1
|
Wei W, Xie Z, Yan J, Luo R, He J. Progress in research on induced sputum in asthma: a narrative review. J Asthma 2025; 62:189-204. [PMID: 39290080 DOI: 10.1080/02770903.2024.2395383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/01/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE To explore the clinical significance of induced sputum in asthma through a retrospective analysis of induced sputum in patients with asthma. DATA SOURCES The data and references cited in this article were obtained from PubMed, Sci-Hub, and Web of Science. STUDY SELECTION Observational studies with reliable data were selected. CONCLUSIONS The cytological count, -omics, and pathogen detection of induced sputum are helpful for the clinical diagnosis of asthma and in guiding medication choices.
Collapse
Affiliation(s)
- Wenjie Wei
- Department of Respiratory and Critical Care Medicine, Hunan University of Medicine General Hospital, Huaihua, People's Republic of China
| | - Zhihao Xie
- Pediatric Department, The First Affiliated Hospital of Hunan University of Medicine, Huaihua, People's Republic of China
| | - Jun Yan
- Pediatric Department, The First Affiliated Hospital of Hunan University of Medicine, Huaihua, People's Republic of China
| | - Renrui Luo
- Department of Respiratory and Critical Care Medicine, Hunan University of Medicine General Hospital, Huaihua, People's Republic of China
| | - Jianbin He
- Department of Respiratory and Critical Care Medicine, Hunan University of Medicine General Hospital, Huaihua, People's Republic of China
| |
Collapse
|
2
|
Najafi Z, Rahmanian-Devin P, Baradaran Rahimi V, Nokhodchi A, Askari VR. Challenges and opportunities of medicines for treating tendon inflammation and fibrosis: A comprehensive and mechanistic review. Fundam Clin Pharmacol 2024; 38:802-841. [PMID: 38468183 DOI: 10.1111/fcp.12999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 01/20/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Tendinopathy refers to conditions characterized by collagen degeneration within tendon tissue, accompanied by the proliferation of capillaries and arteries, resulting in reduced mechanical function, pain, and swelling. While inflammation in tendinopathy can play a role in preventing infection, uncontrolled inflammation can hinder tissue regeneration and lead to fibrosis and impaired movement. OBJECTIVES The inability to regulate inflammation poses a significant limitation in tendinopathy treatment. Therefore, an ideal treatment strategy should involve modulation of the inflammatory process while promoting tissue regeneration. METHODS The current review article was prepared by searching PubMed, Scopus, Web of Science, and Google Scholar databases. Several treatment approaches based on biomaterials have been developed. RESULTS This review examines various treatment methods utilizing small molecules, biological compounds, herbal medicine-inspired approaches, immunotherapy, gene therapy, cell-based therapy, tissue engineering, nanotechnology, and phototherapy. CONCLUSION These treatments work through mechanisms of action involving signaling pathways such as transforming growth factor-beta (TGF-β), mitogen-activated protein kinases (MAPKs), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), all of which contribute to the repair of injured tendons.
Collapse
Affiliation(s)
- Zohreh Najafi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouria Rahmanian-Devin
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Nokhodchi
- Lupin Pharmaceutical Research Center, 4006 NW 124th Ave., Coral Springs, Florida, Florida, 33065, USA
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Brighton, BN1 9QJ, UK
| | - Vahid Reza Askari
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Martín-Cruz L, Benito-Villalvilla C, Sirvent S, Angelina A, Palomares O. The Role of Regulatory T Cells in Allergic Diseases: Collegium Internationale Allergologicum (CIA) Update 2024. Int Arch Allergy Immunol 2024; 185:503-518. [PMID: 38408438 DOI: 10.1159/000536335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 01/16/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Allergy represents a major health problem of increasing prevalence worldwide with a high socioeconomic impact. Our knowledge on the molecular mechanisms underlying allergic diseases and their treatments has significantly improved over the last years. The generation of allergen-specific regulatory T cells (Tregs) is crucial in the induction of healthy immune responses to allergens, preventing the development and worsening of allergic diseases. SUMMARY In the last decades, intensive research has focused on the study of the molecular mechanisms involved in Treg development and Treg-mediated suppression. These mechanisms are essential for the induction of sustained tolerance by allergen-specific immunotherapy (AIT) after treatment discontinuation. Compelling experimental evidence demonstrated altered suppressive capacity of Tregs in patients suffering from allergic rhinitis, allergic asthma, food allergy, or atopic dermatitis, as well as the restoration of their numbers and functionality after successful AIT. KEY MESSAGE The better understanding of the molecular mechanisms involved in Treg generation during allergen tolerance induction might well contribute to the development of novel strategies for the prevention and treatment of allergic diseases.
Collapse
Affiliation(s)
- Leticia Martín-Cruz
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University, Madrid, Spain
| | - Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University, Madrid, Spain
| | - Sofía Sirvent
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| |
Collapse
|
4
|
Sharma S, Leonard A, Phoenix K, Chang HY, Wang J, Hansel S. Systemically Administered Anti-uPAR Antibody Plasma and Lung ELF Pharmacokinetics Characterized by Minimal Lung PBPK Model. AAPS PharmSciTech 2023; 24:236. [PMID: 37989972 DOI: 10.1208/s12249-023-02689-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 10/18/2023] [Indexed: 11/23/2023] Open
Abstract
Antibody-based therapeutics have recently gained keen attention for the treatment of pulmonary indications. However, systemically administered antibody exposure in the lungs needs to be better understood and remains a topic of interest. In this study, we evaluated the exposure of two different uPAR (urokinase-type plasminogen activator receptor) targeting full-length monoclonal IgGs in plasma and lung epithelial lining fluid (ELF) of mice after IP and IV administration. Antibody AK17 exhibited linear pharmacokinetics (PK) in plasma and ELF at 3 and 30 mg/kg single IV dose. The average plasma and ELF half-lives for AK17 and AK21 ranged between ~321-411 h and ~230-345 h, respectively, indicating sustained systemic and lung exposure of antibodies. The average ELF to the plasma concentration ratio of antibodies was ~0.01 and ~0.03 with IP and IV dosing, respectively, over 2 weeks post single dose. We simultaneously characterized plasma and ELF PK of antibody in mice by developing a minimal lung PBPK model for antibody. This model reasonably captured the plasma and ELF PK data while estimating three parameters. The model accounts for the convective transport of antibody into the tissues via blood and lymph flow. FcRn-mediated transcytosis was incorporated into the model for antibody distribution across the lung epithelial barrier. This model serves as a platform to predict the pulmonary PK of systemically administered antibodies and to support optimal dose selection for desired exposure in the lungs as the site of action.
Collapse
Affiliation(s)
- Sharad Sharma
- Biotherapeutics Discovery, Research & Development, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd./P.O. Box 368, Ridgefield, Connecticut, 06877-0368, USA.
| | - Antony Leonard
- Biotherapeutics Discovery, Research & Development, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd./P.O. Box 368, Ridgefield, Connecticut, 06877-0368, USA
| | - Kathryn Phoenix
- Biotherapeutics Discovery, Research & Development, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd./P.O. Box 368, Ridgefield, Connecticut, 06877-0368, USA
| | - Hsueh Yuan Chang
- Biotherapeutics Discovery, Research & Development, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd./P.O. Box 368, Ridgefield, Connecticut, 06877-0368, USA
| | - Jun Wang
- Biotherapeutics Discovery, Research & Development, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd./P.O. Box 368, Ridgefield, Connecticut, 06877-0368, USA
| | - Steven Hansel
- Biotherapeutics Discovery, Research & Development, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd./P.O. Box 368, Ridgefield, Connecticut, 06877-0368, USA
| |
Collapse
|
5
|
Wen J, Wang C, Xia J, Giri M, Guo S. Relationship between serum iron and blood eosinophil counts in asthmatic adults: data from NHANES 2011-2018. Front Immunol 2023; 14:1201160. [PMID: 37731511 PMCID: PMC10507334 DOI: 10.3389/fimmu.2023.1201160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND So far, quite a few studies have revealed that systemic iron levels are related to asthmatic inflammatory reactions. And most studies have focused on the correlation between systemic iron levels and asthma, with inconsistent findings. Yet, few studies have investigated the connection between serum iron and blood eosinophil counts. Hence, we have explored the connection between serum iron and blood eosinophil counts in asthmatics by utilizing data from NHANES. METHODS A total of 2549 individuals were included in our study after screening NHANES participants from 2011 to 2018. The linear regression model and XGBoost model were used to discuss the potential connection. Linear or nonlinear association was further confirmed by the generalized additive model and the piecewise linear regression model. And we also performed stratified analyses to figure out specific populations. RESULTS In the multivariable linear regression models, we discovered that serum iron levels were inversely related to blood eosinophil counts in asthmatic adults. Simultaneously, we found that for every unit increase in serum iron (umol/L), blood eosinophil counts reduced by 1.41/uL in model 3, which adjusted for all variables excluding the analyzed variables. Furthermore, the XGBoost model of machine learning was applied to assess the relative importance of chosen variables, and it was determined that vitamin C intake, age, vitamin B12 intake, iron intake, and serum iron were the five most important variables on blood eosinophil counts. And the generalized additive model and piecewise linear regression model further verify this linear and inverse association. CONCLUSION Our investigation discovered that the linear and inverse association of serum iron with blood eosinophil counts in asthmatic adults, indicating that serum iron might be related to changes in the immunological state of asthmatics. Our work offers some new thoughts for next research on asthma management and therapy. Ultimately, we hope that more individuals become aware of the role of iron in the onset, development, and treatment of asthma.
Collapse
Affiliation(s)
- Jun Wen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Changfen Wang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Jing Xia
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Mohan Giri
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Shuliang Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Wang J, Wang P, Shao Y, He D. Advancing Treatment Strategies: A Comprehensive Review of Drug Delivery Innovations for Chronic Inflammatory Respiratory Diseases. Pharmaceutics 2023; 15:2151. [PMID: 37631365 PMCID: PMC10458134 DOI: 10.3390/pharmaceutics15082151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Chronic inflammatory respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis, present ongoing challenges in terms of effective treatment and management. These diseases are characterized by persistent inflammation in the airways, leading to structural changes and compromised lung function. There are several treatments available for them, such as bronchodilators, immunomodulators, and oxygen therapy. However, there are still some shortcomings in the effectiveness and side effects of drugs. To achieve optimal therapeutic outcomes while minimizing systemic side effects, targeted therapies and precise drug delivery systems are crucial to the management of these diseases. This comprehensive review focuses on the role of drug delivery systems in chronic inflammatory respiratory diseases, particularly nanoparticle-based drug delivery systems, inhaled corticosteroids (ICSs), novel biologicals, gene therapy, and personalized medicine. By examining the latest advancements and strategies in these areas, we aim to provide a thorough understanding of the current landscape and future prospects for improving treatment outcomes in these challenging conditions.
Collapse
Affiliation(s)
- Junming Wang
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; (J.W.); (P.W.); (Y.S.)
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Pengfei Wang
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; (J.W.); (P.W.); (Y.S.)
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Yiru Shao
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; (J.W.); (P.W.); (Y.S.)
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
| | - Daikun He
- Center of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China; (J.W.); (P.W.); (Y.S.)
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai 201508, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai 201508, China
- Department of General Practice, Jinshan Hospital, Fudan University, Shanghai 201508, China
- Department of General Practice, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
7
|
Striz I, Golebski K, Strizova Z, Loukides S, Bakakos P, Hanania N, Jesenak M, Diamant Z. New insights into the pathophysiology and therapeutic targets of asthma and comorbid chronic rhinosinusitis with or without nasal polyposis. Clin Sci (Lond) 2023; 137:727-753. [PMID: 37199256 PMCID: PMC10195992 DOI: 10.1042/cs20190281] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/22/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023]
Abstract
Asthma and chronic rhinosinusitis with nasal polyps (CRSwNP) or without (CRSsNP) are chronic respiratory diseases. These two disorders often co-exist based on common anatomical, immunological, histopathological, and pathophysiological basis. Usually, asthma with comorbid CRSwNP is driven by type 2 (T2) inflammation which predisposes to more severe, often intractable, disease. In the past two decades, innovative technologies and detection techniques in combination with newly introduced targeted therapies helped shape our understanding of the immunological pathways underlying inflammatory airway diseases and to further identify several distinct clinical and inflammatory subsets to enhance the development of more effective personalized treatments. Presently, a number of targeted biologics has shown clinical efficacy in patients with refractory T2 airway inflammation, including anti-IgE (omalizumab), anti-IL-5 (mepolizumab, reslizumab)/anti-IL5R (benralizumab), anti-IL-4R-α (anti-IL-4/IL-13, dupilumab), and anti-TSLP (tezepelumab). In non-type-2 endotypes, no targeted biologics have consistently shown clinical efficacy so far. Presently, multiple therapeutical targets are being explored including cytokines, membrane molecules and intracellular signalling pathways to further expand current treatment options for severe asthma with and without comorbid CRSwNP. In this review, we discuss existing biologics, those under development and share some views on new horizons.
Collapse
Affiliation(s)
- Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Subdivision of Allergology and Clinical Immunology, Institute for Postgraduate Education in Medicine, Prague, Czech Republic
| | - Kornel Golebski
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands
| | - Zuzana Strizova
- Institute of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Stelios Loukides
- Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros Bakakos
- First Respiratory Medicine Department, National and Kapodistrian University of Athens, Athens, Greece
| | - Nicola A. Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Milos Jesenak
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital in Martin, Slovakia
- Department of Pediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital in Martin, Slovakia
- Department of Clinical Immunology and Allergology, University Hospital in Martin, Slovakia
| | - Zuzana Diamant
- Department of Microbiology Immunology and Transplantation, KU Leuven, Catholic University of Leuven, Belgium
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Skane University Hospital, Lund University, Lund, Sweden
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
8
|
Therapeutic strategies targeting pro-fibrotic macrophages in interstitial lung disease. Biochem Pharmacol 2023; 211:115501. [PMID: 36921632 DOI: 10.1016/j.bcp.2023.115501] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the representative phenotype of interstitial lung disease where severe scarring develops in the lung interstitium. Although antifibrotic treatments are available and have been shown to slow the progression of IPF, improved therapeutic options are still needed. Recent data indicate that macrophages play essential pro-fibrotic roles in the pathogenesis of pulmonary fibrosis. Historically, macrophages have been classified into two functional subtypes, "M1" and "M2," and it is well described that "M2" or "alternatively activated" macrophages contribute to fibrosis via the production of fibrotic mediators, such as TGF-β, CTGF, and CCL18. However, highly plastic macrophages may possess distinct functions and phenotypes in the fibrotic lung environment. Thus, M2-like macrophages in vitro and pro-fibrotic macrophages in vivo are not completely identical cell populations. Recent developments in transcriptome analysis, including single-cell RNA sequencing, have attempted to depict more detailed phenotypic characteristics of pro-fibrotic macrophages. This review will outline the role and characterization of pro-fibrotic macrophages in fibrotic lung diseases and discuss the possibility of treating lung fibrosis by preventing or reprogramming the polarity of macrophages. We also utilized a systematic approach to review the literature and identify novel and promising therapeutic agents that follow this treatment strategy.
Collapse
|
9
|
Inotodiol, an antiasthmatic agent with efficacy and safety, preferentially impairs membrane-proximal signaling for mast cell activation. Int Immunopharmacol 2023; 117:109854. [PMID: 36812673 DOI: 10.1016/j.intimp.2023.109854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/23/2023] [Accepted: 02/04/2023] [Indexed: 02/22/2023]
Abstract
While inhaled corticosteroids (ICSs) are the mainstay of asthma treatment, due to compliance, drug safety, and resistance issues, new medications to replace ICSs are in high demand. Inotodiol, a fungal triterpenoid, showed a unique immunosuppressive property with a preference for mast cells. It exerted a mast cell-stabilizing activity equally potent to dexamethasone in mouse anaphylaxis models when orally administered in a lipid-based formulation, upgrading bioavailability. However, it was four to over ten times less effective in suppressing other immune cell subsets, depending on the subsets, than dexamethasone showing invariably potent inhibition. Accordingly, inotodiol affected the membrane-proximal signaling for activating mast cell functions more profoundly than other subsets. Inotodiol also effectively prevented asthma exacerbation. Importantly, considering the no-observed-adverse-effect level of inotodiol was over 15 times higher than dexamethasone, its therapeutic index would be at least eight times better,implying that inotodiol is a viable option for replacing CSs in treating asthma.
Collapse
|
10
|
Savlevich EL, Zurochka AV, Kurbacheva OM, Egorov VI, Shilovskiy IP, Mitrofanova ES, Lyubimova EV. [Pleiomorphism of the cytokine profile in nasal polyp tissue depending on the phenotype of chronic rhinosinusitis with nasal polyps]. Vestn Otorinolaringol 2023; 88:50-56. [PMID: 36867144 DOI: 10.17116/otorino20228801150] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
UNLABELLED Clinical phenotypes of chronic rhinosinusitis with nasal polyps (CRSwNP) are characterized with different inflammation patterns of mRNA expression of cytokines and depend on presence of allergic rhinitis (AR), atopic bronchial asthma (aBA) or nonatopic bronchial asthma (nBA). OBJECTIVE To compare inflammation response in patients with different phenotypes of CRSwNP according to level secretion of the key cytokines in nasal polyp tissue. MATERIAL AND METHODS 292 patients with CRSwNP were divided into four phenotypes: group 1 - CRSwNP without respiratory allergy (RA) and without BA; group 2a - CRSwNP+ AR with aBA; group 2b - CRSwNP+AR without aBA; group 3 - CRSwNP+nBA. Control group (n=36) included patients with hypertrophic rhinitis without atopy or BA. Using multiplex assay we defined the level of IL-1β, IL-4, IL-5, IL-6, IL-13, IFN-γ, TGF-β1, TGF-β2, TGF-β3 in nasal polyp tissue. RESULTS The evaluation of cytokines levels in nasal polyps in different CRSwNP phenotypes showed a pleiotropy of different cytokine secretion depending on different comorbid pathology. In control group we estimated the lowest levels of all detected cytokines in comparison with other CRS groups. High levels of local proteins IL-5 and IL-13 and low levels of all isoform of TGF-β characterized CRSwNP without RA and BA. The combination of CRSwNP with AR showed high levels of proinflammatory cytokines IL-6 and IL-1β, and high levels of TGF-β1 and TGF-β2. The combination of CRSwNP with aBA estimated low levels of proinflammatory cytokines IL-1β, IFN-γ; in case of CRS+nBA we determined the highest levels of TGF-β1, TGF-β2 and TGF-β3 in nasal polyp tissue. CONCLUSIONS Each CRSwNP phenotype is characterized by different mechanism of local inflammation. This underlies the necessity to diagnose BA and respiratory allergy among these patients. The evaluation of local cytokine profile in different CRSwNP phenotypes can help to determine the target anticytokine therapy for patients who has low efficacy of basic corticosteroid therapy.
Collapse
Affiliation(s)
- E L Savlevich
- Central State Medical Academy of Administrative Directorate of the President of the Russian Federation, Moscow, Russia
| | - A V Zurochka
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
- South Ural State University (National Research University), Chelyabinsk, Russia
| | | | - V I Egorov
- Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | | | - E S Mitrofanova
- Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies, Moscow, Russia
| | | |
Collapse
|
11
|
Alsaab HO, Alharbi FD, Alhibs AS, Alanazi NB, Alshehri BY, Saleh MA, Alshehri FS, Algarni MA, Almugaiteeb T, Uddin MN, Alzhrani RM. PLGA-Based Nanomedicine: History of Advancement and Development in Clinical Applications of Multiple Diseases. Pharmaceutics 2022; 14:pharmaceutics14122728. [PMID: 36559223 PMCID: PMC9786338 DOI: 10.3390/pharmaceutics14122728] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Research on the use of biodegradable polymers for drug delivery has been ongoing since they were first used as bioresorbable surgical devices in the 1980s. For tissue engineering and drug delivery, biodegradable polymer poly-lactic-co-glycolic acid (PLGA) has shown enormous promise among all biomaterials. PLGA are a family of FDA-approved biodegradable polymers that are physically strong and highly biocompatible and have been extensively studied as delivery vehicles of drugs, proteins, and macromolecules such as DNA and RNA. PLGA has a wide range of erosion times and mechanical properties that can be modified. Many innovative platforms have been widely studied and created for the development of methods for the controlled delivery of PLGA. In this paper, the various manufacturing processes and characteristics that impact their breakdown and drug release are explored in depth. Besides different PLGA-based nanoparticles, preclinical and clinical applications for different diseases and the PLGA platform types and their scale-up issues will be discussed.
Collapse
Affiliation(s)
- Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
- Correspondence: ; Tel.: +966-556047523
| | - Fatima D. Alharbi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alanoud S. Alhibs
- Department of Pharmacy, King Fahad Medical City, Riyadh 11564, Saudi Arabia
| | - Nouf B. Alanazi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Bayan Y. Alshehri
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Marwa A. Saleh
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11754, Egypt
| | - Fahad S. Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| | - Majed A. Algarni
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Turki Almugaiteeb
- Taqnia-Research Products Development Company, Riyadh 13244, Saudi Arabia
| | | | - Rami M. Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| |
Collapse
|
12
|
Maggi E, Parronchi P, Azzarone BG, Moretta L. A pathogenic integrated view explaining the different endotypes of asthma and allergic disorders. Allergy 2022; 77:3267-3292. [PMID: 35842745 DOI: 10.1111/all.15445] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
The inflammation of allergic diseases is characterized by a complex interaction between type 2 and type 3 immune responses, explaining clinical symptoms and histopathological patterns. Airborne stimuli activate the mucosal epithelium to release a number of molecules impacting the activity of resident immune and environmental cells. Signals from the mucosal barrier, regulatory cells, and the inflamed tissue are crucial conditions able to modify innate and adaptive effector cells providing the selective homing of eosinophils or neutrophils. The high plasticity of resident T- and innate lymphoid cells responding to external signals is the prerequisite to explain the multiplicity of endotypes of allergic diseases. This notion paved the way for the huge use of specific biologic drugs interfering with pathogenic mechanisms of inflammation. Based on the response of the epithelial barrier, the activity of resident regulatory cells, and functions of structural non-lymphoid environmental cells, this review proposes some immunopathogenic scenarios characterizing the principal endotypes which can be associated with a precise phenotype of asthma. Recent literature indicates that similar concepts can also be applied to the inflammation of other non-respiratory allergic disorders. The next challenges will consist in defining specific biomarker(s) of each endotype allowing for a quick diagnosis and the most effective personalized therapy.
Collapse
Affiliation(s)
- Enrico Maggi
- Department of Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paola Parronchi
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | | | - Lorenzo Moretta
- Department of Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
13
|
Saxena J, Bisen M, Misra A, Srivastava VK, Kaushik S, Siddiqui AJ, Mishra N, Singh A, Jyoti A. Targeting COPD with PLGA-Based Nanoparticles: Current Status and Prospects. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5058121. [PMID: 35309178 PMCID: PMC8933108 DOI: 10.1155/2022/5058121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/01/2022] [Accepted: 02/11/2022] [Indexed: 11/17/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is pulmonary emphysema characterized by blockage in the airflow resulting in the long-term breathing problem, hence a major cause of mortality worldwide. Excessive generation of free radicals and the development of chronic inflammation are the major two episodes underlying the pathogenesis of COPD. Currently used drugs targeting these episodes including anti-inflammatory, antioxidants, and corticosteroids are unsafe, require high doses, and pose serious side effects. Nanomaterial-conjugated drugs have shown promising therapeutic potential against different respiratory diseases as they are required in small quantities which lower overall treatment costs and can be effectively targeted to diseased tissue microenvironment hence having minimal side effects. Poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) are safe as their breakdown products are easily metabolized in the body. Drugs loaded on the PLGA NPs have been shown to be promising agents as anticancer, antimicrobial, antioxidants, and anti-inflammatory. Surface modification of PLGA NPs can further improve their mechanical properties, drug loading potential, and pharmacological activities. In the present review, we have presented a brief insight into the pathophysiological mechanism underlying COPD and highlighted the role, potential, and current status of PLGA NPs loaded with drugs in the therapy of COPD.
Collapse
Affiliation(s)
- Juhi Saxena
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab 140413, India
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh 173229, India
| | - Monish Bisen
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh 173229, India
| | - Aditya Misra
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh 173229, India
| | - Vijay Kumar Srivastava
- Amity Institute of Biotechnology, Amity University Rajasthan, Amity Education Valley, Kant Kalwar, NH-11C, Jaipur-Delhi Highway, Jaipur, India
| | - Sanket Kaushik
- Amity Institute of Biotechnology, Amity University Rajasthan, Amity Education Valley, Kant Kalwar, NH-11C, Jaipur-Delhi Highway, Jaipur, India
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Neetu Mishra
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, Maharashtra 412115, India
| | - Abhijeet Singh
- Department of Biosciences, Manipal University Jaipur, Rajasthan 303007, India
| | - Anupam Jyoti
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab 140413, India
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh 173229, India
| |
Collapse
|
14
|
Gunaseelan S, Ariffin MZ, Khanna S, Ooi MH, Perera D, Chu JJH, Chua JJE. Pharmacological perturbation of CXCL1 signaling alleviates neuropathogenesis in a model of HEVA71 infection. Nat Commun 2022; 13:890. [PMID: 35173169 PMCID: PMC8850555 DOI: 10.1038/s41467-022-28533-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 01/27/2022] [Indexed: 12/14/2022] Open
Abstract
Hand, foot and mouth disease (HFMD) caused by Human Enterovirus A71 (HEVA71) infection is typically a benign infection. However, in minority of cases, children can develop severe neuropathology that culminate in fatality. Approximately 36.9% of HEVA71-related hospitalizations develop neurological complications, of which 10.5% are fatal. Yet, the mechanism by which HEVA71 induces these neurological deficits remain unclear. Here, we show that HEVA71-infected astrocytes release CXCL1 which supports viral replication in neurons by activating the CXCR2 receptor-associated ERK1/2 signaling pathway. Elevated CXCL1 levels correlates with disease severity in a HEVA71-infected mice model. In humans infected with HEVA71, high CXCL1 levels are only present in patients presenting neurological complications. CXCL1 release is specifically triggered by VP4 synthesis in HEVA71-infected astrocytes, which then acts via its receptor CXCR2 to enhance viral replication in neurons. Perturbing CXCL1 signaling or VP4 myristylation strongly attenuates viral replication. Treatment with AZD5069, a CXCL1-specific competitor, improves survival and lessens disease severity in infected animals. Collectively, these results highlight the CXCL1-CXCR2 signaling pathway as a potential target against HFMD neuropathogenesis.
Collapse
Affiliation(s)
- Saravanan Gunaseelan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, National University of Singapore, Singapore, 117597, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore, 117456, Singapore
| | - Mohammed Zacky Ariffin
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sanjay Khanna
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore, 117456, Singapore
| | - Mong How Ooi
- Department of Paediatrics, Sarawak General Hospital, Kuching, Sarawak, Malaysia
- Institute of Health and Community Medicine, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - David Perera
- Institute of Health and Community Medicine, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, National University of Singapore, Singapore, 117597, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
- Infectious Disease Translational Research Programme, National University of Singapore, Singapore, 117597, Singapore.
| | - John Jia En Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- LSI Neurobiology Programme, National University of Singapore, Singapore, 117456, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
15
|
Uwagboe I, Adcock IM, Lo Bello F, Caramori G, Mumby S. New drugs under development for COPD. Minerva Med 2022; 113:471-496. [PMID: 35142480 DOI: 10.23736/s0026-4806.22.08024-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The characteristic features of chronic obstructive pulmonary disease (COPD) include inflammation and remodelling of the lower airways and lung parenchyma together with activation of inflammatory and immune processes. Due to the increasing habit of cigarette smoking worldwide COPD prevalence is increasing globally. Current therapies are unable to prevent COPD progression in many patients or target many of its hallmark characteristics which may reflect the lack of adequate biomarkers to detect the heterogeneous clinical and molecular nature of COPD. In this chapter we review recent molecular data that may indicate novel pathways that underpin COPD subphenotypes and indicate potential improvements in the classes of drugs currently used to treat COPD. We also highlight the evidence for new drugs or approaches to treat COPD identified using molecular and other approaches including kinase inhibitors, cytokine- and chemokine-directed biologicals and small molecules, antioxidants and redox signalling pathway inhibitors, inhaled anti-infectious agents and senolytics. It is important to consider the phenotypes/molecular endotypes of COPD patients together with specific outcome measures to target new therapies to particular COPD subtypes. This will require greater understanding of COPD molecular pathologies and a focus on biomarkers of predicting disease subsets and responder/non-responder populations.
Collapse
Affiliation(s)
- Isabel Uwagboe
- Airways Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| | - Ian M Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College, London, UK -
| | - Federica Lo Bello
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Sharon Mumby
- Airways Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
16
|
Yamaguchi H, Hsu JM, Yang WH, Hung MC. Mechanisms regulating PD-L1 expression in cancers and associated opportunities for novel small-molecule therapeutics. Nat Rev Clin Oncol 2022; 19:287-305. [DOI: 10.1038/s41571-022-00601-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
|
17
|
Concepts of advanced therapeutic delivery systems for the management of remodeling and inflammation in airway diseases. Future Med Chem 2022; 14:271-288. [PMID: 35019757 PMCID: PMC8890134 DOI: 10.4155/fmc-2021-0081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic respiratory disorders affect millions of people worldwide. Pathophysiological changes to the normal airway wall structure, including changes in the composition and organization of its cellular and molecular constituents, are referred to as airway remodeling. The inadequacy of effective treatment strategies and scarcity of novel therapies available for the treatment and management of chronic respiratory diseases have given rise to a serious impediment in the clinical management of such diseases. The progress made in advanced drug delivery, has offered additional advantages to fight against the emerging complications of airway remodeling. This review aims to address the gaps in current knowledge about airway remodeling, the relationships between remodeling, inflammation, clinical phenotypes and the significance of using novel drug delivery methods.
Collapse
|
18
|
Novel Immunomodulatory Therapies for Respiratory Pathologies. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC8238403 DOI: 10.1016/b978-0-12-820472-6.00073-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
19
|
Ogulur I, Pat Y, Ardicli O, Barletta E, Cevhertas L, Fernandez‐Santamaria R, Huang M, Bel Imam M, Koch J, Ma S, Maurer DJ, Mitamura Y, Peng Y, Radzikowska U, Rinaldi AO, Rodriguez‐Coira J, Satitsuksanoa P, Schneider SR, Wallimann A, Zhakparov D, Ziadlou R, Brüggen M, Veen W, Sokolowska M, Baerenfaller K, Zhang L, Akdis M, Akdis CA. Advances and highlights in biomarkers of allergic diseases. Allergy 2021; 76:3659-3686. [PMID: 34519063 PMCID: PMC9292545 DOI: 10.1111/all.15089] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 05/19/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
During the past years, there has been a global outbreak of allergic diseases, presenting a considerable medical and socioeconomical burden. A large fraction of allergic diseases is characterized by a type 2 immune response involving Th2 cells, type 2 innate lymphoid cells, eosinophils, mast cells, and M2 macrophages. Biomarkers are valuable parameters for precision medicine as they provide information on the disease endotypes, clusters, precision diagnoses, identification of therapeutic targets, and monitoring of treatment efficacies. The availability of powerful omics technologies, together with integrated data analysis and network‐based approaches can help the identification of clinically useful biomarkers. These biomarkers need to be accurately quantified using robust and reproducible methods, such as reliable and point‐of‐care systems. Ideally, samples should be collected using quick, cost‐efficient and noninvasive methods. In recent years, a plethora of research has been directed toward finding novel biomarkers of allergic diseases. Promising biomarkers of type 2 allergic diseases include sputum eosinophils, serum periostin and exhaled nitric oxide. Several other biomarkers, such as pro‐inflammatory mediators, miRNAs, eicosanoid molecules, epithelial barrier integrity, and microbiota changes are useful for diagnosis and monitoring of allergic diseases and can be quantified in serum, body fluids and exhaled air. Herein, we review recent studies on biomarkers for the diagnosis and treatment of asthma, chronic urticaria, atopic dermatitis, allergic rhinitis, chronic rhinosinusitis, food allergies, anaphylaxis, drug hypersensitivity and allergen immunotherapy. In addition, we discuss COVID‐19 and allergic diseases within the perspective of biomarkers and recommendations on the management of allergic and asthmatic patients during the COVID‐19 pandemic.
Collapse
|
20
|
Xu X, Reitsma S, Wang DY, Fokkens WJ. Highlights in the advances of chronic rhinosinusitis. Allergy 2021; 76:3349-3358. [PMID: 33948955 DOI: 10.1111/all.14892] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/15/2021] [Accepted: 04/26/2021] [Indexed: 12/27/2022]
Abstract
Chronic rhinosinusitis (CRS) is a complex upper airway inflammatory disease with a broad spectrum of clinical variants. As our understanding of the disease pathophysiology evolves, so too does our philosophy towards the approach and management of CRS. Endotyping is gaining favour over phenotype-based classifications, owing to its potential in prognosticating disease severity and delivering precision treatment. Endotyping is especially useful in challenging CRS with nasal polyposis cases, for whom novel treatment options such as biologicals are now available. The latest European Position Paper on Rhinosinusitis and Nasal Polyps (EPOS2020) reflects these changes with updated rhinosinusitis classifications and new integrated care pathways. With the coronavirus disease 2019 (COVID-19) pandemic, physicians and rhinologists have to balance the responsibility of managing their patients' upper airway while adequately protecting themselves from droplet and aerosol transmission. This review summarises the key updates from EPOS2020, endotype-based classification and biomarkers. The role of biologicals in CRS and the lessons we can draw from their use in severe asthma will be examined. Finally, the principles of CRS management during COVID-19 will also be discussed.
Collapse
Affiliation(s)
- Xinni Xu
- Department of Otolaryngology ‐ Head & Neck Surgery National University Hospital Singapore Singapore
| | - Sietze Reitsma
- Department of Otorhinolaryngology Amsterdam University Medical Centers, location AMC Amsterdam The Netherlands
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Wytske J. Fokkens
- Department of Otorhinolaryngology Amsterdam University Medical Centers, location AMC Amsterdam The Netherlands
| |
Collapse
|
21
|
Geng B, Craig TJ. Small molecule drugs for atopic dermatitis, rheumatoid arthritis, and hereditary angioedema. Ann Allergy Asthma Immunol 2021; 128:263-268. [PMID: 34673223 DOI: 10.1016/j.anai.2021.10.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To review recent trends in the development of targeted small molecule drugs (SMDs) for the treatment of immunologically driven disorders, including atopic dermatitis, rheumatoid arthritis, and hereditary angioedema. DATA SOURCES Data sources included peer-reviewed published literature from the PubMed database, published abstracts from scientific and medical meetings, and medication information from the Drugs@FDA database. STUDY SELECTIONS Articles with primary or retrospective trial results, articles with patient or physician survey results, articles providing expert perspectives, and commentary on chronic immunologic disorders, Food and Drug Administration package inserts, and abstracts from scientific meetings were selected. RESULTS Targeted biological therapies have greatly improved response rates and symptom relief for patients with long-term immunologically driven disorders over the past 2 decades. However, recent advances in the understanding of molecular pathways involved in the pathogenesis of these disorders have led to the development of novel targeted SMDs, such as tofacitinib and berotralstat, that can be delivered orally or topically. Few head-to-head studies that compare the safety and efficacy of biologics to SMDs in immunologically driven disorders exist, although some studies suggest that oral and topical modes of administration are preferred by patients and may improve patient quality of life over time. CONCLUSION Scientific advances have led to an increase in the development of targeted SMDs for the treatment of chronic immunologic disorders, which may revolutionize the management of these diseases. Head-to-head studies and real-world evidence are needed to fully compare treatment attributes between biologics and SMDs, including safety, efficacy, adherence, impact on quality of life, and cost-effectiveness.
Collapse
Affiliation(s)
- Bob Geng
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of California San Diego, La Jolla, California
| | - Timothy J Craig
- Department of Medicine, Pediatrics, and Biomedical Sciences, Penn State University, Hershey Medical Center, Hershey, Pennsylvania.
| |
Collapse
|
22
|
Abed S, Turner R, Serniuck N, Tat V, Naiel S, Hayat A, Mekhael O, Vierhout M, Ask K, Rullo AF. Cell-specific drug targeting in the lung. Biochem Pharmacol 2021; 190:114577. [PMID: 33887259 DOI: 10.1016/j.bcp.2021.114577] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 11/26/2022]
Abstract
Non-targeted drug delivery systems have several limitations including the decreased bioavailability of the drug, poor stability and rapid clearance in addition to off-target distribution. Cell-specific targeted delivery approaches promise to overcome some of these limitations and enhance therapeutic selectivity. In this review, we aim to discuss cell-specific targeted approachesin the lung at the biochemical and molecular levels. These approaches include;a) directly administered small molecule drugs with intracellular action; b) targeted biologics and synthetic hybrids with extracellular action; c) site activateddrugs; and d) delivery systems.We discuss the pharmaceutical and biochemical parameters that govern the fate of drug molecules at delivery sites while presenting an overview of relevant literature surrounding this area of research and current advancements.
Collapse
Affiliation(s)
- Soumeya Abed
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Rebecca Turner
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Nickolas Serniuck
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Victor Tat
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Safaa Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Aaron Hayat
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Olivia Mekhael
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Megan Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
| | - Anthony F Rullo
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
23
|
Schneider AL, Schleimer RP, Tan BK. Targetable pathogenic mechanisms in nasal polyposis. Int Forum Allergy Rhinol 2021; 11:1220-1234. [PMID: 33660425 DOI: 10.1002/alr.22787] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) represents a challenging disease entity with significant rates of recurrence following appropriate medical and surgical therapy. Recent approval of targeted biologics in CRSwNP compels deeper understanding of underlying disease pathophysiology. Both of the approved biologics for CRSwNP modulate the type 2 inflammatory pathway, and the majority of drugs in the clinical trials pathway are similarly targeted. However, there remain multiple other pathogenic mechanisms relevant to CRSwNP for which targeted therapeutics already exist in other inflammatory diseases that have not been studied directly. In this article we summarize pathogenic mechanisms of interest in CRSwNP and discuss the results of ongoing clinical studies of targeted therapeutics in CRSwNP and other related human inflammatory diseases.
Collapse
Affiliation(s)
| | - Robert P Schleimer
- Department of Otolaryngology, Head and Neck Surgery, Chicago, Illinois, USA.,Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bruce K Tan
- Department of Otolaryngology, Head and Neck Surgery, Chicago, Illinois, USA.,Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
24
|
Bhutani P, Joshi G, Raja N, Bachhav N, Rajanna PK, Bhutani H, Paul AT, Kumar R. U.S. FDA Approved Drugs from 2015-June 2020: A Perspective. J Med Chem 2021; 64:2339-2381. [PMID: 33617716 DOI: 10.1021/acs.jmedchem.0c01786] [Citation(s) in RCA: 340] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the present work, we report compilation and analysis of 245 drugs, including small and macromolecules approved by the U.S. FDA from 2015 until June 2020. Nearly 29% of the drugs were approved for the treatment of various types of cancers. Other major therapeutic areas of focus were infectious diseases (14%); neurological conditions (12%); and genetic, metabolic, and cardiovascular disorders (7-8% each). Itemization of the approved drugs according to the year of approval, sponsor, target, chemical class, major drug-metabolizing enzyme(s), route of administration/elimination, and drug-drug interaction liability (perpetrator or/and victim) is presented and discussed. An effort has been made to analyze the pharmacophores to identify the structural (e.g., aromatic, heterocycle, and aliphatic), elemental (e.g., boron, sulfur, fluorine, phosphorus, and deuterium), and functional group (e.g., nitro drugs) diversity among the approved drugs. Further, descriptor-based chemical space analysis of FDA approved drugs and several strategies utilized for optimizing metabolism leading to their discoveries have been emphasized. Finally, an analysis of drug-likeness for the approved drugs is presented.
Collapse
Affiliation(s)
- Priyadeep Bhutani
- Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre, Syngene International Limited, Bangalore 560099, India.,Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Rajasthan 333031, India
| | - Gaurav Joshi
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda 151001, India
| | - Nivethitha Raja
- Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre, Syngene International Limited, Bangalore 560099, India
| | - Namrata Bachhav
- 1015 E Cozza Drive # 12, Spokane Washington 99208, United States
| | - Prabhakar K Rajanna
- Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre, Syngene International Limited, Bangalore 560099, India
| | - Hemant Bhutani
- Pharmaceutical Development, Biocon Bristol-Myers Squibb R&D Centre, Bristol-Myers Squibb India Private Limited, Bangalore 560099, India
| | - Atish T Paul
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Rajasthan 333031, India
| | - Raj Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda 151001, India
| |
Collapse
|
25
|
Agache I, Akdis CA, Akdis M, Canonica GW, Casale T, Chivato T, Corren J, Chu DK, Del Giacco S, Eiwegger T, Flood B, Firinu D, Gern JE, Hamelmann E, Hanania N, Hernández-Martín I, Knibb R, Mäkelä M, Nair P, O'Mahony L, Papadopoulos NG, Papi A, Park HS, Pérez de Llano L, Pfaar O, Quirce S, Sastre J, Shamji M, Schwarze J, Palomares O, Jutel M. EAACI Biologicals Guidelines-Recommendations for severe asthma. Allergy 2021; 76:14-44. [PMID: 32484954 DOI: 10.1111/all.14425] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Severe asthma imposes a significant burden on patients, families and healthcare systems. Management is difficult, due to disease heterogeneity, co-morbidities, complexity in care pathways and differences between national or regional healthcare systems. Better understanding of the mechanisms has enabled a stratified approach to the management of severe asthma, supporting the use of targeted treatments with biologicals. However, there are still many issues that require further clarification. These include selection of a certain biological (as they all target overlapping disease phenotypes), the definition of response, strategies to enhance the responder rate, the duration of treatment and its regimen (in the clinic or home-based) and its cost-effectiveness. The EAACI Guidelines on the use of biologicals in severe asthma follow the GRADE approach in formulating recommendations for each biological and each outcome. In addition, a management algorithm for the use of biologicals in the clinic is proposed, together with future approaches and research priorities.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine-Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy, Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
| | - Thomas Casale
- Division of Allergy and Immunology, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Tomas Chivato
- School of Medicine, University CEU San Pablo, Madrid, Spain
| | - Jonathan Corren
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Derek K Chu
- Department of Health Research Methods, Evidence and Impact, Division of Immunology and Allergy, and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Division of Immunology and Allergy, Food Allergy and Anaphylaxis Program, The Hospital for Sick Children, Departments of Paediatrics and Immunology, University of Toronto, Toronto, ON, Canada
| | - Breda Flood
- European Federation of Allergy and Airway Diseases, Brussels, Belgium
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - James E Gern
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Eckard Hamelmann
- Children's Center Bethel, Evangelical Hospital Bethel, University of Bielefeld, Bielefeld, Germany
| | - Nicola Hanania
- Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Rebeca Knibb
- Department of Psychology, School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Mika Mäkelä
- Skin and Allergy Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Parameswaran Nair
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Liam O'Mahony
- Departments of Medicine and Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nikolaos G Papadopoulos
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
- Allergy Department, 2nd Pediatric Clinic, National Kapodistrian University of Athens, Athens, Greece
| | - Alberto Papi
- Research Center on Asthma and COPD, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University, Ajou, Korea
| | | | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Santiago Quirce
- Department of Allergy, La Paz University Hospital, IdiPAZ, CIBER of Respiratory Diseases (CIBERES), Universidad Autónoma de Madrid, Madrid, Spain
| | - Joaquin Sastre
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mohamed Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair, Development, National Heart and Lung Institute, London, UK
- Imperial College NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Jurgen Schwarze
- Centre for Inflammation Research, Child Life and Health, The University of Edinburgh, Edinburgh, UK
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, Chemistry School, Complutense University of Madrid, Madrid, Spain
| | - Marek Jutel
- Department of Clinical Immunology, University of Wroclaw, Wroclaw, Poland
- ALL-MED" Medical Research Institute, Wroclaw, Poland
| |
Collapse
|
26
|
Roth-Walter F, Adcock IM, Benito-Villalvilla C, Bianchini R, Bjermer L, Boyman O, Caramori G, Cari L, Fan Chung K, Diamant Z, Eguiluz-Gracia I, Knol EF, Kolios A, Levi-Schaffer F, Nocentini G, Palomares O, Redegeld F, Van Esch B, Stellato C. Immune modulation via T regulatory cell enhancement: Disease-modifying therapies for autoimmunity and their potential for chronic allergic and inflammatory diseases-An EAACI position paper of the Task Force on Immunopharmacology (TIPCO). Allergy 2021; 76:90-113. [PMID: 32593226 DOI: 10.1111/all.14478] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022]
Abstract
Therapeutic advances using targeted biologicals and small-molecule drugs have achieved significant success in the treatment of chronic allergic, autoimmune, and inflammatory diseases particularly for some patients with severe, treatment-resistant forms. This has been aided by improved identification of disease phenotypes. Despite these achievements, not all severe forms of chronic inflammatory and autoimmune diseases are successfully targeted, and current treatment options, besides allergen immunotherapy for selected allergic diseases, fail to change the disease course. T cell-based therapies aim to cure diseases through the selective induction of appropriate immune responses following the delivery of engineered, specific cytotoxic, or regulatory T cells (Tregs). Adoptive cell therapies (ACT) with genetically engineered T cells have revolutionized the oncology field, bringing curative treatment for leukemia and lymphoma, while therapies exploiting the suppressive functions of Tregs have been developed in nononcological settings, such as in transplantation and autoimmune diseases. ACT with Tregs are also being considered in nononcological settings such as cardiovascular disease, obesity, and chronic inflammatory disorders. After describing the general features of T cell-based approaches and current applications in autoimmune diseases, this position paper reviews the experimental models testing or supporting T cell-based approaches, especially Treg-based approaches, in severe IgE-mediated responses and chronic respiratory airway diseases, such as severe asthma and COPD. Along with an assessment of challenges and unmet needs facing the application of ACT in these settings, this article underscores the potential of ACT to offer curative options for patients with severe or treatment-resistant forms of these immune-driven disorders.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Ian M Adcock
- Molecular Cell Biology Group, National Heart & Lung Institute, Imperial College London, London, UK
| | - Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Rodolfo Bianchini
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Lung and Allergy research, Allergy, Asthma and COPD Competence Center, Lund University, Lund, Sweden
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gaetano Caramori
- Department of Biomedical Sciences, Dentistry and Morphological and Functional Imaging (BIOMORF), Respiratory Medicine Unit, University of Messina, Messina, Italy
| | - Luigi Cari
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Kian Fan Chung
- Experimental Studies Medicine at National Heart & Lung Institute, Imperial College London & Royal Brompton & Harefield NHS Trust, London, UK
| | - Zuzana Diamant
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Skane University Hospital, Lund, Sweden
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department of Clinical Pharmacy & Pharmacology, University Groningen, University Medical Center Groningen and QPS-NL, Groningen, Netherlands
| | - Ibon Eguiluz-Gracia
- Allergy Unit, Hospital Regional Universitario de Málaga-Instituto de Investigación Biomédica de Málaga (IBIMA)-ARADyAL, Málaga, Spain
| | - Edward F Knol
- Departments of Immunology and Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Antonios Kolios
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Francesca Levi-Schaffer
- Pharmacology Unit, Faculty of Medicine, Institute for Drug Research, The Hebrew University of Jerusalem, Israel
| | - Giuseppe Nocentini
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Frank Redegeld
- Faculty of Science, Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Betty Van Esch
- Faculty of Science, Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| |
Collapse
|
27
|
Lyly A, Laulajainen-Hongisto A, Gevaert P, Kauppi P, Toppila-Salmi S. Monoclonal Antibodies and Airway Diseases. Int J Mol Sci 2020; 21:E9477. [PMID: 33322143 PMCID: PMC7763928 DOI: 10.3390/ijms21249477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibodies, biologics, are a relatively new treatment option for severe chronic airway diseases, asthma, allergic rhinitis, and chronic rhinosinusitis (CRS). In this review, we focus on the physiological and pathomechanisms of monoclonal antibodies, and we present recent study results regarding their use as a therapeutic option against severe airway diseases. Airway mucosa acts as a relative barrier, modulating antigenic stimulation and responding to environmental pathogen exposure with a specific, self-limited response. In severe asthma and/or CRS, genome-environmental interactions lead to dysbiosis, aggravated inflammation, and disease. In healthy conditions, single or combined type 1, 2, and 3 immunological response pathways are invoked, generating cytokine, chemokine, innate cellular and T helper (Th) responses to eliminate viruses, helminths, and extracellular bacteria/fungi, correspondingly. Although the pathomechanisms are not fully known, the majority of severe airway diseases are related to type 2 high inflammation. Type 2 cytokines interleukins (IL) 4, 5, and 13, are orchestrated by innate lymphoid cell (ILC) and Th subsets leading to eosinophilia, immunoglobulin E (IgE) responses, and permanently impaired airway damage. Monoclonal antibodies can bind or block key parts of these inflammatory pathways, resulting in less inflammation and improved disease control.
Collapse
Affiliation(s)
- Annina Lyly
- Inflammation Centre, Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, P.O. Box 160, 00029 HUS Helsinki, Finland;
- Department of Otorhinolaryngology—Head and Neck Surgery, Helsinki University Hospital, University of Helsinki, 00029 HUS Helsinki, Finland;
| | - Anu Laulajainen-Hongisto
- Department of Otorhinolaryngology—Head and Neck Surgery, Helsinki University Hospital, University of Helsinki, 00029 HUS Helsinki, Finland;
| | - Philippe Gevaert
- Department of Otorhinolaryngology, Upper Airway Research Laboratory, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Paula Kauppi
- Heart and Lung Center, Pulmonary Department, University of Helsinki and Helsinki University Hospital, 00029 HUS Helsinki, Finland;
| | - Sanna Toppila-Salmi
- Inflammation Centre, Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, P.O. Box 160, 00029 HUS Helsinki, Finland;
- Medicum, Haartman Institute, University of Helsinki, 00029 HUS Helsinki, Finland
| |
Collapse
|
28
|
Bousquet J, Grattan CE, Akdis CA, Eigenmann PA, Hoffmann-Sommergruber K, Agache I, Jutel M. Highlights and recent developments in allergic diseases in EAACI journals (2019). Clin Transl Allergy 2020; 10:56. [PMID: 33292572 PMCID: PMC7712618 DOI: 10.1186/s13601-020-00366-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
The European Academy of Allergy and Clinical Immunology (EAACI) owns three journals: Allergy, Pediatric Allergy and Immunology and Clinical and Translational Allergy. One of the major goals of EAACI is to support health promotion in which prevention of allergy and asthma plays a critical role and to disseminate the knowledge of allergy to all stakeholders including the EAACI junior members. There was substantial progress in 2019 in the identification of basic mechanisms of allergic and respiratory disease and the translation of these mechanisms into clinics. Better understanding of molecular and cellular mechanisms, efforts for the development of biomarkers for disease prediction, novel prevention and intervention studies, elucidation of mechanisms of multimorbidities, entrance of new drugs in the clinics as well as recently completed phase three clinical studies and publication of a large number of allergen immunotherapy studies and meta-analyses have been the highlights of the last year.
Collapse
Affiliation(s)
- J Bousquet
- MACVIA-France, Montpellier, France. .,CHRU Arnaud de Villeneuve, 371 Avenue du Doyen Gaston Giraud, 34295, Montpellier Cedex 5, France.
| | - C E Grattan
- St John's Institute of Dermatology, Guy's Hospital, London, UK
| | - C A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - P A Eigenmann
- Pediatric Allergy Unit, University Hospitals of Geneva, Geneva, Switzerland
| | - K Hoffmann-Sommergruber
- Depart of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - I Agache
- Transylvania University Brasov, Brasov, Romania
| | - M Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wrocław, Poland.,ALL-MED Medical Research Institute, Wrocław, Poland
| |
Collapse
|
29
|
Breiteneder H, Peng Y, Agache I, Diamant Z, Eiwegger T, Fokkens WJ, Traidl‐Hoffmann C, Nadeau K, O'Hehir RE, O'Mahony L, Pfaar O, Torres MJ, Wang D, Zhang L, Akdis CA. Biomarkers for diagnosis and prediction of therapy responses in allergic diseases and asthma. Allergy 2020; 75:3039-3068. [PMID: 32893900 PMCID: PMC7756301 DOI: 10.1111/all.14582] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
Modern health care requires a proactive and individualized response to diseases, combining precision diagnosis and personalized treatment. Accordingly, the approach to patients with allergic diseases encompasses novel developments in the area of personalized medicine, disease phenotyping and endotyping, and the development and application of reliable biomarkers. A detailed clinical history and physical examination followed by the detection of IgE immunoreactivity against specific allergens still represents the state of the art. However, nowadays, further emphasis focuses on the optimization of diagnostic and therapeutic standards and a large number of studies have been investigating the biomarkers of allergic diseases, including asthma, atopic dermatitis, allergic rhinitis, food allergy, urticaria and anaphylaxis. Various biomarkers have been developed by omics technologies, some of which lead to a better classification of distinct phenotypes or endotypes. The introduction of biologicals to clinical practice increases the need for biomarkers for patient selection, prediction of outcomes and monitoring, to allow for an adequate choice of the duration of these costly and long‐lasting therapies. Escalating healthcare costs together with questions about the efficacy of the current management of allergic diseases require further development of a biomarker‐driven approach. Here, we review biomarkers in diagnosis and treatment of asthma, atopic dermatitis, allergic rhinitis, viral infections, chronic rhinosinusitis, food allergy, drug hypersensitivity and allergen immunotherapy with a special emphasis on specific IgE, the microbiome and the epithelial barrier. In addition, EAACI guidelines on biologicals are discussed within the perspective of biomarkers.
Collapse
Affiliation(s)
- Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Ya‐Qi Peng
- Swiss Institute of Allergy and Asthma Research (SIAF) University Zurich Davos Switzerland
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
- Otorhinolaryngology Hospital The First Affiliated Hospital Sun Yat‐Sen University Guangzhou China
| | - Ioana Agache
- Department of Allergy and Clinical Immunology Faculty of Medicine Transylvania University of Brasov Brasov Romania
| | - Zuzana Diamant
- Department of Respiratory Medicine & Allergology Institute for Clinical Science Skane University Hospital Lund University Lund Sweden
- Department of Respiratory Medicine First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
- Department of Clinical Pharmacy & Pharmacology University of GroningenUniversity Medical Center Groningen Groningen Netherlands
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute Hospital for Sick Children Toronto ON Canada
- Department of Immunology University of Toronto Toronto ON Canada
- Division of Immunology and Allergy Food Allergy and Anaphylaxis Program The Hospital for Sick Children Departments of Paediatrics and Immunology University of Toronto Toronto ON Canada
| | - Wytske J. Fokkens
- Department of Otorhinolaryngology Amsterdam University Medical Centres Amsterdam The Netherlands
| | - Claudia Traidl‐Hoffmann
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- ZIEL ‐ Institute for Food & Health Technical University of Munich Freising‐Weihenstephan Germany
| | - Kari Nadeau
- Sean N. Parker Center for Allergy & Asthma Research Stanford University Stanford CA USA
| | - Robyn E. O'Hehir
- Department of Allergy, immunology and Respiratory Medicine Central Clinical School Monash University Melbourne Vic. Australia
- Allergy, Asthma and Clinical Immunology Service Alfred Health Melbourne Vic. Australia
| | - Liam O'Mahony
- Departments of Medicine and Microbiology APC Microbiome Ireland National University of Ireland Cork Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital MarburgPhilipps‐Universität Marburg Marburg Germany
| | - Maria J. Torres
- Allergy Unit Regional University Hospital of Malaga‐IBIMA‐UMA‐ARADyAL Malaga Spain
| | - De‐Yun Wang
- Department of Otolaryngology Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery and Department of Allergy Beijing TongRen Hospital Beijing China
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University Zurich Davos Switzerland
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
| |
Collapse
|
30
|
Üzülmez Ö, Kalic T, Breiteneder H. Advances and novel developments in molecular allergology. Allergy 2020; 75:3027-3038. [PMID: 32882057 PMCID: PMC7756543 DOI: 10.1111/all.14579] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 12/21/2022]
Abstract
The continuous search for new allergens and the design of allergen derivatives improves the understanding of their allergenicity and aids the design of novel diagnostic and immunotherapy approaches. This article discusses the recent developments in allergen and epitope discovery, allergy diagnostics and immunotherapy. Structural information is crucial for the elucidation of cross-reactivity of marker allergens such as the walnut Jug r 6 or that of nonhomologous allergens, as shown for the peanut allergens Ara h 1 and 2. High-throughput sequencing, liposomal nanoallergen display, bead-based assays, and protein chimeras have been used in epitope discovery. The binding of natural ligands by the birch pollen allergen Bet v 1 or the mold allergen Alt a 1 increased the stability of these allergens, which is directly linked to their allergenicity. We also report recent findings on the use of component-resolved approaches, basophil activation test, and novel technologies for improvement of diagnostics. New strategies in allergen-specific immunotherapy have also emerged, such as the use of virus-like particles, biologics or novel adjuvants. The identification of dectin-1 as a key player in allergy to tropomyosins and the formyl peptide receptor 3 in allergy to lipocalins are outstanding examples of research into the mechanism of allergic sensitization.
Collapse
Affiliation(s)
- Öykü Üzülmez
- Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Tanja Kalic
- Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| |
Collapse
|
31
|
AlKhater SA. Dynamic Interplay Between Microbiota and Mucosal Immunity in Early Shaping of Asthma and its Implication for the COVID-19 Pandemic. J Asthma Allergy 2020; 13:369-383. [PMID: 33061464 PMCID: PMC7532070 DOI: 10.2147/jaa.s272705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/06/2020] [Indexed: 12/11/2022] Open
Abstract
The crosstalk between host immunity and the external environment in the mucous membranes of the gastrointestinal and respiratory tracts in bronchial asthma has recently been scrutinized. There is compelling evidence that the microbiota at these sites may play an important role in the pathogenesis of this chronic airway disease. The appearance of bacteria early in life in the gut before dissemination to the airways plays a pivotal role in shaping mucosal immunity. Loss of microbial diversity or dysbiosis can result in aberrant immune-mediated inflammation and mucosal barrier disruption, which coincides clinically with the successive development of the "allergic march" in asthma. Microbial manipulation may be effective in curbing asthma development by indirectly preserving homeostatic epithelial barrier functions. The protective effects and mechanisms of immunity-microbiome crosstalk at mucosal sites require further investigation to identify therapeutic and preventive measures in asthma. This topical review aims to highlight new evidence that compromised epithelial barrier function, which results in deregulated crosstalk between the microbiome and host mucosal immune system, is an important disease mechanism in asthma. In the light of current COVID-19 pandemic, the collective findings on the impact of mucosal microbiota on the suceptibility to SARS-CoV-2 infection and severity of COVID-19 is explored. The possible therapeutic implications to target these abnormalities are further discussed.
Collapse
Affiliation(s)
- Suzan A AlKhater
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Department of Pediatrics, King Fahad Hospital of the University, Al-Khobar, Saudi Arabia
| |
Collapse
|
32
|
Xu X, Ong YK, Wang DY. Novel findings in immunopathophysiology of chronic rhinosinusitis and their role in a model of precision medicine. Allergy 2020; 75:769-780. [PMID: 31512248 DOI: 10.1111/all.14044] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/26/2019] [Accepted: 09/01/2019] [Indexed: 02/06/2023]
Abstract
Our understanding of the pathophysiology of chronic rhinosinusitis (CRS) is continuously evolving. The traditional description of CRS in terms of two phenotypes based on the presence or absence of nasal polyps belies the underlying intricate immunopathophysiological processes responsible for this condition. CRS is being increasingly recognized as a disease spectrum encompassing a range of inflammatory states in the sinonasal cavity, with non-type 2 inflammatory disease on one end, type 2 inflammatory, eosinophil-heavy disease on the other and an overlap of both in different proportions in between. Abundance in research on the immune mechanisms of CRS has revealed various new endotypes that hold promise as biomarkers for the development of targeted therapies in severe, uncontrolled CRS. The introduction of precision medicine to manage this chronic, complex condition is a step forward in providing individualized care for all patients with CRS. In this review, the latest research on the pathophysiology of CRS with a focus on potential novel biomarkers and treatment options over the last 2 years are summarized and integrated into a suggested model of precision medicine in CRS.
Collapse
Affiliation(s)
- Xinni Xu
- Department of Otolaryngology‐Head and Neck Surgery National University Hospital System (NUHS) Singapore Singapore
| | - Yew Kwang Ong
- Department of Otolaryngology‐Head and Neck Surgery National University Hospital System (NUHS) Singapore Singapore
| | - De Yun Wang
- Department of Otolaryngology Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| |
Collapse
|
33
|
Passali D, Bellussi LM, Damiani V, Tosca MA, Motta G, Ciprandi G. Chronic rhinosinusitis with nasal polyposis: the role of personalized and integrated medicine. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:11-18. [PMID: 32073556 PMCID: PMC7947745 DOI: 10.23750/abm.v91i1-s.9243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/01/2020] [Indexed: 02/06/2023]
Abstract
Chronic rhinosinusitis with nasal polyposis (CRSwNP) is a frequent disorder. From a clinical and an immunopathological point of view, different phenotypes and endotypes have been identified. The frequent comorbidity with asthma allowed to pave the way to the use of biological agents for the treatment of CRSwNP. Biological agents are targeted to antagonize IgE, interleukin (IL) 4, IL-5, and IL-13 at present. However, a correct and appropriate workup is mandatory, mainly concerning the exact definition of the specific pheno-endotype. The preliminary outcomes are promising, even though there is a need for well-established indications, criteria of responsiveness, duration, and safety. On the other hand, this personalized medicine could be fruitfully integrated with gold-standard medications, such as intranasal corticosteroids. As CRSwNP is a chronic disorder, treatment should be long-lasting, so complementary anti-inflammatory treatments could be opportunely integrated and/or alternated to steroids.
Collapse
Affiliation(s)
| | | | | | | | - Gaetano Motta
- ENT Department, University Luigi Vanvitelli, Naples, Italy.
| | | |
Collapse
|
34
|
Hufnagl K, Pali-Schöll I, Roth-Walter F, Jensen-Jarolim E. Dysbiosis of the gut and lung microbiome has a role in asthma. Semin Immunopathol 2020; 42:75-93. [PMID: 32072252 PMCID: PMC7066092 DOI: 10.1007/s00281-019-00775-y] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023]
Abstract
Worldwide 300 million children and adults are affected by asthma. The development of asthma is influenced by environmental and other exogenous factors synergizing with genetic predisposition, and shaping the lung microbiome especially during birth and in very early life. The healthy lung microbial composition is characterized by a prevalence of bacteria belonging to the phyla Bacteroidetes, Actinobacteria, and Firmicutes. However, viral respiratory infections are associated with an abundance of Proteobacteria with genera Haemophilus and Moraxella in young children and adult asthmatics. This dysbiosis supports the activation of inflammatory pathways and contributes to bronchoconstriction and bronchial hyperresponsiveness. Exogenous factors can affect the natural lung microbiota composition positively (farming environment) or negatively (allergens, air pollutants). It is evident that also gut microbiota dysbiosis has a high influence on asthma pathogenesis. Antibiotics, antiulcer medications, and other drugs severely impair gut as well as lung microbiota. Resulting dysbiosis and reduced microbial diversity dysregulate the bidirectional crosstalk across the gut-lung axis, resulting in hypersensitivity and hyperreactivity to respiratory and food allergens. Efforts are undertaken to reconstitute the microbiota and immune balance by probiotics and engineered bacteria, but results from human studies do not yet support their efficacy in asthma prevention or treatment. Overall, dysbiosis of gut and lung seem to be critical causes of the increased emergence of asthma.
Collapse
Affiliation(s)
- Karin Hufnagl
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Isabella Pali-Schöll
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Franziska Roth-Walter
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria.
- Center for Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University Vienna, Währinger G. 18-20, 1090, Vienna, Austria.
| |
Collapse
|
35
|
Breiteneder H, Diamant Z, Eiwegger T, Fokkens WJ, Traidl‐Hoffmann C, Nadeau K, O’Hehir RE, O’Mahony L, Pfaar O, Torres MJ, Wang DY, Zhang L, Akdis CA. Future research trends in understanding the mechanisms underlying allergic diseases for improved patient care. Allergy 2019; 74:2293-2311. [PMID: 31056763 PMCID: PMC6973012 DOI: 10.1111/all.13851] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/28/2019] [Accepted: 04/12/2019] [Indexed: 12/16/2022]
Abstract
The specialties of allergy and clinical immunology have entered the era of precision medicine with the stratification of diseases into distinct disease subsets, specific diagnoses, and targeted treatment options, including biologicals and small molecules. This article reviews recent developments in research and patient care and future trends in the discipline. The section on basic mechanisms of allergic diseases summarizes the current status and defines research needs in structural biology, type 2 inflammation, immune tolerance, neuroimmune mechanisms, role of the microbiome and diet, environmental factors, and respiratory viral infections. In the section on diagnostic challenges, clinical trials, precision medicine and immune monitoring of allergic diseases, asthma, allergic and nonallergic rhinitis, and new approaches to the diagnosis and treatment of drug hypersensitivity reactions are discussed in further detail. In the third section, unmet needs and future research areas for the treatment of allergic diseases are highlighted with topics on food allergy, biologics, small molecules, and novel therapeutic concepts in allergen‐specific immunotherapy for airway disease. Unknowns and future research needs are discussed at the end of each subsection.
Collapse
Affiliation(s)
- Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Zuzana Diamant
- Department of Respiratory Medicine & Allergology, Institute for Clinical Science, Skane University Hospital Lund University Lund Sweden
- Department of Respiratory Medicine, First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
| | - Thomas Eiwegger
- Division of Immunology and Allergy Food Allergy and Anaphylaxis Program The Department of Pediatrics The Hospital for Sick Children Toronto Ontario Canada
- Research Institute, The Hospital for Sick Children, Translational Medicine Program Toronto Ontario Canada
- Department of Immunology The University of Toronto Toronto Ontario Canada
| | - Wytske J. Fokkens
- Department of Otorhinolaryngology Amsterdam University Medical Centres, Location AMC Amsterdam The Netherlands
| | - Claudia Traidl‐Hoffmann
- Chair and Institute of Environmental Medicine UNIKA‐T, Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- Christine Kühne Center for Allergy Research and Education Davos Switzerland
| | - Kari Nadeau
- Sean N. Parker Center for Allergy & Asthma Research Stanford University Stanford California
| | - Robyn E. O’Hehir
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Central Clinical School Monash University Melbourne Victoria Australia
- Allergy, Asthma and Clinical Immunology Service Alfred Health Melbourne Victoria Australia
| | - Liam O’Mahony
- Departments of Medicine and Microbiology, APC Microbiome Ireland National University of Ireland Cork Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy University Hospital Marburg, Philipps‐Universität Marburg Marburg Germany
| | - Maria J. Torres
- Allergy Unit Regional University Hospital of MalagaIBIMA‐UMA‐ARADyAL Malaga Spain
| | - De Yun Wang
- Department of Otolaryngology Yong Loo Lin School of Medicine National University of Singapore Singapore
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery and Department of Allergy Beijing Tongren Hospital Beijing China
| | - Cezmi A. Akdis
- Christine Kühne Center for Allergy Research and Education Davos Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich Davos Switzerland
| |
Collapse
|