Efficacy and safety of salmeterol/fluticasone combination therapy in infants and preschool children with asthma insufficiently controlled by inhaled corticosteroids.
Drug Res (Sttutg.)., 66 (2016), pp. 371-376
[896]
E.J.L.E. Vrijlandt, G. El Azzi, M. Vandewalker, N. Rupp, T. Harper, L. Graham, et al.
Safety and efficacy of tiotropium in children aged 1-5 years with persistent asthmatic symptoms: a randomised, double-blind, placebo-controlled trial.
Lancet Respir Med., 6 (2018), pp. 127-137
[897]
P.J. Barnes.
Immunology of asthma and chronic obstructive pulmonary disease.
Nat Rev Immunol., 8 (2008), pp. 183-192
[898]
D.S. Postma, K.F. Rabe.
The asthma-COPD overlapy syndrome.
N Engl J Med., 373 (2015), pp. 1241-1249
[899]
J.L. Perret, S.C. Dharmage, M.C. Matheson, D.P. Johns, L.C. Gurrin, J.A. Burgess, et al.
The interplay between the effects of lifetime asthma, smoking, and atopy on fixed airflow obstruction in middle age.
Am J Respir Crit Care Med., 187 (2013), pp. 42-48
[900]
L.P. Hayden, M.H. Cho, B.A. Raby, T.H. Beaty, E.K. Silverman, C.P. Hersh, COPDGene Investigators.
Childhood asthma is associated with COPD and known asthma variants in COPDGene: a genome-wide association study.
Respir Res., 19 (2018), pp. 209
[901]
D.S. Bui, J.A. Burgess, A.J. Lowe, J.L. Perret, C.J. Lodge, M. Bui, et al.
Childhood Lung Function Predicts Adult Chronic Obstructive Pulmonary Disease and Asthma-Chronic Obstructive Pulmonary Disease Overlap Syndrome.
Am J Respir Crit Care Med, 196 (2017), pp. 39-46
[902]
A. Singh, C. Liu, B. Putman, R. Zeig-Owens, C.B. Hall, T. Schwartz, et al.
Predictors of Asthma/COPD Overlap in FDNY Firefighters With World Trade Center Dust Exposure: A Longitudinal Study.
Chest., 154 (2018), pp. 1301-1310
[903]
T. To, J. Zhu, K. Larsen, J. Simatovic, L. Feldman, K. Ryckman, et al.
Canadian Respiratory Research Network. Progression from Asthma to Chronic Obstructive Pulmonary Disease. Is Air Pollution a Risk Factor?.
Am J Respir Crit Care Med, 194 (2016), pp. 429-438
[904]
V. Plaza, F. Álvarez, M. Calle, C. Casanova, B.G. Cosío, A. López-Viña, et al.
Consensus on the Asthma-COPD Overlap Syndrome (ACOS) Between the Spanish COPD Guidelines (GesEPOC) and the Spanish Guidelines on the Management of Asthma (GEMA).
Arch Bronconeumol., 53 (2017), pp. 443-449
[905]
P.G. Gibson, J.L. Simpson.
The overlap syndrome of asthma and COPD: what are its features and how important is it?.
Thorax., 64 (2009), pp. 728-735
[906]
M. Hardin, M. Cho, M.L. McDonald, T. Beaty, J. Ramsdell, Bhatt 6, et al.
The clinical and genetic features of COPD-asthma overlap syndrome.
Eur Respir J., 44 (2014), pp. 341-350
[907]
M. Miravitlles, J.B. Soriano, J. Ancochea, L. Muñoz, E. Durán-Tauleria, G. Sánchez, et al.
Characterisation of the overlap COPD-asthma phenotype: focus on physical activity and health status.
Respir Med., 107 (2013), pp. 1053-1060
[908]
V. Koblizek, J. Chlumsky, V. Zindr, K. Neumannova, J. Zatloukal, J. Zak, et al.
Chronic obstructive pulmonary disease: official diagnosis and treatment guidelines of the Czech Pneumological and Phthisiological society; a novel phenotypic approach to COPD with patient-oriented care.
Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub., 157 (2013), pp. 189-201
[909]
GINA-GOLD diagnosis of disease of chronic airflow limitation: asthma, COPD and asthma-COPD overlap syndrome (ACOS). 2015. Disponible en www.goldcopd.org/asthma-copd-overlap.html.
[910]
H. Kankaanranta, T. Harju, M. Kilpeläinen, W. Mazur, J.T. Lehto, M. Katajisto, et al.
Diagnosis and pharmacotherapy of stable chronic obstructive pulmonary disease: the Finnish guidelines.
Basic Clin Pharmacol Toxicol., 116 (2015), pp. 291-307
[911]
D.D. Sin, M. Miravitlles, D.M. Mannino, J.B. Soriano, D. Price, B.R. Celli, et al.
What is asthma-COPD overlap syndrome?.
Towards a consensus definition from a round table discussion. Eur Respir J., 48 (2016), pp. 664-673
[912]
Global Initiative for Chronic Obstructive Lung Disea (GINA-GOLD) 2017. Disponible en https://goldcopd.org/wp-content/uploads/2016/04/wms-spanish-Pocket-Guide-GOLD-2017.pdf.
[913]
M. Miravitlles, F. Álvarez-Gutiérrez, M. Calle, C. Casanova, B.G. Cosío, A. López-Viña, et al.
Algorithm for identification of ACO: consensus between the Spanish COPD and asthma guidelines.
Eur Respir J., 49 (2017),
[914]
H. Joo, D. Han, J.H. Lee, C.K. Rhee.
Heterogeneity of asthma- COPD overlap syndrome.
Int J Chron Obstruct Pulmon Dis., 12 (2017), pp. 697-703
[915]
B. Alcázar-Navarrete, J.A. Trigueros, J.A. Riesco, A. Campuzano, J. Pérez.
Geographic variations of the prevalence and distribution of COPD phenotypes in Spain: “the ESPIRAL-ES study”.
Int J Chron Obstruct Pulmon Dis., 13 (2018), pp. 1115-1124
[916]
J.H. Song, C.H. Lee, D.K. Kim, H. Yoon, M.K. Byun, C.K. Rhee, et al.
Differences in prevalence of asthma-COPD overlap according to different criteria.
Medicine (Baltimore)., 97 (2018), pp. e12049
[917]
A. Barczyk, M. Maskey-Warzechowska, K. Górska, M. Barczyk, K. Kuziemski, P. Sliwinski, H. Batura-Gabryel, et al.
Asthma-COPD Overlap-A Discordance Between Patient Populations Defined by Different Diagnostic Criteria.
J Allergy Clin Immunol Pract., (2019 Apr 26),
[918]
R. De Marco, G. Pesce, A. Marcon, S. Accordini, L. Antonicelli, M. Bugiani, et al.
The coexistence of asthma and chronic obstructive pulmonary disease (COPD): prevalence and risk factors in young, middle-aged and elderly people from the general population.
[919]
A.M. Menezes, M.M. de Oca, R. Pérez-Padilla, G. Nadeau, F.C. Wehrmeister, M.V. Lopez-Varela, et al.
Increased risk of exacerbation and hospitalization in subjects with an overlap phenotype COPD asthma.
Chest., 145 (2014), pp. 297-304
[920]
M. Barrecheguren, C. Esquinas, M. Miravitlles.
The asthma-chronic obstructive pulmonary disease overlap syndrome (ACOS): opportunities and challenges.
Curr Opin Pulm Med., 21 (2015), pp. 74-79
[921]
A. Mendy, E. Forno, T. Niyonsenga, R. Carnahan, J. Gasana.
Prevalence and Features of Asthma-COPD Overlap in the U.S. 2007-2012.
Clin Respir J., 12 (2018), pp. 2369-2377
[922]
L. Ekerljung, R. Mincheva, S. Hagstad, A. Bjerg, G. Telg, G. Stratelis, et al.
Prevalence, clinical characteristics and morbidity of the Asthma-COPD overlap in a general population sample.
Journal of Asthma., 55 (2018), pp. 461-469
[923]
B.G. Cosío, J.B. Soriano, J.L. López-Campos, M. Calle-Rubio, J.J. Soler-Cataluña, J.P. de-Torres, et al.
Defining the Asthma-COPD Overlap Syndrome in a COPD Cohort.
Chest., 149 (2016), pp. 45-52
[924]
F. Nissen, D.R. Morales, H. Mullerova, L. Smeeth, I.J. Douglas, J.K. Quint.
Concomitant diagnosis of asthma and COPD: a quantitative study in UK primary care.
Br J Gen Pract., 68 (2018), pp. e775-e782
[925]
J.A. Krishnan, A. Nibber, A. Chisholm, D. Price, E.D. Bateman, L. Bjermer, et al.
Prevalence and Characteristics of Asthma-COPD Overlap in Routine Primary Care Practices.
Ann Am Thorac Soc., 16 (2019), pp. 1143-1150
[926]
F.T. Shaya, D. Dongyi, M.O. Akazawa, C.M. Blanchette, J. Wang, D.W. Mapel, et al.
Burden of concomitant asthma and COPD in a medicaid population.
Chest., 134 (2008), pp. 14-19
[927]
P. Kauppi, H. Kupiainen, A. Lindqvist, L. Tammilehto, M. Kilpeläinen, V.L. Kinnula, et al.
Overlap syndrome of asthma and COPD predicts low quality of life.
[928]
S.Y. Park, H. Jung, J.H. Kim, B. Seo, O.Y. Kwon, S. Choi, et al.
Longitudinal analysis to better characterize Asthma-COPD syndrome: Findings from an adult asthma cohort in Korea (COREA).
Clin Exp Allergy., 49 (2019), pp. 603-614
[929]
M. Calle, R. Casamor, M. Miravitlles.
Identification and distribution of COPD phenotypes in clinical practice according to Spanish COPD Guidelines: the FENEPOC study.
Int J Chron Obstruct Pulmon Dis., 12 (2017), pp. 2373-2383
[930]
J.L. Izquierdo-Alonso, J.M. Rodríguez-González, P. de Lucas-Ramos, I. Unzueta, X. Ribera, E. Antón, et al.
Prevalence and characteristics of three clinical phenotypes of chronic obstructive pulmonary disease (COPD).
Respir Med., 107 (2013), pp. 724-731
[931]
M. Miravitlles, M. Barrecheguren, M. Román-Rodríguez.
Frequency and characteristics of different clinical phenotypes of COPD.
Int J Tub Lung Dis., 19 (2015), pp. 992-998
[932]
J.F.M. Van Boven, M. Román-Rodríguez, J.F. Palmer, N. Toledo-Pons, B.G. Cosío, J.B. Soriano.
Comorbidome, Pattern, and Impact of Asthma-COPD Overlap Syndrome in Real Life.
Chest., 149 (2016), pp. 1011-1020
[933]
H. Andersén, P. Lampela, A. Nevanlinna, O. Säynäjäkangas, T. Keistinen.
High hospital burden in overlap syndrome of asthma and COPD.
Clin Respir J., 7 (2013), pp. 342-346
[934]
C.K. Rhee, H.K. Yoon, K.H. Yoo, Y.S. Kim, S.W. Lee, Y.B. Park, et al.
Medical utilization and cost in patients with overlap syndrome of chronic obstructive pulmonary disease and asthma.
COPD., 11 (2014), pp. 163-170
[935]
M. Sadatsafavi, H. Tavakoli, T. Kendzerska, A. Gershon, T. To, S.D. Aaron, J.M. FitzGerald, Canadian Respiratory Research Network.
History of Asthma in Patients with Chronic Obstructive Pulmonary Disease. A Comparative Study of Economic Burden.
Ann Am Thorac Soc., 13 (2016), pp. 188-196
[936]
R.M. Turner, M. DePietro, B. Ding.
Overlap of Asthma and Chronic Obstructive Pulmonary Disease in Patients in the United States: Analysis of Prevalence, Features, and Subtypes.
JMIR Public Health Surveill., 4 (2018), pp. e60
[937]
J.P. Llanos, H. Ortega, G. Germain, M.S. Duh, M.H. Lafeuille, S. Tiggelaar, et al.
Health characteristics of patients with asthma, COPD and asthma-COPD overlap in the NHANES database.
Int J Chron Obstruct Pulmon Dis., 13 (2018), pp. 2859-2868
[938]
B.G. Cosío, J.B. Soriano, J.L. López-Campos, M. Calle, J.J. Soler, J.P. de Torres, et al.
Distribution and Outcomes of a Phenotype-Based Approach to Guide COPD Management: Results from the CHAIN Cohort.
PLoS One., 11 (2016), pp. e0160770
[939]
K.E. Wurst, K. Kelly-Reif, G.A. Bushnell, S. Pascoe, N. Barnes.
Understanding asthma-chronic obstructive pulmonary disease overlap syndrome.
Respir Med., 110 (2016), pp. 1-11
[940]
C. Sorino, C. Pedone, N. Scichilone.
Fifteen-year mortality of patients with asthma-COPD overlap syndrome.
Eur J Intern Med., 34 (2016), pp. 72-77
[941]
R. Golpe, M. Suárez-Valor, I. Martín-Robles, P. Sanjuán-López, E. Cano-Jiménez, O. Castro-Añón, et al.
Mortality in COPD patients according to clinical phenotypes.
Int J Chron Obstruct Pulmon Dis., 13 (2018), pp. 1433-1439
[942]
S. Kumbhare, C. Strange.
Mortality in Asthma-Chronic Obstructive Pulmonary Disease Overlap in the United States.
South Med J., 111 (2018), pp. 293-298
[943]
GEMA4. 4. Guía española para el manejo del asma. Madrid: Luzán 5. 2019. Disponible en www.gemasma.com.
[944]
M. Miravitlles, J.J. Soler-Cataluña, M. Calle, J. Molina, P. Almagro, J.A. Quintano, et al.
Guía española de la enfermedad pulmonar obstructiva crónica (GesEPOC) 2017b. Tratamiento farmacológico en fase estable.
Archivos de Bronconeumología, 53 (2017), pp. 324-335
[945]
V. Plaza (Coord).
GEMA4.0. Guía Española para el Manejo del Asma.
Arch Bronconeumol, 51 (2015), pp. 2-54
[946]
S.A. Christenson, K. Steiling, M. van den Berge, K. Hijazi, P.S. Hiemstra, D.S. Postma, et al.
Asthma-COPD overlap. Clinical relevance of genomic signatures of type 2 inflammation in chronic obstructive pulmonary disease.
Am J Respir Crit Care., 191 (2015), pp. 758-766
[947]
M. Suzuki, H. Makita, S. Konno, K. Shimizu, H. Kimura, H. Kimura, et al.
Asthma-like features and clinical course of chronic obstructive pulmonary disease. An analysis from the Hokkaido COPD Cohort Study.
Am J Respir Crit Care Med., 194 (2016), pp. 1358-1365
[948]
B.G. Cosío, D. Dacal, L. Pérez de Llano.
Asthma-COPD overlap: identification and optimal treatment.
Ther Adv Respir Dis., 12 (2018),
[949]
D.J. Maselli, N.A. Hanania.
Management of Asthma COPD Overlap.
Ann Allergy Asthma Immunol., (2019 Jul 31),
[950]
S. Louie, A.A. Zeki, M. Schivo, A.L. Chan, K.Y. Yoneda, M. Avdalovic, et al.
The asthma-chronic obstructive pulmonary disease overlap syndrome: pharmacotherapeutic considerations.
Expert Rev Clin Pharmacol., 6 (2013), pp. 197-219
[951]
A.S. Gershon, M.A. Campitelli, R. Croxford, M.B. Stanbrook, T. To, R. Upshur, et al.
Combination long-acting ß-agonists and inhaled corticosteroids compared with long-acting ß-agonists alone in older adults with chronic obstructive pulmonary disease.
JAMA., 312 (2014), pp. 1114-1121
[952]
Y. Ishiura, M. Fujimura, Y. Shiba, N. Ohkura, J. Hara, K. Kasahara.
A comparison of the efficacy of once-daily fluticasone furoate/vilanterol with twice-daily fluticasone propionate/salmeterol in asthma-COPD overlap syndrome.
Pulm Pharmacol Ther., 35 (2015), pp. 28-33
[953]
S. Pascoe, N. Locantore, M. Dransfield, N.C. Barnes, I.D. Pavord.
Blood eosinophil counts, exacerbations and response to the addition of inhaled fluticasone furoate to vilanterol in patients with chronic obstructive pulmonary disease: A secondary analysis of data from two parallel randomized controlled trials.
Lancet Respir Med., 3 (2015), pp. 435-442
[954]
H.Y. Park, H. Lee, W.J. Koh, S. Kim, I. Jeong, H.K. Koo, et al.
Association of blood eosinophils and plasma periostin with FEV1 response after 3-month inhaled corticosteroid and long-acting beta2-agonist treatment in stable COPD patients.
Int J Chron Obstruct Pulmon Dis., 11 (2016), pp. 23-30
[955]
S.Y. Lee, H.Y. Park, E.K. Kim, S.Y. Lim, C.K. Rhee, Y.I. Hwang, et al.
Combination therapy of inhaled steroids and long-acting beta2-agonists in asthma-COPD overlap syndrome.
Int J Chron Obstruct Pulmon Dis., 11 (2016), pp. 2797-2803
[956]
D.P. Tashkin, B. Celli, S. Senn, D. Burkhart, S. Kesten, S. Menjoge, et al.
UPLIFT Study Investigators. A 4-year trial of tiotropium in chronic obstructive pulmonary disease.
N Engl J Med, 359 (2008), pp. 1543-1554
[957]
H.A. Kerstjens, M. Engel, R. Dahl, P. Paggiaro, E. Beck, M. Vandewalker, et al.
Tiotropium in asthma poorly controlled with standard combination therapy.
N Engl J Med., 367 (2012), pp. 1198-1207
[958]
T.S. Tat, A. Cilli.
Omalizumab treatment in asthma-COPD overlap syndrome.
J Asthma., 53 (2016), pp. 1048-1050
[959]
A.D. Yalcin, B. Celik, A.N. Yalcin.
Omalizumab (anti-IgE) therapy in the asthma-COPD overlap syndrome (ACOS) and its effects on circulating cytokine levels.
Immunopharmacol Immunotoxicol., 38 (2016), pp. 253-256
[960]
S. Maltby, P.G. Gibson, H. Powell, V.M. McDonald.
Omalizumab Treatment Response in a Population With Severe Allergic Asthma and Overlapping COPD.
[961]
N.A. Hanania, B.E. Chipps, N.M. Griffin, B. Yoo, A. Iqbal, T.B. Casale.
Omalizumab effectiveness in asthma-COPD overlap: Post hoc analysis of PROSPERO.
J Allergy Clin Immunol., 143 (2019), pp. 1629-1633
[962]
T.B. Casale, A.T. Luskin, W. Busse, R.S. Zeiger, B. Trzaskoma, M. Yang, et al.
Omalizumab Effectiveness by Biomarker Status in Patients with Asthma: Evidence From PROSPERO. A Prospective Real-World Study.
J Allergy Clin Immunol Pract, 7 (2019), pp. 156-164
[963]
J.P. Llanos, C.F. Bell, E. Packnett, E. Thiel, D.E. Irwin, B. Hahn, et al.
Real-world characteristics and disease burden of patients with asthma prior to treatment initiation with mepolizumab or omalizumab: a retrospective cohort database study.
J Asthma Allergy., 12 (2019), pp. 43-58
[964]
C.E. Brightling, E.R. Bleecker, R.A. Panettieri Jr., M. Bafadhel, D. She, C.K. Ward, et al.
Benralizumab for chronic obstructive pulmonary disease and sputum eosinophilia: a randomised, double-blind, placebo-controlled, phase 2a study.
Lancet Respir Med., 2 (2014), pp. 891-990
[965]
G.J. Criner, B.R. Celli, C.E. Brightling, A. Agusti, A. Papi, D. Singh, et al.
Benralizumab for the Prevention of COPD Exacerbations.
N Engl J Med., (2019 May 20),
[966]
A. Dasgupta, M. Kjarsgaard, D. Capaldi, K. Radford, F. Aleman, C. Boylan, et al.
A pilot randomised clinical trial of mepolizumab in COPD with eosinophilic bronchitis.
Eur Respir J., 49 (2017), pp. 3-43
[967]
I.D. Pavord, P. Chanez, G.J. Criner, H.A.M. Kerstjens, S. Korn, N. Lugogo, et al.
Mepolizumab for Eosinophilic Chronic Obstructive Pulmonary Disease.
N Engl J Med., 377 (2017), pp. 1613-1629
[968]
A. Yousuf, W. Ibrahim, N.J. Greening, C.E. Brightling.
T2 Biologics for Chronic Obstructive Pulmonary Disease.
J Allergy Clin Immunol Pract, 7 (2019), pp. 1405-1416
[969]
C. Bonham, K. Patterson, M. Strek.
Asthma Outcomes and Management During Pregnancy.
Chest., 152 (2018), pp. 515-527
[970]
A. Grosso, F. Locatelli, E. Gini, F. Albicini, C. Tirelli, I. Cerveri, et al.
The course of asthma during pregnancy in a recent, multicase-control study on respiratory health.
Allergy, Asthma & Clinical Immunology., 17 (2018), pp. 14-16
[971]
L.E. Grzeskowiak, B. Smith, A. Roy, G.A. Dekker, V.L. Clifton.
Patterns, predictors and outcomes of asthma control and exacerbations during pregnancy: a prospective cohort study.
ERJ Open Research., 2 (2016), pp. 00054-2015
[972]
E. Martínez-Moragón, A. Romero-Falcón, J.L. García-Rivero.
Algorithm for the management of asthma in pregnant women: a protocol to optimize processes in healthcare.
Expert Rev Respir Med., 11 (2017), pp. 1003-1012
[973]
G. Wang, V.E. Murphy, J. Namazy, H. Powell, M. Schatz, C. Chambers, et al.
The risk of maternal and placental complications in pregnant women with asthma: a systematic review and meta-analysis.
J Matern-Fetal Neonatal Med Off J Eur Assoc Perinat Med Fed Asia Ocean Perinat Soc Int Soc Perinat Obstet., 27 (2014), pp. 934-942
[974]
C. Tidemandsen, P. Egerup, C.S. Ulrik, V. Backer, D. Westergaard, A.P. Mikkelsen, et al.
Asthma Is Associated With Pregnancy Loss and Recurrent Pregnancy Loss: A Nationwide Cohort Study.
J Allergy Clin Immunol Pract, 10 (2022 Sep), pp. 2326-2332.e3
[975]
K. Abdullah, J. Zhu, A. Gershon, S. Dell, T. To.
Effect of asthma exacerbation during pregnancy in women with asthma: a population-based cohort study.
Eur Respir J., 55 (2020), pp. 1901335
[976]
Z. Ali, A.V. Hansen, C.S. Ulrik.
Exacerbations of asthma during pregnancy: Impact on pregnancy complications and outcome.
J Obstet Gynaecol., 36 (2016), pp. 455-461
[977]
C.B. Baarnes, A.V. Hansen, C.S. Ulrik.
Enrolment in an Asthma Management Program during Pregnancy and Adherence with Inhaled Corticosteroids: The ‘Management of Asthma during Pregnancy’ Program.
Respiration., 92 (2016), pp. 9-15
[978]
H. Powell, V.E. Murphy, M.J. Hensley, W. Giles, V.L. Clifton, P.G. Gibson.
Rhinitis in pregnant women with asthma is associated with poorer asthma control and quality of life.
J Asthma., 52 (2015), pp. 1023-1030
[979]
Z. Ali, L. Nilas, C.S. Ulrik.
Determinants if low risk of asthma exacerbations during pregnancy.
Clin Exp Allergy., 48 (2018), pp. 23-28
[980]
A. Lim, K. Stewart, K. König, J. George.
Systematic review of the safety of regular preventive asthma medications during pregnancy.
Ann Pharmacother., 45 (2011), pp. 931-945
[981]
V.E. Murphy, M.E. Jensen, P.G. Gibson.
Asthma during Pregnancy: Exacerbations, Management, and Health Outcomes for Mother and Infant.
Semin Respir Crit Care Med., 38 (2017), pp. 160-173
[982]
R.A. Charlton, J.M. Snowball, A.L. Nightingale, et al.
Safety of Fluticasone Propionate Prescribed for Asthma During Pregnancy: A UK Population-Based Cohort Study.
J Allergy Clin Immunol Pract., 3 (2015), pp. 772-779
[983]
NAEP. National Heart, Lung, Blood Institute, National Asthma Education, Prevention Program Asthma, Pregnancy Working Group.
NAEPP expert panel report. Managing asthma during pregnancy: recommendations for pharmacologic treatment-2004 update.
J Allergy Clin Immunol., 115 (2005), pp. 34-46
[984]
B. Kallen, H. Rydhstroem, A. Aberg.
Congenital malformations after the use of inhaled budesonid in early pregnancy. Obstet.
Gynecol., 93 (1999), pp. 392-395
[985]
E. Garne, A.V. Hansen, J. Morris, L. Zaupper, I. Barisic, M. Gatt, et al.
Use of asthma medication during pregnancy and risk of specific congenital abnormalities: a European case-malformed control study.
J Allergy Clin Immunol., 136 (2015), pp. 1496-1502
[986]
S. Eltonsy, F.-Z. Kettani, L. Blais.
Beta2-agonists use during pregnancy and perinatal outcomes: a systematic review.
Respir Med., 108 (2014), pp. 9-33
[987]
J.A. Namazy, M. Schatz.
Management of Asthma during Pregnancy: Optimizing Outcomes and Minimizing Risk.
Semin Respir Crit Care Med., 39 (2018), pp. 29-35
[988]
J.A. Namazy, L. Blais, E.B. Andrews, A.E. Scheuerle, M.D. Cabana, J.M. Thorp, et al.
Pregnancy outcomes in the omalizumab pregnancy registry and a disease-matched comparator cohort.
J Allergy Clin Immunol., 145 (2020), pp. 528-536.e1
[989]
J. Namazy, M.D. Cabana, A.E. Scheuerle, J.M. Thorp Jr., H. Chen, G. Carrigan, et al.
The Xolair Pregnancy Registry (EXPECT): the safety of omalizumab use during pregnancy.
J Allergy Clin Immunol, 135 (2015), pp. 407-412
[990]
G.V. De Arujo, D.F. Leite, J.A. Rizzo, E.S. Sarinho.
Asthma in pregnancy: association between the Asthma Control Test and the Global Initiative for Asthma classification and comparisons with spirometry.
Eur J Obstet Gynecol Reprod Biol., 203 (2016), pp. 25-29
[991]
V.E. Murphy, M.E. Jensen, J. Mattes, M.J. Hensley, W.B. Giles, M.J. Peek, et al.
The Breathing for Life Trial: a randomised controlled trial of fractional exhaled nitric oxide (FENO)-based management of asthma during pregnancy and its impact on perinatal outcomes and infant and childhood respiratory health.
BMC Pregnancy Childbirth, 16 (2016), pp. 111
[992]
K. Palmsten, M. Schatz, P.H. Chan, D.L. Johnson, C.D. Chambers.
Validation of the Pregnancy Asthma Control Test.
J Allergy Clin Immunol Pract., 4 (2016), pp. 310-315
[993]
E. Zairina, M.J. Abramson, C.F. McDonald, J. Li, T. Dharmasiri, K. Stewart, et al.
Telehealth to improve asthma control in pregnancy: A randomized controlled trial.
Respirology., 21 (2016), pp. 867-874
[994]
S.M. Tarlo, C. Lemier.
Occupational asthma.
N Engl J Med., 370 (2014), pp. 640-649
[995]
S.M. Tarlo, J. Balmes, R. Balkisson, J. Beach, W. Beckett, D. Bernstein, et al.
Diagnosis and Management of Work-Related Asthma. American College of Chest Physicians Consensus Statement.
Chest., 134 (2008), pp. 1S-41S
[996]
M. Kogevinas, J.P. Zock, D. Jarvis, H. Kromhout, L. Lillienberg, E. Plana, et al.
Exposure to substances in the workplace and new-onset asthma: an international prospective population-based study (ECRHS-II).
Lancet., 370 (2007), pp. 336-341
[997]
Nicholson PJ, Cullinan P, Burge PS, Boyle C. Occupational Asthma: Prevention, Identification and Management: Systematic Review and Recommendations. British Occupational Health Research Foundation. 2010. http://www.bohrf.org.uk/downloads/OccupationalAsthmaEvidenceReview-Mar2010.pdf
[998]
O. Meca, M.-J. Cruz, M. Sánchez-Ortiz, F.-J. González-Barcala, I. Ojanguren, X. Munoz.
Do Low Molecular Weight Agents Cause More Severe Asthma than High Molecular Weight Agents?.
PLoS ONE., 11 (2016), pp. e0156141
[999]
C. Beretta, C. Rifflart, G. Evrard, J. Jamart, J. Thimpont, O. Vandenplas.
Assessment of eosinophilic airway inflammation as a contribution to the diagnosis of occupational asthma.
Allergy., 73 (2018), pp. 206-213
[1000]
G. Moscato, G. Pala, C. Barnig, F. de Blay, S.R. del Giacco, I. Folletti, et al.
EAACI consensus statment for investigation of work-related asthma in non-specialized centres.
[1001]
S.M. Brooks, M.A. Weiss, I.L. Berstein.
Reactive airways dysfunction syndrome (RADS). Persistent asthma syndrome after high level irritant exposures.
Chest., 88 (1985), pp. 376-384
[1002]
S.M. Tarlo, I. Broder.
Irritant-induced occupational asthma.
Chest., 96 (1989), pp. 297-300
[1003]
D.G. Kern.
Outbreak of the reactive airways dysfunction syndrome after a spill of glacial acetic acid.
Am Rev Respir Dis., 144 (1991), pp. 1058-1064
[1004]
D. Heederik, P.K. Henneberg, C.A. Redlich.
Primary prevention: exposure reduction, skin exposure and respiratory protection.
Eur Respir Rev., 21 (2012), pp. 112-124
[1005]
D. Gautrin, H. Ghezzo, C. Infante-Rivard, J.L. Malo.
Incidence and determinants of IgE-mediated sensitizacion in apprentices: a prospective study.
Am J Respir Crit. Care Med., 162 (2000), pp. 1222-1228
[1006]
O. Vandenplas, J. Godet, L. Hurdubaea, C. Rifflart, H. Suojalehto, M. Wiszniewska, et al.
European network for the PHenotyping of OCcupational ASthma (E-PHOCAS) investigators. Are high- and low-molecular-weight sensitizing agents associated with different clinical phenotypes of occupational asthma?.
Allergy., 74 (2019), pp. 261-272
[1007]
A. Adisesh, L. Gruszka, E. Robinson, G. Evans.
Smoking status and immunoglobulin E seropositivity to workplace allergens.
Occup Med (Lond)., 61 (2011), pp. 62-64
[1008]
O. Vandenplas, H. Ghezzo, X. Munoz, G. Moscato, L. Perfetti, C. Lemière, et al.
What are the questionnaire items most useful in identifying subjects with occupational asthma?.
Eur Respir J., 26 (2005), pp. 1056-1063
[1009]
M.J. Cruz, X. Muñoz.
The current diagnostic role of the specific occupational laboratory challenge test.
Curr Opin Allergy Clin Inmunol., 12 (2012), pp. 119-125
[1010]
J. Cote, S. Kennedy, M. Chan-Yeung.
Sensitivity and specificity of PC20 and peak expiratory flow rate in cedar asthma.
J Allergy Clinical Immunol., 85 (1990), pp. 592-598
[1011]
A. Cartier, J. Sastre.
Clinical assessment of occupational asthma and its differential diagnosis.
Inmunol Allergy Clin North Am., 31 (2011), pp. 717-728
[1012]
J.A. Pralong, C. Lemière, T. Rochat, J. L’Archevêque, M. Labrecque, A. Cartier.
Predictive value of nonspecific bronchial responsiveness in occupational asthma.
Journal of Allergy and Clinical Immunology, 137 (2016), pp. 412-416
[1013]
M.P. Berges-Gimeno, R.A. Simon, D.D. Stevenson.
The natural history and clinical characteristics of aspirin-exacerbated respiratory disease.
Ann. Allergy Asthma Immunol., 89 (2002), pp. 474-478
[1014]
H. Suojalehto, K. Suuronen, P. Cullinan.
Specific challenge testing for occupational asthma: revised handbook.
Eur Respir J., 54 (2019), pp. 1901026
[1015]
P.K. Henneberger, J.R. Patel, G.J. de Groene, J. Beach, S.M. Tarlo, T.M. Pal, et al.
Workplace interventions for treatment of occupational asthma.
Cochrane Database Syst Rev., 10 (2019), pp. CD006308
[1016]
J.P. Parsons, C. Kaeding, G. Phillips, D. Jarloura, G. Wadley, J.G. Mastronarde.
Prevalence of exercise-induced bronchospasm in a cohort of varsity college athletes.
Med Sci Sports Exerc., 39 (2007), pp. 1487-1492
[1017]
J.P. Parsons, T.S. Hallstrand, J.G. Mastronarde, D.A. Kaminsky, K.W. Rundell, J.H. Hull, et al.
American Thoracic Society Subcommittee on Exercise-induced Bronchoconstriction.
An official American Thoracic Society clinical practice guideline: exercise-induced bronchoconstriction. Am J Respir Crit Care Med, 187 (2013), pp. 1016-1027
[1018]
M.A. Krafczyk, C.A. Asplund.
Exercise-induced bronchoconstriction: diagnosis and management.
Am Fam Physician., 84 (2011), pp. 427-434
[1019]
K.W. Rundell, J.B. Slee.
Exercise and other indirect challenges to demonstrate asthma or exercise induced bronchoconstriccion in athletes.
J Allergy Clin Immunol., 122 (2008), pp. 238-248
[1020]
J.M. Weiler, J.D. Brannan, C.C. Randolph, T.S. Hallstrand, J. Parsons, W. Silvers, et al.
Exercise-induced bronchoconstriction update-2016.
J Allergy Clin Immunol., 138 (2016), pp. 1292-1295.e36
[1021]
S.D. Anderson, P. Kippelen.
Airway injury as a mechanism for exercise-induced bronchoconstriccion in elite athletes.
J Allergy Clin Immunol., 122 (2008), pp. 225-235
[1022]
J. Ciencewicki, S. Trivedi, S.R. Kleeberger.
Oxidants and the phatogesis of lung deseases.
J Allergy Clin Immunol., 122 (2008), pp. 456-468
[1023]
F. De Baets, E. Bodart, M. Dramaix-Wilmet, S. van Daele, G. de Bilderling, S. Masset, et al.
Exercise-induced respiratory symthoms are poor predictors of bronchoconstriccions.
Pediatr Pulmonol., 39 (2005), pp. 301-305
[1024]
T.S. Hallstrand, J.D. Leuppi, G. Joos, L.H. Graham, K.H. Carlsen, D.A. Kaminsky, et al.
ERS technical standard on bronchial challenge testing: pathophysiology and methodology of indirect airway challenge testing.
Eur Respir J, 52 (2018), pp. 1801033
[1025]
M. Weimberger, M. Abu-Hasan.
Perceptions and pathophysiopatology of dyspnea and exercise intolerance.
Pediatr Clin North Am., 56 (2009), pp. 33-48
[1026]
R.O. Crapo, R. Casaburi, A.L. Coates, P.L. Enright, J.L. Hankinson, C.G. Irvin, et al.
Guidelines for methacholine and exercise challenge testing-1999.
Am J Respir Crit Care Med., 161 (2000), pp. 309-329
[1027]
J.M. Weiler, S. Bonini, R. Coifman, T. Craig, L. Delgado, M. Capão-Filipe, et al.
American Academy of Allergy, Asthma and Inmmunology Work Group report: exercise-induced asthma.
J Allergy Clin Immunol, 119 (2007), pp. 1349-1358
[1028]
D.M. Dryden, C.H. Spooner, M.K. Stickland, B. Vandermeer, L. Tjosvold, L. Bialy, et al.
Exercise-induced bronchoconstriction and asthma.
Evid Rep Technol Assess (Full Rep)., 189 (2010), pp. 1-154
[1029]
M. Weinberger.
Long-acting beta-agonists and exercise.
J Allergy ClinImmunol., 122 (2008), pp. 251-253
[1030]
P. Kippelen, J. Larsson, S.D. Anderson, J.D. Brannan, I. Delin, B. Dahlen, et al.
Acute effects of beclomethasone on hyperpnea-induced bronchoconstriction.
Med Sci Sports Exerc., 42 (2010), pp. 273-280
[1031]
G. Philip, D.S. Pearlman, C. Villaran, C. Legrand, T. Loeys, R.B. Langdon, et al.
Single-dose montelukast or salmeterol as protection against exercise-induced bronchoconstriction.
Chest., 132 (2007), pp. 875-883
[1032]
F.S. Ram, S.M. Robinson, P.N. Black, J. Picot.
Physical training for asthma.
Cochrane Database Syst Rev., (2005), pp. CD001116
[1033]
M.K. Stickland, B.H. Rowe, C.H. Spooner, B. Vandermeer, D.M. Dryden.
Effect of warm-up exercise on exercise-induced bronchoconstriction.
Med Sci Sports Exerc., 44 (2012), pp. 389-391
[1034]
T.D. Mickleborough.
A nutritional approach to managing exercise-induced asthma.
Exerc Sport Sci Rev., 36 (2008), pp. 135-144
[1035]
J.P. Rajan, N.E. Wineinger, D.D. Stevenson, A.A. White.
Prevalence of aspirin-exacerbated respiratory disease among asthmatic patients: a meta-analysis of the literature.
J Allergy Clin Immunol., 135 (2015), pp. 676-681
[1036]
C. Jenkins, J. Costello, L. Hodge.
Systematic review of prevalence of aspirin induced asthma and its implications for clinical practice.
[1037]
M. Sanak.
Eicosanoid mediators in the airway inflammation of asthmatic patients: what is new?.
Allergy Asthma Immunol Res., 8 (2016), pp. 481-490
[1038]
K.N. Cahill, J.C. Bensko, J.A. Boyce, T.M. Laidlaw.
Prostaglandin D(2): a dominant mediator of aspirin-exacerbated respiratory disease.
J Allergy Clin Immunol, 135 (2015), pp. 245-252
[1039]
J.J. Eastman, K.J. Cavagnero, A.S. Deconde, A.S. Kim, M.R. Karta, D.H. Broide, et al.
Group 2 innate lymphoid cells are recruited to the nasal mucosa in patients with aspirin-exacerbated respiratory disease.
J Allergy Clin Immunol., 140 (2017), pp. 101-108
[1040]
M.L. Kowalski, I. Agache, S. Bavbek, A. Bakirtas, M. Blanca, G. Bochenek, et al.
Diagnosis and management of NSAID-Exacerbated Respiratory Disease (N-ERD)-a EAACI position paper.
Allergy., 74 (2019), pp. 28-39
[1041]
G. Bochenek, T. Stachura, K. Szafraniec, H. Plutecka, M. Sanak, E. Nizankowska-Mogilnicka, et al.
Diagnostic Accuracy of Urinary LTE4 Measurement to Predict Aspirin-Exacerbated Respiratory Disease in Patients with Asthma.
J Allergy Clin Immunol Pract, 6 (2018), pp. 528-535
[1042]
A. Alonso-Llamazares, C. Martínez-Cócera, J. Dominguez-Ortega, T. Robledo-Echarren, M. Cimarra-Álvarez, M. Mesa del Castillo.
Nasal Provocation test (NPT) with aspirin: a sensitive and safe method to diagnose aspirin-induced asthma (AIA).
[1043]
P. Barranco, I. Bobolea, J.I. Larco, N. Prior, M.C. López-Serrano, S. Quirce.
Diagnosis of Aspirin-Induced Asthma combining the bronchial and the oral challenge tests: A pilot study.
J Investig Allergol Clin Immunol., 19 (2009), pp. 446-452
[1044]
J. Quiralte-Castillo, M.R. Ávila-Castellano, S. Cimbollek, P. Benaixa, S. Leguisamo, K. Baynova, et al.
Nasal ketorolac challenge using acoustic rhinometry in patients with Aspirin-Exacerbated Respiratory Disease.
J Investig Allergol Clin Immunol., 27 (2017), pp. 169-174
[1045]
A.A. White, D.D. Stevenson.
Aspirin-Exacerbated Respiratory Disease.
N Engl J Med, 379 (2018), pp. 1060-1070
[1046]
S.E. Dahlén, K. Malmstrom, E. Nizankowska.
Improvement of aspirin-intolerant asthma by montelukast, a leukotriene antagonist: a randomized, double-blind, placebo-controlled trial.
Am J Respir Crit Care Med., 165 (2002), pp. 9-14
[1047]
G. Stryjewska-Makuch, M. Humeniuk-Arasiewicz, E. Jura-Szoltys, J. Glück.
The effect of Antileukotrienes on the results of postoperative treatment of paranasal sinuses in patients with Non-Steroidal Anti-Inflammatory Drug-Exacerbated Respiratory Disease.
Int Arch Allergy Immunol., 179 (2019), pp. 281-289
[1048]
T. Jean, V. Eng, J. Sheikh, M.S. Kaplan, B. Goldberg, S. Jau Yang, et al.
Effect of omalizumab on outcomes in patients with aspirin-exacerbated respiratory disease.
Allergy Asthma Proc., 40 (2019), pp. 316-320
[1049]
H. Hayashi, C. Mitsui, E. Nakatani, Y. Fukutomi, K. Kajiwara, K. Watai, et al.
Omalizumab reduces cysteinyl leukotriene and 9?.,11?.-prostaglandin F2 overproduction in aspirin-exacerbated respiratory disease.
J Allergy Clin Immunol., 137 (2016), pp. 1585-1587
[1050]
E. Phillips-Anglés, P. Barranco, M. Lluch-Bernal, J. Domínguez-Ortega, V. López-Carrasco, S. Quirce.
Aspirin tolerance in patients with nonsteroidal anti-inflammatory drug-exacerbated respiratory disease following treatment with omalizumab.
J Allergy Clin Immunol Pract., 5 (2017), pp. 842-845
[1051]
P. Gevaert, N. van Bruaene, T. Cattaert, K. van Steen, T. van Zele, F. Acke, et al.
Mepolizumab, a humanized anti-IL-5 mAb, as a treatment option for severe nasal polyposis.
J Allergy Clin Immunol., 128 (2011), pp. 989-995
[1052]
A.G. Kartush, J.K. Schumacher, R. Shah, M.O. Patadia.
Biologic Agents for the treatment of chronic rhinosinusitis with nasal polyps.
Am J Rhinol Allergy, 33 (2019), pp. 203-211
[1053]
C. Bachert, P.W. Hellings, J. Mullol, R.M. Naclerio, J. Chao, N. Amin, et al.
Dupilumab improves patient-reported outcomes in patients with chronic rhinosinusitis with nasal polyps and comorbid asthma.
J Allergy Clin Immunol Pract., 7 (2019), pp. 2447-2449
[1054]
K.A. Cook, D.D. Stevenson.
Current complications and treatment of aspirin-exacerbated respiratory disease.
Exp Rev Respir Med., 10 (2016), pp. 1305-1316
[1055]
D.D. Stevenson.
Aspirin and NSAID sensitivity.
Immunol Allergy Clin North Am., 24 (2004), pp. 491-505
[1056]
Y. El Miedany, S. Youssef, I. Ahmed, M. El Gaafary.
Safety of etoricoxib, a specific cyclooxygenase-2 inhibitor, in asthmatic patients with aspirin-exacerbated respiratory disease.
Ann Allergy Asthma Immunol., 97 (2006), pp. 105-109
[1057]
A. Prieto, M. de Barrio, E. Martín, M. Fernández-Bohórquez, F.J. de Castro, F.J. Ruiz, et al.
Tolerability to nabumetone and meloxicam in patients with nonsteroidal anti-inflammatory drug intolerance.
J Allergy Clin Immunol., 119 (2007), pp. 960-964
[1058]
J.Y. Lee, R.A. Simon, D.D. Stevenson.
Selection of aspirin dosages for aspirin desensitization treatment in patients with aspirin-exacerbated respiratory disease.
J Allergy Clin Immunol., 119 (2007), pp. 157-164
[1059]
D.K. Chu, D.J. Lee, K.M. Lee, H.J. Schünemann, W. Szczeklik, J.M. Lee.
Benefits and harms of aspirin desensitization for aspirin-exacerbated respiratory disease: a systematic review and meta-analysis.
Int Forum Allergy Rhinol., (2019 Sep 13),
[1060]
M. Swierczynska-Krepa, M. Sanak, G. Bochenek, P. Strek, A. Cmiel, A. Gielicz, et al.
Aspirin desensitization in patients with aspirin-induced and aspirin-tolerant asthma: a double-blind study.
J Allergy Clin Immunol., 134 (2014), pp. 883-890
[1061]
K.M. Walters, J.D. Waldram, K.M. Woessner, A.A. White.
Long-term clinical outcomes of aspirin desensitization with continuous daily aspirin therapy in Aspirin-Exacerbated Respiratory Disease.
Am J Rhinol Allergy., 32 (2018), pp. 280-286
[1062]
L. Renjiao, L. Fengming.
The safety and efficacy of aspirin desensitization combined with long-term aspirin therapy in Aspirin-exacerbated respiratory disease.
J Investig Allergol Clin Immunol., (2019 Jul 8),
[1063]
A. Rozsasi, D. Polzehl, T. Deutschle, E. Smith, K. Wiesmiller, H. Riechelmann, et al.
Long-term treatment with aspirin desensitization: a prospective clinical trial comparing 100 and 300 mg apirin daily.
Allergy., 63 (2008), pp. 1228-1234
[1064]
W.W. Pleskow, D.D. Stevenson, D.A. Mathison, R.A. Simon, M. Schatz, R.S. Zeiger.
Aspirin desensitization in aspirin-sensitive asthmatic patients: clinical manifestations and characterization of the refractory period.
J Allergy Clin Immunol., 69 (1982), pp. 11-19
[1065]
T. Halvorsen, E.S. Walsted, C. Bucca, A. Bush, G. Cantarella, G. Friedrich, et al.
Inducible laryngeal obstruction: an official joint European Respiratory Society and European Laryngological Society statement.
European Respiratory Journal., 50 (2017), pp. 1602221
[1066]
K. Low, L. Ruane, N. Uddin, P. Finlay, K.K. Lau, K. Hamza, et al.
Abnormal vocal cord movement in patients with and without airway obstruction and asthma symptoms.
Clinical and Experimental Allergy., 47 (2017), pp. 200-207
[1067]
J. Haines, S.H.K. Chua, J. Smith, C. Slinger, A.J. Simpson, S.J. Fowler.
Triggers of breathlessness in inducible laryngeal obstruction and asthma.
Clinical and Experimental Allergy., 50 (2020), pp. 1230-1237
[1068]
J. Ye, M. Nourie, F. Hoguin, A.I. Gillespie.
The Ability of Patient-Symptom Questionnaires to Differentiate PVFMD From Asthma.
Journal of Voice, 31 (2017), pp. 382.e1-382.e8
[1069]
M.J. Morris, K.L. Christopher.
Diagnostic criteria for the classification of vocal cord dysfunction.
Chest., 138 (2010), pp. 1213-1223
[1070]
A. Fretzayas, M. Moustaki, I. Loukou, K. Douros.
Differentiating vocal cord dysfunction from asthma.
Journal of Asthma and Allergy., 10 (2017), pp. 277283
[1071]
N. Denipah, C.M. Dominguez, E.P. Kraai, T.L. Kraai, P. Leos, D. Braude.
Acute Management of Paradoxical Vocal Fold Motion (Vocal Cord Dysfunction).
Annals of Emergency Medicine, 69 (2017), pp. 18-23
[1072]
B. De Silva, D. Crenshaw, L. Matrka, L.A. Forrest.
Vocal fold botulinum toxin injection for refractory paradoxical vocal fold motion disorder.
Laryngoscope., 129 (2019), pp. 808-811
[1073]
Liyanagedara S, McLeod R, Elhassan HA. Exercise induced laryngeal obstruction: a review of diagnosis and management. European Archives of Oto-Rhino-Laryngology. Springer. 2017. Disponible en https://doi.org/10.1007/s00405-016-4338-1.
[1074]
D.P. Park, J.G. Ayres, D.T. McLeod, A.H. Mansur.
Vocal cord dysfunction treated with long-term tracheostomy: 2 case studies.
Ann Allergy Asthma Immunol., 98 (2007), pp. 591-594
[1075]
WHO. Report of the WHO-China Joint Mission on Coronavirus Disease 2019 (COVID-19) [Internet]. 2020. Disponible en: https://www.who.int/docs/default-source/coronaviruse/who-china-joint-mission-on- covid-19-final-report.pdf.
[1076]
W. Guan, Z. Ni, Y. Hu, W. Liang, C. Ou, J. He, China Medical Treatment Expert Group for Covid-19, et al.
Clinical Characteristics of Coronavirus Disease 2019 in China.
N Engl J Med., (2020 Feb 28),
[1077]
C. Huang, Y. Wang, X. Li, L. Ren, J. Zhao, Y. Hu, et al.
Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China.
Lancet., 395 (2020), pp. 497-506
[1078]
D. Wang, B. Hu, C. Hu, F. Zhu, X. Liu, J. Zhang, et al.
Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan.
China. JAMA., (2020 February),
[1079]
P. Wu, X. Hao, E.H.Y. Lau, J.Y. Wong, K.S.M. Leung, J.T. Wu, et al.
Real-time tentative assessment of the epidemiological characteristics of novel coronavirus infections in Wuhan, China, as at 22 January 2020.
Euro Surveill Bull Eur Sur Mal Transm Eur Commun Dis Bull., 25 (2020),
[1080]
J.J. Zhang, X. Dong, Y.Y. Cao, Y.D. Yuan, Y.B. Yang, Y.Q. Yan, et al.
Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan.
China. Allergy., (2020),
[1081]
F. Zhou, T. Yu, R. Du, G. Fan, Y. Liu, Z. Liu, et al.
Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study.
[1082]
R.M. Viner, O.T. Mytton, C. Bonell, G.J. Melendez-Torres, J. Ward, L. Hudson, et al.
Susceptibility to SARS-CoV-2 Infection Among Children and Adolescents Compared With Adults A Systematic Review and Meta-analysis.
JAMA Pediatrics., 175 (2021), pp. 143-156
[1083]
Y. Zhu, C.J. Bloxham, K.D. Hulme, J.E. Sinclair, Z.W.M. Tong, L.E. Steele, et al.
A meta-analysis on the role of children in SARS-CoV-2 in household transmission clusters.
Clin Infect Dis., (2020 Dec 6), pp. ciaa1825
[1084]
P. Brodin.
Why is COVID-19 so mild in children?.
Acta Paediatr., (2020),
[1085]
Y. Dong, X. Mo, Y. Hu, X. Qi, F. Jiang, Z. Jiang, et al.
Epidemiological Characteristics of 2143 Pediatric Patients With 2019 Coronavirus Disease in China.
Pediatrics, (2020 Mar 16),
[1086]
L.M. Yonker, A.M. Neilan, Y. Bartsch, A.B. Patel, J. Regan, P. Arya, et al.
Pediatric SARS-CoV-2: Clinical presentation infectivity, and immune responses.
J Pediatr, 227 (2020), pp. 45-52.e5
[1087]
T. Lupia, S. Scabini, S. Mornese, G. di Perri, F.G. de Rosa, S. Corcione.
2019 novel coronavirus (2019-nCoV) outbreak: A new challenge.
J Glob Antimicrob Resist., 21 (2020), pp. 22-27
[1088]
X. Dong, Y.Y. Cao, X.X. Lu, J.J. Zhang, H. Du, Y.Q. Yan, et al.
Eleven Faces of Coronavirus Disease 2019.
Allergy, (2020 Mar 20),
[1089]
S. Lovinsky-Desir, D.R. Deshpande, A. De, L. Murray, J.A. Stingone, A. Chan, et al.
Asthma among hospitalized patients with COVID-19 and related outcomes.
J Allergy Clin Immunol., 146 (2020), pp. 1027-1034.e4
[1090]
K.D. Chhiba, G.B. Patel, T.H.T. Vu, M.M. Chen, A. Guo, E. Kudlaty, et al.
Prevalence and characterization of asthma in hospitalized and nonhospitalized patients with COVID-19.
J Allergy Clin Immunol., 146 (2020 Aug), pp. 307-314.e4
[1091]
P.D. Terry, R.E. Heidel, R. Dhand.
Asthma in Adult Patients with COVID-19. Prevalence and Risk of Severe Disease.
Am J Respir Crit Care Med., 203 (2021 Apr 1), pp. 893-905
[1092]
K. Eger, S. Hashimoto, G.J. Braunstahl, A.T. Brinke, K.W. Patberg, A. Beukert, et al.
Poor outcome of SARS-CoV-2 infection in patients with severe asthma on biologic therapy.
Respir Med., 177 (2020 Dec 24), pp. 106287
[1093]
E.J. Williamson, A.J. Walker, K. Bhaskaran, S. Bacon, C. Bates, C.E. Morton, et al.
Factors associated with COVID-19-related death using OpenSAFELY.
Nature., 584 (2020), pp. 430-436
[1094]
S.C. Lee, K.J. Son, C.H. Han, J.Y. Jung, S.C. Park.
Impact of comorbid asthma on severity of coronavirus disease (COVID-19).
Sci Rep., 10 (2020), pp. 21805
[1095]
S. Hanon, G. Brusselle, M. Deschampheleire, R. Louis, A. Michils, et al.
COVID-19 and biologics in severe asthma: data from the Belgian Severe Asthma Registry.
Eur Respir J., 56 (2020), pp. 2002857
[1096]
M.J. Rial, M. Valverde, V. del Pozo, F.J. González-Barcala, C. Martínez-Rivera, X. Muñoz, et al.
Clinical characteristics in 545 patients with severe asthma on biological treatment during the COVID-19 outbreak.
J Allergy Clin Immunol Pract., 9 (2021), pp. 487-489.e1
[1097]
SEPAR. Recomendaciones de prevención de infección por coronavirus en las unidades de función pulmonar de los diferentes ámbitos asistenciales. Marzo de 2020. Disponible en: https://drive.google.com/file/d/1JPyCJ0_qiewcUTybJd0sxkruj-lbr8Z9/view.
[1098]
J.M. Olaguibel, I. Alobid, M. Álvarez, A. Crespo-Lessmann, J. Domínguez Ortega, F. García-Rio, et al.
Functional Examination of the Upper and Lower Airways in Asthma and Respiratory Allergic Diseases: Considerations in the Post-SARS-CoV-2 Era.
J Investig Allergol Clin Immunol., 31 (2021), pp. 17-35
[1099]
Crespo-Lessmann A, Plaza V; Consensus Group, Almonacid C, Caballero Soto ML, Cañas JA, Carretero Gracia JÁ, Cruz MJ, Dacal Rivas D, Del Camino Muñiz M, Pérez Del Llano L, Del Pozo V, López Carrasco V, Marín Trigo JM, Mateus E, Muñoz X, Olaguíbel JM, Quirce S, Ramos D, Sanz Rubio D, Sastre J, Vázquez Martín S, Vera Solsona E. Multidisciplinary consensus on sputum induction biosafety during the COVID-19 pandemic. Allergy. 2020 Dec 12. 10.1111/all.14697.
[1100]
D.S. Hui, B.K. Chow, S.S. Ng, L.C.Y. Chu, S.D. Hall, T. Gin, et al.
Exhaled Air Dispersion Distances During Noninvasive Ventilation via Different Respironics Face Masks.
Chest., 136 (2009), pp. 998-1005
[1101]
Cinesi Gómez C, Peñuelas O, Luján M, Egea C, Massa F. Recomendaciones de consenso respecto al soporte respiratorio no invasivo en el paciente adulto con insuficiencia respiratoria aguda secundaria a infección por SARSCoV-2. Arch Bronconeumol 2020. En prensa.
[1102]
E.A. Abrams, S.J. Szefler.
Managing Asthma during Coronavirus Disease-2019: An Example for Other Chronic Conditions in Children and Adolescents.
J Pediatr., 222 (2020), pp. 221-226
[1103]
S. Matsuyama, M. Kawase, N. Nao, K. Shirato, M. Ujike, W. Kamitani, et al.
The Inhaled Steroid Ciclesonide Blocks SARS-CoV-2 RNA Replication by Targeting the Viral Replication-Transcription Complex in Cultured Cells.
J Virol., 95 (2020), pp. e01648-e1720
[1104]
J.L. Izquierdo, C. Almonacid, Y. González, C. Del Rio-Bermúdez, J. Ancochea, R. Cárdenas, et al.
The Impact of COVID-19 on Patients with Asthma.
Eur Respir J., 17 (2020), pp. 2003142
[1105]
S. Ramakrishnan, D.V. Nicolau Jr., B. Langford, M. Mahdi, H. Jeffers, C. Mwasuku, et al.
Inhaled budesonide in the treatment of early COVID-19 (STOIC): a phase 2, open-label, randomised controlled trial.
Lancet Respir Med., (2021 Apr 9),
[1106]
Grupo Neumo SFH. Grupo de trabajo de patologías respiratorias (Grupo NEUMO). Interacciones entre fármacos COVID19 y asma. Sociedad Española de Farmacia Hospitalaria 2020. Disponible en https://www.sefh.es/fichadjuntos/RESUMENINTERACCIONESCOVID19asma.pdf.
[1107]
F. Villar-Alvarez, M.A. Martinez-Garcia, D. Jimenez, F. Fariñas-Guerreiro, R. Ortiz de Lejarazu-Leonardo, J.L. Lopez Campos, et al.
Open Respiratory Archives., 3 (2021), pp. 100097
[1108]
R. Agarwal.
Allergic bronchopulmonary aspergillosis.
Chest., 135 (2009), pp. 805
[1109]
R.B. Moss.
Pathophysiology and immunology of allergic bronchopulmonary aspergillosis.
Med Mycol., 43 (2005), pp. S203-S206
[1110]
S. Gago, D.W. Denning, P. Bowyer.
Pathophysiological aspects of Aspergillus colonization in disease.
Med Mycol., 57 (2019 01), pp. S219-S227
[1111]
D.W. Denning, A. Pleuvry, D.C. Cole.
Global burden of allergic bronchopulmonary aspergillosis with asthma and its complication chronic pulmonary aspergillosis in adults.
Med Mycol., 51 (2013), pp. 361-370
[1112]
V.N. Maturu, R. Agarwal.
Prevalence of Aspergillus sensitization and allergic bronchopulmonary aspergillosis in cystic fibrosis: systematic review and meta-analysis.
Clin Exp Allergy., 45 (2015), pp. 1765-1778
[1113]
R. Agarwal, D. Gupta, A.N. Aggarwal, A.K. Saxena, A. Chakrabarti, S.K. Jindal.
Clinical significance of hyperattenuating mucoid impaction in allergic bronchopulmonary aspergillosis: an analysis of 155 patients.
Chest., 132 (2007), pp. 1183-1190
[1114]
R. Agarwal, A. Chakrabarti, A. Shah, D. Gupta, J.F. Meis, R. Guleria, Denning, For the ABPA complicating asthma ISHAM working group, et al.
Allergic bronchopulmonary aspergillosis: review of literature and proposal of new diagnostic and classification criteria, 2013.
Clinical & Experimental Allergy., 43 (2013), pp. 850-873
[1115]
R. Agarwal, I.S. Sehgal, S. Dhooria, A.N. Aggarwal.
Developments in the diagnosis and treatment of allergic bronchopulmonary aspergillosis.
Expert Rev Respir Med., 10 (2016), pp. 1317-1334
[1116]
K. Asano, A. Hebisawa, T. Ishiguro, N. Takayanagi, Y. Nakamura, J. Suzuki, et al.
New clinical diagnostic criteria for allergic bronchopulmonary aspergillosis/mycosis and its validation.
J Allergy Clin Immunol., 147 (2021 Apr), pp. 1261-1268.e5
[1117]
R. Agarwal, D. Maskey, A.N. Aggarwal, B. Saikia, M. Garg, D. Gupta, et al.
Diagnostic performance of various tests and criteria employed in allergic bronchopulmonary aspergillosis: a latent class analysis.
[1118]
R. Agarwal, D. Dua, H. Choudhary, A.N. Aggarwal, I.S. Sehgal, S. Dhooria, et al.
Role of Aspergillus fumigatus specific IgG in diagnosis and monitoring treatment response in allergic bronchopulmonary aspergillosis.
Mycoses., 60 (2017), pp. 33-39
[1119]
R. Agarwal, D. Gupta, A.N. Aggarwal, A.K. Saxena, B. Saikia, A. Chakrabarti, et al.
Clinical significance of decline in serum IgE levels in allergic bronchopulmonary aspergillosis.
Respir Med., 104 (2010), pp. 204-210
[1120]
R. Agarwal, A. Khan, A.N. Aggarwal, N. Varma, M. Garg, B. Saikia, et al.
Clinical relevance of peripheral blood eosinophil count in allergic bronchopulmonary aspergillosis.
J Infect Public Health., 4 (2011), pp. 235-243
[1121]
R. Agarwal, A. Khan, M. Garg, A.N. Aggar-wal, D. Gupta.
Chest radiographic and computed tomographic manifestations in allergic bronchopulmonary aspergillosis.
World J Radiol., 4 (2012), pp. 141-150
[1122]
R. Agarwal, A. Khan, D. Gupta, A.N. Aggarwal, A.K. Saxena, A. Chakrabarti.
An alternate method of classifying allergic bronchopulmonary aspergillosis based on high-attenuation mucus.
PLoS One., 5 (2010 15), pp. e15346
[1123]
P.A. Greenberger, R.K. Bush, J.G. Demain, A. Luong, R.G. Slavin, A.P. Knutsen.
Allergic bronchopulmonary aspergillosis.
J Allergy Clin Immunol Pract., 2 (2014), pp. 703-708
[1124]
P.A. Greenberger.
Allergic bronchopulmonary aspergillosis.
J Allergy Clin Immunol., 110 (2002), pp. 685-692
[1125]
R. Agarwal, A.N. Aggarwal, S. Dhooria, I. Singh Sehgal, M. Garg, et al.
A randomised trial of glucocorticoids in acute stage allergic bronchopulmonary aspergillosis complicating asthma.
Eur Respir J., 47 (2016), pp. 490-498
[1126]
R. Agarwal, D. Gupta, A.N. Aggarwal, D. Behera, S.K. Jindal.
Allergic bronchopulmonary aspergillosis: lessons from 126 patients attending a chest clinic in north India.
Chest., 130 (2006), pp. 442-448
[1127]
T.F. Patterson, G.R. Thompson 3rd, D.W. Denning, J.A. Fishman, S. Hadley, R. Herbrecht, et al.
Practice guidelines for the diagnosis and management of aspergillosis: 2016 update by the Infectious Diseases Society of America.
Clin Infect Dis., 63 (2016), pp. 1-60
[1128]
L. Chishimba, R.M. Niven, J. Cooley, D.W. Denning.
Voriconazole and posaconazole improve asthma severity in allergic bronchopulmonary aspergillosis and severe asthma with fungal sensitization.
J Asthma., 49 (2012), pp. 423-433
[1129]
J.X. Li, L.Ch. Fan, M.H. Li, W.J. Cao, J.F. Xu.
Beneficial effects of omalizumab therapy in allergic bronchopulmonary aspergilosis: a synthesis review of published literature.
Respir Med., 122 (2017), pp. 33-42
[1130]
M.C. Altman, J. Lenington, S. Bronson, A.G. Ayars.
Combination omalizumab and mepolizumab therapy for refractory allergic bronchopulmonary aspergillosis.
J Allergy Clin Immunol Pract., 5 (2017), pp. 1137-1139
[1131]
Tolebeyan A, Mohammedi O, Vaezi Z, Animi A. Mepolizumab as posible treatment for allergic bronchopulmonary aspergilosis: a review of eight cases. Cureus. 12(8): e9684. DOI 10.7759/cureus.9684.
[1132]
Matsuura H, Fujiwara K, Omori H, Onishi K, Kuribayashi K, Mitsumune S, et al. Successful Treatment with Benralizumab for Allergic Bronchopulmonary Aspergillosis that Developed after Disastrous Heavy Rainfall in Western Japan. Intern Med Advance Publication. DOI:10.2169/internalmedicine.6217-20.
[1133]
K. Tsubouchi, M. Arimura-Omori, S. Inoue, Y. Okamatsu, K. Inoue, T. Harada, et al.
A case of allergic bronchopulmonary aspergillosis with marked peripheral blood eosinophilia successfully treated with benralizumab.
Respirator Medicine case reports., 32 (2021), pp. 101339
[1134]
S. Hirota, Y. Kobayashi, T. Ishiguro, T. Nishida, N. Kagiyama, Y. Shimizu, et al.
Allergic bronchopulmonary aspergillosis successfully treated with mepolizumab: Case report and review of the literature.
Respir Med Case Rep, 26 (2019), pp. 59-62
[1135]
L. Bernal-Rubio, B. de-la-Hoz Caballer, C. Almonacid-Sanchez, D. Gonzalez-de-Olano.
Successful treatment of allergic bronchopulmonary aspergillosis with benralizumab after no response to omalizumab.
J Investig Allergol Clin Immunol 2020 J Investig Allergol Clin Immunol., 30 (2020), pp. 378-379
[1136]
K. Tomomatsu, Y. Sugino, N. Okada, J. Tanaka, T. Oguma, K. Asano.
Rapid clearance of mepolizumab-resistant bronchial mucus plugs inallergic bronchopulmonary aspergillosis with benralizumab treatment.
Allergol Int., 69 (2020), pp. 636-638
[1137]
J.C. Jennette, R.J. Falk, P.A. Bacon, N. Basu, M.C. Cid, F. Ferrario, et al.
2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides.
Arthritis Rheum., 65 (2013 Jan), pp. 1-11
[1138]
A. Matucci, F. Nencini, E. Maggi, A. Vultaggio.
Systemic hypereosinophilic syndromes: when autoimmunity is Th2 mediated.
Curr Opin Allergy Clin Immunol., 20 (2020 Apr), pp. 175-180
[1139]
l. Raffray, l. Guillevin.
Updates for the treatment of EGPA.
Presse Med., 49 (2020 Oct), pp. 104036
[1140]
R.W. Jakes, N. Kwon, B. Nordstrom, R. Goulding, K. Fahrbach, J. Tarpey, et al.
Burden of illness associated with eosinophilic granulomatosis with polyangiitis: a systematic literature review and meta-analysis.
Clin Rheumatol., 40 (2021 Dec), pp. 4829-4836
[1141]
S. Furuta, T. Iwamoto, H. Nakajima.
Update on eosinophilic granulomatosis with polyangiitis.
Allergol Int., 68 (2019 Oct), pp. 430-436
[1142]
M. Gokhale, C.F. Bell, S. Doyle, J. Fairburn-Beech, J. Steinfeld, M.K. Van Dyke.
Prevalence of Eosinophilic Granulomatosis With Polyangiitis and Associated Health Care Utilization Among Patients With Concomitant Asthma in US Commercial Claims Database.
J Clin Rheumatol., 27 (2021 Apr 1), pp. 107-113
[1143]
A. Vaglio, I. Casazza, C. Grasselli, D. Corradi, R.A. Sinico, C. Buzio.
Churg-Strauss syndrome.
Kidney Int., 76 (2009 Nov), pp. 1006-1011
[1144]
E.Y. Wu, M.L. Hernández, J.C. Jennette, R.J. Falk.
Eosinophilic Granulomatosis with Polyangiitis: Clinical Pathology Conference and Review.
J Allergy Clin Immunol Pract., 6 (2018 Sep-Oct), pp. 1496-1504
[1145]
M. Groh, C. Pagnoux, C. Baldini, E. Bel, P. Bottero, V. Cottin, et al.
Eosinophilic granulomatosis with polyangiitis (Churg-Strauss) (EGPA) Consensus Task Force recommendations for evaluation and management.
Eur J Intern Med., 26 (2015 Sep), pp. 545-553
[1146]
A.T. Masi, G.G. Hunder, J.T. Lie, B.A. Michel, D.A. Bloch, W.P. Arend, et al.
The American College of Rheumatology 1990 criteria for the classification of Churg-Strauss syndrome (allergic granulomatosis and angiitis).
Arthritis Rheum., 33 (1990 Aug), pp. 1094-1100
[1147]
P.C. Grayson, C. Ponte, R. Suppiah, J.C. Robson, A. Craven, A. Judge, et al.
2022 American College of Rheumatology/European Alliance of Associations for Rheumatology Classification Criteria for Eosinophilic Granulomatosis with Polyangiitis.
Ann Rheum Dis., 81 (2022 Mar), pp. 309-314
[1148]
S.A. Chung, C.A. Langford, M. Maz, A. Abril, M. Gorelik, G. Guyatt, et al.
2021 American College of Rheumatology/Vasculitis Foundation Guideline for the Management of Antineutrophil Cytoplasmic Antibody-Associated Vasculitis.
Arthritis Rheumatol., 73 (2021 Aug), pp. 1366-1383
[1149]
B. Hellmich, B. Sánchez-Álamo, J.H. Shirmer, A. Berti, D. Blockmans, M.C. Cid, et al.
EULAR recommendations for the management of ANCA-associated vasculitis: 2022 Update.
Ann Rheum Dis, 0 (2023), pp. 1-18
[1150]
P. Cohen, C. Pagnoux, A. Mahr, J.P. Arène, l. Mouthon, V. Le Guern, et al.
Churg-Strauss syndrome with poor-prognosis factors: A prospective multicenter trial comparing glucocorticoids and six or twelve cyclophosphamide pulses in forty-eight patients.
Arthritis Rheum., 57 (2007 May 15), pp. 686-693
[1151]
M. Gayraud, l. Guillevin, P. Cohen, F. Lhote, P. Cacoub, P. Deblois, et al.
Treatment of good-prognosis polyarteritis nodosa and Churg-Strauss syndrome: comparison of steroids and oral or pulse cyclophosphamide in 25 patients. French Cooperative Study Group for Vasculitides.
Br J Rheumatol., 36 (1997 Dec), pp. 1290-1297
[1152]
V. Cottin, C. Khouatra, R. Dubost, J.C. Glérant, J.F. Cordier.
Persistent airflow obstruction in asthma of patients with Churg-Strauss syndrome and long-term follow-up.
Allergy., 64 (2009 Apr), pp. 589-595
[1153]
A.J. Mohammad, A. Hot, F. Arndt, F. Moosig, M.J. Guerry, N. Amudala, et al.
Rituximab for the treatment of eosinophilic granulomatosis with polyangiitis (Churg-Strauss).
Ann Rheum Dis., 75 (2016 Feb), pp. 396-401
[1154]
Centro de información online de medicamentos de la AEMPS - CIMA. Ficha técnica Nucala 100 MG polvo para solución inyectable. Disponible en https://cima.aemps.es/cima/dochtml/ft/1151043001/FT_1151043001.html.
[1155]
M.E. Wechsler, P. Akuthota, D. Jayne, P. Khoury, A. Klion, C.A. Langford, et al.
Mepolizumab or placebo for eosinophilic granulomatosis with polyangiitis.
N Engl J Med., 376 (2017), pp. 1921-1932
[1156]
S.J. Ackerman, B.S. Bochner.
Mechanisms of eosinophilia in the pathogenesis of hypereosinophilic disorders.
Immunol Allergy Clin North Am., 27 (2007), pp. 357-375
[1157]
C. Curtis, P. Ogbogu.
Hypereosinophilic syndrome.
Clin Rev Allergy Immunol., 50 (2016), pp. 240-251
[1158]
W. Shomali, J. Gotlib.
World Health Organization-defined eosinophilic disorders: 2019 update on diagnosis, risk stratification, and management.
Am J Hematol., 94 (2019), pp. 1149-1167
[1159]
J.E. Kahn, M. Groh, G. Lefevre.
(A critical appraisal of) classification of hypereosinophilic disorders.
Front Med (Lausanne)., 4 (2017 Dec 5), pp. 216
[1160]
G. Requena, J. Logie, D.C. Gibbons, J. Steinfeld, M.K. Van Dyke.
The increasing incidence and prevalence of hypereosinophilic syndrome in the United Kingdom.
Immun Inflamm Dis., 9 (2021 Dec), pp. 1447-1451
[1161]
P. Valent, A.D. Klion, H.P. Horny, F. Roufosse, J. Gotlib, P.F. Weller, et al.
Contemporary consensus proposal on criteria and classification of eosinophilic disorders and related syndromes.
J Allergy Clin Immunol., 130 (2012), pp. 607-612.e9
[1162]
P.U. Ogbogu, B.S. Bochner, J.H. Butterfield, G.J. Gleich, J. Huss-Marp, J.E. Kahn, et al.
Hypereosinophilic syndrome: a multicenter, retrospective analysis of clinical characteristics and response to therapy.
J Allergy Clin Immunol., 124 (2009), pp. 1319-1325.e3
[1163]
A. Wolf, D. Dedman, J. Campbell, H. Booth, D. Lunn, J. Chapman, et al.
Data resource profile: Clinical Practice Research Datalink (CPRD) Aurum.
Int J Epidemiol., 48 (2019 Dec 1),
[1164]
H.R. Noh, G.G. Magpantay.
Hypereosinophilic syndrome.
Allergy Asthma Proc., 38 (2017), pp. 78-81
[1165]
A.D. Klion.
How I treat hypereosinophilic syndromes.
Blood., 126 (2015), pp. 1069-1077
[1166]
J. Gotlib.
World Health Organization-defined eosinophilic disorders: 2017 update on diagnosis, risk stratification, and management.
Am J Hematol, 92 (2017 Nov), pp. 1243-1259
[1167]
S.G. Plötz, H.U. Simon, U. Darsow, D. Simon, E. Vassina, S. Yousefi, et al.
Use of an anti-interleukin-5 antibody in the hypereosinophilic syndrome with eosinophilic dermatitis.
N Engl J Med., 349 (2003 Dec 11), pp. 2334-2339
[1168]
J.K. Garrett, S.C. Jameson, B. Thomson, M.H. Collins, L.E. Wagoner, D.K. Freese, et al.
Anti-interleukin-5 (mepolizumab) therapy for hypereosinophilic syndromes.
J Allergy Clin Immunol., 113 (2004), pp. 115-119
[1169]
M.E. Rothenberg, A.D. Klion, F.E. Roufosse, J.E. Kahn, P.F. Weller, H.U. Simon, et al.
Treatment of patients with the hypereosinophilic syndrome with mepolizumab.
N Engl J Med., 358 (2008), pp. 1215-1228
[1170]
F. Roufosse, A. de Lavareille, l. Schandene, E. Cogan, A. Georgelas, l. Wagner, et al.
Mepolizumab as a corticosteroid-sparing agent in lymphocytic variant hypereosinophilic syndrome.
J Allergy Clin Immunol., 126 (2010), pp. 828-835.e3
[1171]
F.E. Roufosse, J.E. Kahn, G.J. Gleich, L.B. Schwartz, A.D. Singh, L.J. Rosenwasser, et al.
Long-term safety of mepolizumab for the treatment of hypereosinophilic syndromes.
J Allergy Clin Immunol., 131 (2013), pp. 461-467.e1-5
[1172]
F. Roufosse, J.E. Kahn, M.E. Rothenberg, A.J. Wardlaw, A.D. Klion, S.Y. Kirby, et al.
Efficacy and safety of mepolizumab in hypereosinophilic syndrome: a phase III, randomized, placebo-controlled trial.
J Allergy Clin Immunol., 146 (2020 Dec), pp. 1397-1405
[1173]
G.J. Gleich, F. Roufosse, G. Chupp, S. Faguer, B. Walz, A. Reiter, et al.
Safety and Efficacy of Mepolizumab in Hypereosinophilic Syndrome: An Open-Label Extension Study.
J Allergy Clin Immunol Pract., 9 (2021 Dec), pp. 4431-4440.e1
[1174]
F.J. Álvarez, J. Delgado, J.A. Quintano.
Continuidad asistencial en el asma bronquial.
Espacioasma., 8 (2015), pp. 27-28
[1175]
J. Haggerty, R. Reid, G.K. Freeman, B.H. Starfield, C.E. Adair, R. McKendry.
Continuity of care: a multidisciplinary review.
BMJ., 327 (2003), pp. 1219-1221
[1176]
K.R. Murphy, E.O. Meltzer, M.S. Blaiss, R.A. Nathan, S.W. Stoloff, D.E. Doherty.
Asthma management and control in the United States: results of the 2009 Asthma Insight and Management survey.
Allergy Asthma Proc., 33 (2012), pp. 54-64
[1177]
K. Hasegawa, A.F. Sullivan, E. Tovar, T.J. Gaeta, C. Fee, S.J. Turner, Multicenter Airway Research Collaboration-36 Investigators, et al.
A multicenter observational study of US adults with acute asthma: who are the frequent users of the emergency department?.
J Allergy Clin Immunol Pract., 2 (2014), pp. 733-740
[1178]
Royal College of Physicians; National Review of Asthma Deaths; 2014. Why asthma still kills: the National Review of Asthma Deaths (NRAD); Confidential Enquiry Report. Available from: https://www.rcplondon.ac.uk/projects/outputs/why-asthma-still-kills.
[1179]
D. Orozco-Beltrán, C. Carratalá-Munuera, J.M. Arriero, P. Campo, E. Martínez-Moragón, J. Molina, Working Group for the Consensus Document on the Management of Severe Asthma in Adults in Primary Health Care, et al.
Management and referral of patients with severe and poorly controlled asthma in primary care.
Fam Pract., 33 (2016), pp. 678-683
[1180]
V. Plaza, P. Rodríguez del Río, F. Gómez, A. López, J. Molina, J.A. Quintano, et al.
Identification of gaps in the clinical patient care of asthma in Spain. Results of the OPTIMA-GEMA survey.
An Sist Sanit Navar., 39 (2016), pp. 181-201
[1181]
L. Pérez de Llano, E. Martínez-Moragón, V. Plaza, A. Trisán, C.A. Sánchez, F.J. Callejas, et al.
Unmet therapeutic goals and potential treatable traits in a population of patients with severe uncontrolled asthma in Spain.
ENEAS study. Respir Med., 151 (2019), pp. 49-54
[1182]
G.K. Freeman, F. Olesen, P. Hjortdahl.
Continuity of care: an essential element of modern general practice?.
Family Practice., 20 (2003), pp. 623-627
[1183]
M.L. Vázquez, I. Vargasa, R. Nuño, N. Toro.
Organizaciones sanitarias integradas y otros ejemplos de colaboración entre proveedores.
Informe SESPAS 2012. Gac Sanit., 26 (2012), pp. 94-101
[1184]
A.L. Dima, M. de Bruin, E. van Ganse, on behalf of the ASTRO-LAB group.
Mapping the Asthma Care Process: Implications for Research and Practice.
J. Allergy Clin Immunol., 4 (2016), pp. 868-876
[1185]
A. Capelastegui.
Cómo mejorar la calidad de la asistencia al paciente con asma. Rev.
Asma., 2 (2017), pp. 23-28
[1186]
T. Haahtela, L.E. Tuomisto, A. Pietinalho, T. Klaukka, M. Erhola, M. Kaila, et al.
A 10 year asthma programme in Finland: major change for the better.
Thorax., 61 (2006), pp. 663-670
[1187]
S. Mangiapane, M. Schulz, S. Mühlig, P. Ihle, I. Schubert, H.-C. Waldmann.
Community Pharmacy-Based Pharmaceutical Care for Asthma Patients.
Annals of Pharmacotherapy, 39 (2005), pp. 1817-1822
[1188]
N. Mubarak, E.M. Hatah, T.M. Khan, C.S. Zin.
A systematic review and meta-analysis of the impact of collaborative practice between community pharmacist and general practitioner on asthma management.
Journal of Asthma and Allergy., 12 (2019), pp. 109-153
[1189]
V. Plaza (coord.).
GEMA4.0. Guía Española para el Manejo del Asma.
Arch Bronconeumol, 51 (2015), pp. 2-54
[1190]
E. Martínez-Moragón, M. Palop, A. de Diego, J. Serra, C. Pellicer, P. Casán, ASMACOST Study Group, et al.
Factors affecting quality of life of asthma patients in Spain: the importance of patient education.
Allergol Immunopathol (Madr)., 42 (2014), pp. 476-484
[1191]
A. Kouri, A. Kaplan, L.P. Boulet, S. Gupta.
New evidence-based tool to guide the creation of asthma action plans for adults.
Can Fam Physician., 65 (2019), pp. 103-106
[1192]
S. Evans-Lacko, M. Jarrett, P. McCrone, G. Thornicroft.
Facilitators and barriers to implementing clinical care pathways.
BMC Health Serv Res., 10 (2010), pp. 182
[1193]
M. Blanco, J. Delgado, J. Molina, J.T. Gómez, F. Gómez, F.J. Álvarez, et al.
Referral criteria for asthma: Consensus document.
J Investig Allergol Clin Immunol., (2019 Apr 1),
[1194]
M. Engelkes, H.M. Janssens, J.C. de Jongste, M.C. Sturkenboom, K.M. Verhamme.
Medication adherence and the risk of severe asthma exacerbations: a systematic review.
Eur Respir J., 45 (2015), pp. 396-407
[1195]
M. George, B. Bender.
New insights to improve treatment adherence in asthma and COPD.
Patient Prefer Adherence., 13 (2019), pp. 1325-1334
[1196]
Ministerio de Sanidad, Consumo y Bienestar Social. Marco Estratégico para la Atención Primaria y Comunitaria. Estrategia C. Objetivos C.1 y C.2. Plan de acción C.2.1. 10 de abril de 2019. Disponible en http://www.mscbs.gob.es/profesionales/proyectosActividades/docs/Marco_Estrategico_APS_25Abril_2019.pdf.
[1197]
Instituto de Información Sanitaria. Agencia de Calidad del Sistema Nacional de Salud (SNS). El sistema de historia clínica digital del SNS. Capítulo 3. Principios estratégicos. Apartado 3.1. Utilidad para profesionales y ciudadanos. Disponible en https://www.mscbs.gob.es/organizacion/sns/planCalidadSNS/docs/HCDS-NS_Castellano.pdf.
[1198]
L. Pérez de Llano, R. Villoro, M. Merino, M.C. Gómez-Neira, M. Camino, A. Hidalgo.
Coste-efectividad de una unidad monográfica de asma.
Arch Bronconeumol., 52 (2016), pp. 196-203
[1199]
C. Miles, E. Arden-Close, M. Thomas, A. Bruton, L. Yardley, M. Hankins, et al.
Barriers and facilitators of effective self-management in asthma: systematic review and thematic synthesis of patient and healthcare professional views.
NPJ Prim Care Respir Med., 27 (2017), pp. 57
[1200]
K. Harris, D. Kneale, T.J. Lasserson, V.M. McDonald, J. Grigg, J. Thomas.
School-based self-management interventions for asthma in children and adolescents: a mixed methods systematic review.
Cochrane Database of Systematic Reviews., (2019), pp. CD011651
[1201]
S. O’Connell, V.J. McCarthy, E. Savage.
Self-management support preferences of people with asthma or chronic obstructive pulmonary disease: A systematic review and meta-synthesis of qualitative studies.
[1202]
Banco de datos de asma. SEPAR. Disponible en https://www.bancodatosasma.com/.
[1203]
A. Sá-Sousa, J.A. Fonseca, A.M. Pereira, A. Ferreira, A. Arrobas, A. Mendes, et al.
The Portuguese Severe Asthma Registry: Development, Features, and Data Sharing Policies.
Biomed Res Int., (2018), pp. 1495039
[1204]
Almonacid C. Telemedicina y Asma. Revisiones asma; 9. Disponible en: http://www.revisionesasma.com/telemedicina-y-asma/.
[1205]
S. Gupta, C. Price, G. Agarwal, D. Chan, S. Goel, L.P. Boulet, et al.
The Electronic Asthma Management System (eAMS) improves primary care asthma management.
Eur Respir J., 53 (2019), pp. 1802241
[1206]
S. Erickson, I. Tolstykh, J.V. Selby, G. Mendoza, C. Iribarren, M.D. Eisner.
The impact of allergy and pulmonary specialist care on emergency asthma utilization in a large managed care organization.
Health Serv Res., 40 (2005), pp. 1443-1465
[1207]
M. Schatz, R.S. Zeiger, D. Mosen, A.J. Apter, W.M. Vollmer, T.B. Stibolt, et al.
Improved asthma outcomes from allergy specialist care: a population-based cross-sectional analysis.
J Allergy Clin Immunol., 116 (2005), pp. 1307-1313
[1208]
M.E. Wechsler.
Managing asthma in primary care: putting new guideline recommendations into context.
Mayo Clin Proc., 84 (2009), pp. 707-717
[1209]
D. Price, L. Bjermer, D.A. Bergin, R. Martinez.
Asthma referrals: a key component of asthma management that needs to be addressed.
J Asthma Allergy., 10 (2017), pp. 209-223
[1210]
J.A. Carretero Gracia, F. Rodríguez Fernández, J.T. Gómez Sáenz, J. Molina París, F. Gómez Ruiz, V. López Carrasco, et al.
Criterios de derivación en asma: Actualización documento de consenso.
Open Respir Arch., 3 (2021), pp. 4
[1211]
A.N. Van der Meer, H. Pasam, W. Kempenaar-Okkema, J.A. Pelinck, M. Schutten, H. Storm, et al.
A 1-day visit in a severe asthma centre: effect on asthma control, quality of life and healthcare use.
Eur Respir J., 4883 (2016), pp. 726-733
[1212]
C. Cisneros, R.M. Díaz-Campos, N. Marina, C. Melero, A. Padilla, S. Pascual, on behalf of the DUMA Study Group, et al.
Accreditation of specialized asthma units for adults in Spain: an applicable experience for the management of difficult-to-control asthma.
J Asthma Allergy., 10 (2017), pp. 163-169
[1213]
Acreditación de Unidades de Asma Grave. Disponible en: https://www.seaic.org/profesionales/acreditacion-unidades-de-asma-grave.
[1214]
M. Thomas.
Why aren’t we doing better in asthma: Time for personalised medicine?.
NPJ Prim Care Respir Med., 25 (2015), pp. 15004
[1215]
V.M. McDonald, A.E. Vertigan, P.G. Gibson.
How to set up a severe asthma service.
Respirology., 16 (2011), pp. 900-911
[1216]
H. Burke, J. Davis, S. Evans, L. Flower, A. Tan, R.J. Kurukulaaratchy.
A multidisciplinary team case management approach reduces the burden of frequent asthma admissions.
ERJ Open Res., 2 (2016),
[1217]
J. Kryworuchko, D. Stacey, N. Bai, I.D. Graham.
Twelve years of clinical practice guideline development, dissemination and evaluation in Canada (1994 to 2005).
Implement Sci., 4 (2009), pp. 49
[1218]
V. Plaza, J. Bellido-Casado, P. Alonso-Coello, G. Rodrigo.
Guías de Práctica Clínica para el asma. Luces y sombras.
Arch Bronconeumol., 45 (2009), pp. 25-29
[1219]
L.P. Boulet, M.J. FitzGerald, M.L. Levy, A.A. Cruz, S. Pedersen, T. Haahtela, et al.
A guide to the translation of the Global Initiative for Asthma (GINA) strategy into improved care.
Eur Respir J., 39 (2012), pp. 1220-1229
[1220]
A. Acuna-Izcaray, E. Sánchez-Angarita, V. Plaza, G. Rodrigo, M. Montes de Oca, I. Gich, et al.
Quality assessment of asthma clinical practice guidelines: a systematic appraisal.
Chest., 144 (2013), pp. 390-397
[1221]
I.D. Graham, J. Logan, M.B. Harrison, S.E. Straus, J. Tetroe, W. Caswell, et al.
Lost in knowledge translation: time for a map?.
J Contin Educ Health Prof., 26 (2006), pp. 13-24
[1222]
E.A. Miller.
Solving the disjuncture between research and practice, telehealth trends in the 21st Century.
Health Policy., 82 (2007), pp. 133-141
[1223]
B. Chongmelaxme, S. Lee, T. Dhippayom, S. Saokaew, N. Chaiyakunapruk, P. Dilokthornsakul.
The Effects of Telemedicine on Asthma Control and Patients’ Quality of Life in Adults: A Systematic Review and Meta-analysis.
J Allergy Clin Immunol Pract, 7 (2019), pp. 199-216.e11
[1224]
G. McLean, E. Murray, R. Band, K.R. Moffat, P. Hanlon, A. Bruton, et al.
Interactive digital interventions to promote self-management in adults with asthma: systematic review and meta-analysis.
BMC Pulm Med., 16 (2016), pp. 83
Article options
Tools
Supplemental materials
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos