1
|
Sahnoon L, Bajbouj K, Mahboub B, Hamoudi R, Hamid Q. Targeting IL-13 and IL-4 in Asthma: Therapeutic Implications on Airway Remodeling in Severe Asthma. Clin Rev Allergy Immunol 2025; 68:44. [PMID: 40257546 PMCID: PMC12011922 DOI: 10.1007/s12016-025-09045-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/22/2025]
Abstract
Asthma is a chronic respiratory disorder affecting individuals across all age groups. It is characterized by airway inflammation and remodeling and leads to progressive airflow restriction. While corticosteroids remain a mainstay therapy, their efficacy is limited in severe asthma due to genetic and epigenetic alterations, as well as elevated pro-inflammatory cytokines interleukin-4 (IL-4), interleukin-13 (IL-13), and interleukin-5 (IL-5), which drive structural airway changes including subepithelial fibrosis, smooth muscle hypertrophy, and goblet cell hyperplasia. This underscores the critical need for biologically targeted therapies. This review systematically examines the roles of IL-4 and IL-13, key drivers of type-2 inflammation, in airway remodeling and their potential as therapeutic targets. IL-4 orchestrates eosinophil recruitment, immunoglobulin class switching, and Th2 differentiation, whereas IL-13 directly modulates structural cells, including fibroblasts and epithelial cells, to promote mucus hypersecretion and extracellular matrix (ECM) deposition. Despite shared signaling pathways, IL-13 emerges as the dominant cytokine in remodeling processes including mucus hypersecretion, fibrosis and smooth muscle hypertrophy. While IL-4 primarily amplifies inflammatory cascades by driving IgE switching, promoting Th2 cell polarization that sustain cytokine release, and inducing chemokines to recruit eosinophils. In steroid-resistant severe asthma, biologics targeting IL-4/IL-13 show promise in reducing exacerbations and eosinophilic inflammation. However, their capacity to reverse established remodeling remains inconsistent, as clinical trials prioritize inflammatory biomarkers over long-term structural outcomes. This synthesis highlights critical gaps in understanding the durability of IL-4/IL-13 inhibition on airway structure and advocates for therapies combining biologics with remodeling-specific strategies. Through the integration of mechanistic insights and clinical evidence, this review emphasizes the need for long-term studies utilizing advanced imaging, histopathological techniques, and patient-reported outcomes to evaluate how IL-4/IL-13-targeted therapies alter airway remodeling and symptom burden, thereby informing more effective treatment approaches for severe, steroid-resistant asthma.
Collapse
Affiliation(s)
- Lina Sahnoon
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Khuloud Bajbouj
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Bassam Mahboub
- Rashid Hospital, Dubai Health, 4545, Dubai, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Division of Surgery and Interventional Science, University College London, London, UK.
- Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, Sharjah, United Arab Emirates.
| | - Qutayba Hamid
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Meakins-Christie Laboratories, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
2
|
Lewandowska AA, Rybacki C, Graczyk M, Waśniowska D, Kołodziej M. Is There a Place for Cannabinoids in Asthma Treatment? Int J Mol Sci 2025; 26:3328. [PMID: 40244178 PMCID: PMC11989515 DOI: 10.3390/ijms26073328] [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: 03/14/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
The beneficial effects of cannabinoids in the treatment of respiratory diseases have been drawing researchers' attention for several decades. Asthma is a complex disease entity characterized by a variable course, the treatment of which requires the continuous search for alternative, adjuvant treatment strategies designed for patients refractory to available pharmacotherapies. Cannabinoids exert certain physiological responses in the respiratory system due to their immunomodulatory properties and the strong presence of the endocannabinoid system in the lungs. In animal model studies, THC and CBD seem to counteract bronchoconstriction and inhibit pro-inflammatory mediation, respectively, which highlights their possible future contribution to the treatment of respiratory and allergic diseases, such as asthma. However, there are controversies regarding the health consequences of cannabis usage, the extracts' proportions, or equally safe and effective routes of administration, especially considering the alarming reports indicating an increased risk of asthma development among recreational cannabis smokers. The purpose of this review is to analyze the available literature on the influence of the endocannabinoid system, phytocannabinoids, and their modes of action on asthma pathogenesis in an attempt to assess their potential clinical relevance and determine future research directions.
Collapse
Affiliation(s)
- Agata Anna Lewandowska
- Clinical Department of Pulmonology, Allergology and Pulmonary Oncology, 10th Military Clinical Hospital with Polyclinic in Bydgoszcz, 85-681 Bydgoszcz, Poland (C.R.)
| | - Cezary Rybacki
- Clinical Department of Pulmonology, Allergology and Pulmonary Oncology, 10th Military Clinical Hospital with Polyclinic in Bydgoszcz, 85-681 Bydgoszcz, Poland (C.R.)
- Faculty of Medicine, Bydgoszcz University of Science and Technology, 85-796 Bydgoszcz, Poland
| | - Michał Graczyk
- Department of Palliative Care, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
| | - Dorota Waśniowska
- Clinical Department of Pulmonology, Allergology and Pulmonary Oncology, 10th Military Clinical Hospital with Polyclinic in Bydgoszcz, 85-681 Bydgoszcz, Poland (C.R.)
| | - Małgorzata Kołodziej
- Clinical Department of Oncology, Oncology Center of Professor Franciszek Łukaszczyk in Bydgoszcz, 85-796 Bydgoszcz, Poland
| |
Collapse
|
3
|
Soendergaard MB, Hansen S, Håkansson KEJ, von Bülow A, Bjerrum AS, Schmid JM, Johansson SL, Rasmussen LM, Johnsen CR, Bertelsen BB, Krogh NS, Hilberg O, Ulrik CS, Porsbjerg C. Early Reduction of FeNO on Anti-IL5 Biologics Is Associated With Clinical Remission of Severe Asthma. Allergy 2025; 80:986-995. [PMID: 39673455 DOI: 10.1111/all.16425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/10/2024] [Accepted: 11/03/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND In patients with severe asthma, treatment with anti-interleukin-5 (IL-5) biologics can lead to a reduction in fractional exhaled nitric oxide (FeNO) in some patients. The clinical implications of varying FeNO responses to anti-IL-5 biologics remain unclear. This study aims to categorise patients based on their FeNO response to anti-IL-5 biologics and evaluate the association of these categories with clinical outcomes. METHODS We used the Danish Severe Asthma Register (DSAR) to identify the early FeNO response profiles in patients receiving anti-IL5 biologics. We defined FeNO responders as patients with elevated FeNO levels at baseline and a decrease corresponding to the minimal clinically important difference (MCID) at 4 months of follow-up and FeNO non-responders as those who did not experience a decrease. RESULTS We identified 403 patients on anti-IL5 treatment in DSAR, and 265 (66%) had elevated FeNO levels at baseline. After 4 months of treatment, 151 (57%) patients showed a significant decrease in FeNO levels, and 114 (43%) did not. FeNO responders were more likely to achieve clinical remission of asthma (34% vs. 19%, p = 0.01, OR 2.11, CI 1.04, 5.18, p = 0.03) than FeNO non-responders after 12 months of treatment. The higher remission rates in FeNO responders mainly reflected a higher rate of normalisation of lung function. CONCLUSIONS FeNO levels were reduced after anti-IL5 treatment in a significant proportion of patients treated with anti-IL5, and this was associated with clinical remission. Early FeNO response to anti-IL5 could potentially be used as a biomarker to guide management decisions with biologics towards remission of disease in severe asthma.
Collapse
Affiliation(s)
| | - Susanne Hansen
- Department of Respiratory Medicine, Copenhagen University Hospital-Bispebjerg, Copenhagen, Denmark
- Centre for Clinical Research and Prevention, Frederiksberg Hospital, Copenhagen, Denmark
| | - Kjell Erik Julius Håkansson
- Department of Respiratory Medicine, Copenhagen University Hospital-Bispebjerg, Copenhagen, Denmark
- Department of Respiratory Medicine, Copenhagen University Hospital-Hvidovre, Hvidovre, Denmark
| | - Anna von Bülow
- Department of Respiratory Medicine, Copenhagen University Hospital-Bispebjerg, Copenhagen, Denmark
| | - Anne-Sofie Bjerrum
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | - Johannes Martin Schmid
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | | - Ole Hilberg
- Department of Respiratory Medicine, Sygehus Lillebaelt-Vejle Sygehus, Vejle, Denmark
| | - Charlotte Suppli Ulrik
- Department of Respiratory Medicine, Copenhagen University Hospital-Hvidovre, Hvidovre, Denmark
| | - Celeste Porsbjerg
- Department of Respiratory Medicine, Copenhagen University Hospital-Bispebjerg, Copenhagen, Denmark
| |
Collapse
|
4
|
Hui CW, Wu WC, Tong TK, Shiu C, Ng HL, Leung SO. Discovery of a new anti-γc antibody in clinical development for the treatment of autoimmune diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae020. [PMID: 40163668 DOI: 10.1093/jimmun/vkae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/11/2024] [Indexed: 04/02/2025]
Abstract
Autoimmune disease refers to a condition when the immune system anomalously attacks its own body and healthy cells. Although the exact causes of autoimmune diseases are unknown, it is recognized that excessive or aberrant cytokine responses contribute significantly to the development of autoimmunity. Among them, the common gamma c chain (γc) cytokines driven signaling cascade plays an indispensable role in driving pathogenic immune responses in patients with autoimmune diseases. Thus, we hypothesize that the development of an antibody targeting γc receptor could serve as a potential approach for treating autoimmune diseases and fulfil the unmet medical needs in this area. Here, we demonstrate that a humanized anti-γc antibody, hC2, could show high binding affinity to the human γc receptor and suppress 6 γc cytokines (interleukin [IL]-2, IL-4, IL-7, IL-9, IL-15 and IL-21)-driven STAT phosphorylation, leading to inhibition of autoimmunity and activation in B, T, and natural killer cell lines. Similar inhibitory effects were observed in the human peripheral blood mononuclear cell culture. Moreover, administration of hC2 could reduce expansion and tissue infiltration of T helper and cytotoxic T cells, leading to attenuation of damages to skin, liver, and kidney in the humanized xenograft mouse model. The current study demonstrates the potential of γc blockades for the treatment of T cell-mediated autoimmune diseases and chronic graft-versus-host disease. Anti-γc antibody hC2 might offer a more efficacious therapy compared with antibodies targeting a single γc cytokine and safer therapy than JAK inhibitors to fulfill the unmet medical needs in the autoimmune diseases in the future.
Collapse
Affiliation(s)
- Chin Wai Hui
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Wai Chung Wu
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Tak Keung Tong
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Carol Shiu
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Hoi Lam Ng
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Shui On Leung
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| |
Collapse
|
5
|
Zhong Y, Zhou Y, Tang L, Li W, Li Y, Gao H, Tao F, Wu X. Relationship of prenatal ambient air pollutants exposure with childhood asthma risk and underlying mechanism: Ma'anshan Birth Cohort study. ENVIRONMENTAL RESEARCH 2025; 274:121283. [PMID: 40043928 DOI: 10.1016/j.envres.2025.121283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/16/2025] [Accepted: 03/02/2025] [Indexed: 03/10/2025]
Abstract
INTRODUCTION Association between prenatal exposure to particulate matter speciation and childhood asthma was limited, and study of sensitive exposure window was needed. METHODS Among 1807 children from Ma'anshan Birth Cohort, childhood asthma information was obtained by standardized questionnaire. Family address was collected at birth and in 7 years old and used to assess trimester-specific ambient air pollutants (AAPs) exposure. Restricted cubic spline and mixed effects logistic regression were applied to assess association of AAPs with childhood asthma, stratified by sex. Moreover, potential mechanism of AAPs-childhood asthma association was revealed by constructing adverse outcome pathway. RESULTS There were significant correlations between AAPs. During the third trimester, exposure to PM2.5 exhibited a nearly J-shaped association with childhood asthma risk. When compared to the lowest tertile, childhood asthma risk increased by 59% (95% CI: 1.00-2.52) among children within the highest tertile of PM2.5 exposure (>76.65 μg/m3); and each unit increase in log2-transformed PM2.5 was associated with 102% (95% CI: 1.24-3.27) increase in childhood asthma risk. For chemical compositions of PM2.5, exposure to Cl-, NO3-, NH4+, and NO3- was also significantly associated with increased childhood asthma risk in the third trimester, especially in boys. Up-expression of IL-4 is molecular initiation event in the AAPs-asthma association, followed by decreased fibrinolysis, activated bradykinin, increased proinflammatory mediators, and recruitment of inflammatory cells, ultimately causing hyperinflammation. CONCLUSIONS Association of AAPs with asthma risk varied by trimester and sex, particularly PM2.5. Our findings enhance the public awareness of air pollution, heighten the importance of monitoring and control of AAPs.
Collapse
Affiliation(s)
- Yumei Zhong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yun Zhou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Lijun Tang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Wenge Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yuxi Li
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Hui Gao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xiulong Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
6
|
Castro M, Papi A, Porsbjerg C, Lugogo NL, Brightling CE, González-Barcala FJ, Bourdin A, Ostrovskyy M, Staevska M, Chou PC, Duca L, Pereira AM, Fogarty C, Nadama R, Zhang M, Rodrigues A, Soler X, Sacks HJ, Deniz Y, Rowe PJ, de Prado Gómez L, Jacob-Nara JA. Effect of dupilumab on exhaled nitric oxide, mucus plugs, and functional respiratory imaging in patients with type 2 asthma (VESTIGE): a randomised, double-blind, placebo-controlled, phase 4 trial. THE LANCET. RESPIRATORY MEDICINE 2025; 13:208-220. [PMID: 39947221 DOI: 10.1016/s2213-2600(24)00362-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 03/08/2025]
Abstract
BACKGROUND Asthma is a respiratory disease characterised by chronic airway inflammation and mucus hypersecretion. VESTIGE used functional respiratory imaging to assess changes in airway structure and function, including mucus plugging, in response to dupilumab. METHODS VESTIGE was a randomised, double-blind, placebo-controlled, phase 4 trial done at 72 research sites or academic centres in 14 countries. We recruited adult patients (aged 18-70 years) with physician-diagnosed, uncontrolled, moderate-to-severe type 2 asthma (blood eosinophil count ≥300 cells/μL and fractional exhaled nitric oxide [FeNO] ≥25 parts per billion [ppb]) being treated with medium-dose to high-dose inhaled corticosteroids combined with other controller medications. Patients were randomly assigned (2:1; block size of 6) via interactive voice-web response technology to receive add-on dupilumab 300 mg subcutaneously once every 2 weeks or volume-matched placebo up to week 24. Randomisation was stratified by inhaled corticosteroids dose level and region (eastern Europe vs the rest of the world). Participants and investigators, including those assessing outcomes, were masked to group assignment. The primary endpoints were the proportion of patients with a FeNO concentration below 25 ppb at week 24, and percentage change from baseline to week 24 in airway volumes (specific regional airway volumes corrected for lung volume, [s]iVaw) at total lung capacity (TLC), both assessed in the intention-to-treat population. Safety was assessed in all patients who received at least one dose of study drug or placebo. The trial is registered with ClinicalTrials.gov, NCT04400318, and is completed. FINDINGS Patient recruitment occurred from July 18, 2020, to Jan 6, 2023. Patients (mean age 50·4 years [SD 12·6]; 68 [62%] female and 41 [38%] male) were randomly assigned to receive dupilumab 300 mg (n=72) or placebo (n=37). At week 24, patients in the dupilumab group were significantly more likely than those in the placebo group to have a FeNO concentration below 25 ppb (41 [57%] of 72 patients vs four [11%] of 37; odds ratio: 9·8 [95% CI 3·1 to 30·8]; p<0·001). Treatment with dupilumab versus placebo led to a numerical increase in (s)iVaw at TLC from baseline to week 24, although the difference was not significant (least squares [LS] mean percentage change from baseline to week 24: 19·7% [SE 8·1] for dupilumab and -2·0% [11·5] for placebo; LS mean difference vs placebo: 21·8% [95% CI -7·7 to 51·3]; p=0·14). Safety was consistent with the reported safety profile for dupilumab. Treatment-emergent adverse events related to study intervention were reported in 11 (15%) of 72 patients who received dupilumab and four (11%) of 37 who received placebo; no deaths occurred during the intervention period. INTERPRETATION The full results of this study indicate that dupilumab reduced airway inflammation and mucus plugging, and improved airway volume and flow, corresponding to improved lung function and asthma control. This study highlights the potential of imaging technology to assess disease burden, monitor progression, and evaluate therapeutic responses, which can provide valuable insights to guide clinical decision making for patients with uncontrolled, moderate-to-severe type 2 asthma. FUNDING Sanofi and Regeneron Pharmaceuticals.
Collapse
Affiliation(s)
- Mario Castro
- Department of Medicine, University of Kansas School of Medicine, Kansas City, KS, USA.
| | - Alberto Papi
- Department of Cardiorespiratory Medicine, Respiratory Medicine Unit, University of Ferrara, S Anna University Hospital, Ferrara, Italy
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Njira L Lugogo
- Michigan Medicine Asthma Program, University of Michigan, Ann Arbor, MI, USA
| | - Christopher E Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Francisco-Javier González-Barcala
- Neumología, Hospital Clínico Universitario de Santiago de Compostela-Grupo de Investigación TRIAD, Santiago de Compostela, Spain; Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Arnaud Bourdin
- Department of Respiratory Diseases, PhyMedExp, University of Montpellier, INSERM CNRS, Montpellier, France
| | - Mykola Ostrovskyy
- Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | | | | | | | - Ana Margarida Pereira
- Allergy Unit, Instituto and Hospital CUF, Porto, Portugal; Patient Centered Innovation and Technologies, Center for Health Technology and Services Research, Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Rufai Nadama
- King Khalid University Hospital, Riyadh, Saudi Arabia
| | | | | | | | | | - Yamo Deniz
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | |
Collapse
|
7
|
Latayan J, Akkenapally SV, Madala SK. Emerging Concepts in Cytokine Regulation of Airway Remodeling in Asthma. Immunol Rev 2025; 330:e70020. [PMID: 40116139 PMCID: PMC11926778 DOI: 10.1111/imr.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Asthma, a chronic respiratory condition that has seen a dramatic rise in prevalence over the past few decades, now affects more than 300 million people globally and imposes a significant burden on healthcare systems. The key pathological features of asthma include inflammation, airway hyperresponsiveness, mucus cell metaplasia, smooth muscle hypertrophy, and subepithelial fibrosis. Cytokines released by lung epithelial cells, stromal cells, and immune cells during asthma are critical to pathological tissue remodeling in asthma. Over the past few decades, researchers have made great strides in understanding key cells involved in asthma and the cytokines that they produce. Epithelial cells as well as many adaptive and innate immune cells are activated by environmental signals to produce cytokines, namely, type 2 cytokines (IL-4, IL-5, IL-13), IFN-γ, IL-17, TGF-β, and multiple IL-6 family members. However, the precise mechanisms through which these cytokines contribute to airway remodeling remain elusive. Additionally, multiple cell types can produce the same cytokines, making it challenging to decipher how specific cell types and cytokines uniquely contribute to asthma pathogenesis. This review highlights recent advances and provides a comprehensive overview of the key cells involved in the production of cytokines and how these cytokines modulate airway remodeling in asthma.
Collapse
Affiliation(s)
- Jana Latayan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
- Immunology Graduate ProgramUniversity of CincinnatiCincinnatiOhioUSA
| | - Santhoshi V. Akkenapally
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| | - Satish K. Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| |
Collapse
|
8
|
Zhang W, Li L, Zhang Y, Dai J, Qiu C, Chen R, Shi F. Perturbations in the airway microbiome are associated with type 2 asthma phenotype and severity. Ann Allergy Asthma Immunol 2025; 134:296-305.e9. [PMID: 39549985 DOI: 10.1016/j.anai.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/14/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Airway microbiome has been linked to asthma heterogeneity, yet little is known about the associations between airway microbiota and type 2 (T2) asthma phenotype and severity. OBJECTIVE To determine the relationship of nasopharyngeal (NP) and induced sputum (IS) microbiota to the phenotypic features of T2 asthma. METHODS NP and IS samples from subjects with T2 mild-to-moderate asthma (n = 23), subjects with severe asthma (n = 21), and healthy controls (n = 16) were analyzed. Bacterial microbiota and functional profiles were compared. The correlation between microbial communities and clinical and inflammatory features was evaluated in individuals with asthma of 2 statuses. RESULTS Differences in NP and IS microbiota were associated with T2 asthma phenotype. Alterations in NP microbiota were more reflective of T2 inflammation and severity, with additional stratification of a subgroup characterized by significant elevations in T2 inflammatory biomarkers and reductions in bacterial richness and diversity (P < .05). Burkholderia-Caballeronia-Paraburkholderia, Ralstonia, and Rhodococcus were identified as hub taxa within NP microbial network in T2 severe asthma, which were prevalent in the entire airway and involved in bacterial functions including inflammatory and steroid responses (P < .05). The composition and diversity of IS microbiota were complex, with Veillonella as the most altered genus, having an increase with increasing asthma severity. CONCLUSION Our work revealed the significant associations of microbiota perturbations throughout the entire respiratory tract to the extent of T2 inflammation, phenotype and severity in T2 asthma. The specific taxa identified invite further mechanistic investigations to unravel their possibility as biomarkers and therapeutic targets for T2 severe asthma.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Infectious Diseases, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Lifei Li
- Key Laboratory of Shenzhen Respiratory Diseases, Institute of Shenzhen Respiratory Diseases, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People's Republic of China
| | - Yu Zhang
- Department of Infectious Diseases, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Junjie Dai
- Department of Infectious Diseases, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen, People's Republic of China
| | - Chen Qiu
- Key Laboratory of Shenzhen Respiratory Diseases, Institute of Shenzhen Respiratory Diseases, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People's Republic of China
| | - Rongchang Chen
- Key Laboratory of Shenzhen Respiratory Diseases, Institute of Shenzhen Respiratory Diseases, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People's Republic of China
| | - Fei Shi
- Department of Infectious Diseases, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen, People's Republic of China.
| |
Collapse
|
9
|
Santiago LF, da Silva ES, Dos Santos PS, Salazar-Garcés LF, Santos SPO, Fernandes AMS, Silva RC, Alves VS, Briza P, Ferreira F, Pacheco LGC, Alcantara-Neves NM, Pinheiro CS. The proteome of human adult whipworm Trichuris trichiura: A source of potential immunomodulatory molecules. Acta Trop 2025; 263:107566. [PMID: 39988282 DOI: 10.1016/j.actatropica.2025.107566] [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: 10/15/2024] [Revised: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Soil-transmitted helminths (STHs), including Trichuris trichiura, pose a major global health challenge, particularly in children, causing significant morbidity. However, T. trichiura's ability to modulate host immune responses offers a unique opportunity to discover biomolecules with therapeutic potential for inflammatory, allergic, and autoimmune disorders. This study conducted a proteomic analysis of adult male and female T. trichiura using liquid chromatography-tandem mass spectrometry (LC-MS/MS), identifying 810 parasite proteins. Of these, 177 were exclusive to females, 277 to males, and 356 shared. Gene ontology analysis showed similar cellular component profiles in males and females, mostly involving intracellular structures. However, female-exclusive proteins exhibited more diverse components. Molecular function analysis highlighted hydrolytic and catalytic activities, suggesting enzymatic strategies for nutrition and immune modulation. Notably, immunomodulatory proteins were identified in both sexes, showing therapeutic potential, including a Kunitz protease inhibitor and glutamate dehydrogenase. To evaluate immunomodulatory properties, one identified protein (rc4299) was tested on cultures of peripheral blood mononuclear cells (PBMCs) from allergic individuals. The recombinantly produced rc4299 increased IL-10 secretion, indicating potential for treating autoimmune and allergic diseases. This study uncovers the T. trichiura proteome and highlights promising therapeutic targets, emphasizing the parasite's complex interactions with the host immune system.
Collapse
Affiliation(s)
- Leonardo F Santiago
- Institute of Health Science - ICS, Federal University of Bahia, Salvador, Brazil
| | - Eduardo S da Silva
- Institute of Health Science - ICS, Federal University of Bahia, Salvador, Brazil
| | | | | | - Sara P O Santos
- Institute of Health Science - ICS, Federal University of Bahia, Salvador, Brazil
| | | | - Raphael C Silva
- Institute of Health Science - ICS, Federal University of Bahia, Salvador, Brazil
| | - Vitor S Alves
- Institute of Health Science - ICS, Federal University of Bahia, Salvador, Brazil
| | - Peter Briza
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Fatima Ferreira
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Luis G C Pacheco
- Institute of Health Science - ICS, Federal University of Bahia, Salvador, Brazil
| | | | - Carina S Pinheiro
- Institute of Health Science - ICS, Federal University of Bahia, Salvador, Brazil.
| |
Collapse
|
10
|
Quinn AE, Zhao L, Bell SD, Huq MH, Fang Y. Exploring Asthma as a Protective Factor in COVID-19 Outcomes. Int J Mol Sci 2025; 26:1678. [PMID: 40004141 PMCID: PMC11855143 DOI: 10.3390/ijms26041678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Asthma has long been associated with increased susceptibility to viral respiratory infections, leading to significant exacerbations and poorer clinical outcomes. Contrarily and interestingly, emerging data and research surrounding the COVID-19 pandemic have shown that patients with asthma infected with SARS-CoV-2 experienced decreased severity of disease, lower hospitalization rates, as well as decreased morbidity and mortality. Research has shown that eosinophils could enhance immune defense against viral infections, while inhaled corticosteroids can assist in controlling systematic inflammation. Moreover, reduced ACE-2 expression in individuals with asthma may restrict viral entry, and the Th2 immune response may offset the Th1 response typically observed in severe COVID-19 patients. These factors may help explain the favorable outcomes seen in asthmatic patients during the COVID-19 pandemic. This review highlights potential protective mechanisms seen in asthmatic patients, including eosinophilia, the use of inhaled corticosteroids, reduced ACE-2 expression, and a dominate Th2 immune response. Such a study will be helpful to better manage patients with asthma who have contracted COVID-19.
Collapse
Affiliation(s)
- Anthony E. Quinn
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China;
| | - Scott D. Bell
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Muhammad H. Huq
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
11
|
Fairweather SJ, Hammerton G, Paternoster L, Gilbody S, Jones HJ, Khandaker GM. Childhood allergy and anxiety/depression in early adulthood: A longitudinal study in the ALSPAC birth cohort. Brain Behav Immun 2025; 124:226-236. [PMID: 39662640 DOI: 10.1016/j.bbi.2024.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/30/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Allergic disease and common mental disorders frequently co-occur. However, little is known about the longitudinal impact of childhood allergy on the subsequent risk of developing anxiety or depression, and the possible biological mechanisms for this. METHODS We performed longitudinal analyses of data from the Avon Longitudinal Study of Parents and Children (ALSPAC) birth cohort. The baseline sample comprised n = 5256 children with allergy data available at age 7yrs. We used multivariable regression to test associations between childhood allergy at age 7yrs and: a) four inflammatory markers at age 9yrs; b) depression and anxiety measures between ages 10-24yrs. Allergy measures included biological markers (total serum immunoglobulin E (tIgE), number of positive skin prick tests (SPTs)), and presence of eczema, asthma and/or food allergy (mother reported). Inflammatory markers were interleukin-6 (IL-6), C-reactive protein (CRP), IL-4 and IL-13. We used structural equation modelling to test whether inflammatory markers mediated the association between tIgE and depression/anxiety. RESULTS tIgE and having ≥ 1 positive SPT at age 7 were associated with IL-6 levels at age 9 (adjusted β = 0.09; 95 % CI 0.06-0.13; p < 0.001 and adjusted β = 0.06; 95 % CI 0.03-0.09; p < 0.001 respectively), but not with CRP, IL-4 or IL13 levels. We found no strong evidence of an association between childhood allergy and subsequent depression/anxiety during adolescence and early adulthood. This finding was consistent across biological and mother-reported allergy measures. CONCLUSIONS Biological markers of childhood allergy are associated with IL-6, a key inflammatory cytokine. However, childhood allergy may not have a large long-term effect on subsequent depression/anxiety.
Collapse
Affiliation(s)
- Sophie J Fairweather
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Population Health Sciences, Bristol Medical School, Bristol, UK; NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK; Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Gemma Hammerton
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Population Health Sciences, Bristol Medical School, Bristol, UK; Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lavinia Paternoster
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Population Health Sciences, Bristol Medical School, Bristol, UK; NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Simon Gilbody
- Mental Health and Addiction Research Group, Department of Health Sciences, University of York, York YO10 5DD, UK
| | - Hannah J Jones
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Population Health Sciences, Bristol Medical School, Bristol, UK; NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK; Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Golam M Khandaker
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Population Health Sciences, Bristol Medical School, Bristol, UK; NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK; Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; Avon and Wiltshire Mental Health Partnership NHS Trust, Bristol, UK
| |
Collapse
|
12
|
Pastore D, Lupia C, D'Amato M, Bruni A, Garofalo E, Longhini F, Gallelli L, Vatrella A, Pelaia G, Pelaia C. Emerging biological treatments for asthma. Expert Opin Emerg Drugs 2025:1-11. [PMID: 39873193 DOI: 10.1080/14728214.2025.2460529] [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: 10/06/2024] [Revised: 12/24/2024] [Accepted: 01/27/2025] [Indexed: 01/30/2025]
Abstract
INTRODUCTION Severe asthma is a chronic airway disease characterized by many pathomechanisms known as endotypes. Biological therapies targeting severe asthma endotypes have significantly improved the treatment of this disease, thus remarkably bettering patient quality of life. AREAS COVERED This review aims to describe current biological therapies for severe asthma, highlighting emerging ones. Several studies have confirmed the beneficial effects of currently available monoclonal antibodies targeting immunoglobulin E (IgE), interleukin-5 (IL-5) or its receptor, and interleukin-4 (IL-4)/interleukin-13 (IL-13) receptors (IL-4R/IL-13R). However, patients with T2-low asthma are not eligible for the above biological therapies. EXPERT OPINION New treatments are now moving toward targeting the upstream pathways of asthma pathogenesis, coordinated by innate cytokines such as alarmins. These key proinflammatory mediators orchestrate the activation of complex cellular networks including both innate and adaptive immune responses. Alarmins include thymic stromal lymphopoietin (TSLP), interleukin-25 (IL-25), and interleukin-33 (IL-33), which are released from injured airway epithelial cells. TSLP and the other alarmins are suitable targets of biological therapies which are effective for add-on treatment of type 2 asthma. Moreover, anti-alarmin monoclonal antibodies can be also beneficial for patients with T2-low, poorly controlled severe asthma.
Collapse
Affiliation(s)
- Daniela Pastore
- Department of Health Sciences,University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Chiara Lupia
- Department of Health Sciences,University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Maria D'Amato
- Department of Respiratory Medicine, "V. Monaldi University Hospital, Naples, Italy
| | - Andrea Bruni
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Eugenio Garofalo
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Federico Longhini
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Luca Gallelli
- Department of Health Sciences,University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Girolamo Pelaia
- Department of Health Sciences,University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
13
|
Khalid Salah Al-Sheakly B, Saheb Sharif-Askari F, Saheb Sharif-Askari N, Hundt JE, Halwani R. The potential role of nanobodies in asthma therapy. Front Pharmacol 2025; 15:1510806. [PMID: 39902079 PMCID: PMC11788342 DOI: 10.3389/fphar.2024.1510806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025] Open
Abstract
Asthma is a chronic inflammatory disease of the airways characterized by bronchoconstriction, airway hyperresponsiveness, and mucus production. The pathophysiology of asthma involves a complex interplay of immune cells and mediators, including cytokines, chemokines, and other inflammatory molecules. Despite advances in asthma management, many patients continue to experience symptoms due to the limitations of current therapies. Monoclonal antibodies (mAbs) targeting specific inflammatory mediators have improved treatment outcomes for some patients, but challenges such as poor tissue penetration and high costs remain. Nanobodies (Nbs), a novel class of single-domain antibodies, offer a promising alternative due to their small size, stability, and potential for enhanced tissue penetration. This review discusses the key mediators involved in asthma, challenges in current treatments, and the potential of Nbs as a new therapeutic strategy. We also explore current studies and innovations in nanobody technology.
Collapse
Affiliation(s)
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jennifer E. Hundt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pediatrics, Faculty of Medicine, Prince Abdullah Ben Khaled Celiac Disease Chair, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Nolasco S, Mukherjee M, Nair P. Trajectories of responses to mepolizumab in severe asthma. Eur Respir J 2025; 65:2402023. [PMID: 39884757 DOI: 10.1183/13993003.02023-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 02/01/2025]
Affiliation(s)
- Santi Nolasco
- Research Institute of St. Joe's, St Joseph's Healthcare, Hamilton, Hamilton, ON, Canada
| | - Manali Mukherjee
- Research Institute of St. Joe's, St Joseph's Healthcare, Hamilton, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Parameswaran Nair
- Research Institute of St. Joe's, St Joseph's Healthcare, Hamilton, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
15
|
Brooks SG, King J, Smith JA, Yosipovitch G. Cough and itch: Common mechanisms of irritation in the throat and skin. J Allergy Clin Immunol 2025; 155:36-52. [PMID: 39321991 DOI: 10.1016/j.jaci.2024.09.012] [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: 06/19/2024] [Revised: 08/14/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
Cough and itch are protective mechanisms in the body. Cough occurs as a reflex motor response to foreign body inhalation, while itch is a sensation that similarly evokes a scratch response to remove irritants from the skin. Both cough and itch can last for sustained periods, leading to debilitating chronic disorders that negatively impact quality of life. Understanding the parallels and differences between chronic cough and chronic itch may be paramount to developing novel therapeutic approaches. In this article, we identify connections in the mechanisms contributing to the complex cough and scratch reflexes and summarize potential shared therapeutic targets. An online search was performed using various search engines, including PubMed, Web of Science, Google Scholar, and ClinicalTrials.gov from 1983 to 2024. Articles were assessed for quality, and those relevant to the objective were analyzed and summarized. The literature demonstrated similarities in the triggers, peripheral and central nervous system processing, feedback mechanisms, immunologic mediators, and receptors involved in the cough and itch responses, with the neuronal sensitization processes exhibiting the greatest parallels between cough and itch. Given the substantial impact on quality of life, novel therapies targeting similar neuroimmune pathways may apply to both itch and cough and provide new avenues for enhancing their management.
Collapse
Affiliation(s)
- Sarah G Brooks
- Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, Fla
| | - Jenny King
- Division of Immunology, Immunity to Infection, and Respiratory Medicine, Wythenshawe Hospital, University of Manchester, Manchester, United Kingdom; North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Jaclyn Ann Smith
- Division of Immunology, Immunity to Infection, and Respiratory Medicine, Wythenshawe Hospital, University of Manchester, Manchester, United Kingdom; North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Gil Yosipovitch
- Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, Fla.
| |
Collapse
|
16
|
Hagino T, Saeki H, Fujimoto E, Kanda N. Predictive Factors for Long-Term High Responders to Upadacitinib Treatment in Patients with Atopic Dermatitis. Dermatitis 2025; 36:62-71. [PMID: 39311694 DOI: 10.1089/derm.2024.0230] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Background: Upadacitinib, a Janus kinase 1 inhibitor, is an effective medicine for moderate-to-severe atopic dermatitis (AD). Identifying long-term responders to upadacitinib is crucial for optimal treatment strategies in real-world clinical practice. To identify predictive factors for long-term high responders to upadacitinib 15 mg or 30 mg, defined as achievers of investigator's global assessment (IGA) 0/1 with ≥2-point improvement from baseline IGA at week 48. Methods: A retrospective study was conducted from August 2021 to September 2023 on 63 AD patients treated with upadacitinib 15 mg and 31 patients with 30 mg. Patients of each group were categorized into long-term high responders (achievers of IGA 0/1 at week 48) and low responders (non-achievers). We compared baseline values of clinical indexes and laboratory parameters between long-term responders and nonresponders. Results: In 15 mg group, long-term high responders showed lower rate of bronchial asthma (BA), lower values of baseline eczema area and severity index (EASI) of head and neck, IgE, and systemic inflammatory response index (SIRI) compared with low responders. In 30 mg group, long-term high responders showed lower baseline levels of IgE compared with low responders. Conclusion: Patients with lower baseline EASI of head and neck, IgE, or SIRI or without BA and those with lower baseline IgE may have a higher potential to become long-term high responders to upadacitinib 15 mg and 30 mg treatment, respectively.
Collapse
Affiliation(s)
- Teppei Hagino
- From the Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Japan
| | - Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | | | - Naoko Kanda
- From the Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Japan
| |
Collapse
|
17
|
Nishi K, Matsumoto H, Sunadome H, Nagasaki T, Oguma T, Tashima N, Hayashi Y, Terada S, Morita K, Yoshimura C, Nishizaka Y, Sano A, Iwanaga T, Sano H, Haraguchi R, Tohda Y, Kawaguchi T, Matsuda F, Hirai T. IL1RL1 variant may affect the response to type 2 biologics in patients with severe asthma. ERJ Open Res 2025; 11:00448-2024. [PMID: 39811553 PMCID: PMC11726575 DOI: 10.1183/23120541.00448-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/24/2024] [Indexed: 01/16/2025] Open
Abstract
Background Asthma is a heterogeneous disease with variable response to treatment. Genetic backgrounds are involved in the severity of type 2 asthma, but their effects on responses to biologics remain unknown. This study aimed to clarify the role of genetic factors in response to biologics in patients with severe asthma. Methods Adults with severe asthma receiving biologics were enrolled in this multicentre, observational, real-world study. The responses to biologics were evaluated using Physicians' Global Evaluation of Treatment Effectiveness (GETE). Optimal biologic for each patient was also determined based on the best GETE score for the biologic used or currently used biologic. Three single nucleotide polymorphisms (IL1RL1, rs1420101; IL4RA, rs8832; and TSLP rs1837253) were examined. Results Among the 113 patients analysed, 53 (46.9%) had an excellent GETE score for at least one biologic. These patients with an excellent GETE score for at least one biologic, particularly for benralizumab, had the risk genotype of rs1420101 more frequently than the remaining patients, independent of the clinical demographics. Regarding the optimal biologic for each patient, anti-IL-5 drugs were optimal for patients with the rs1420101 TT or rs8832 GG genotype. Furthermore, dupilumab was similarly effective, regardless of the risk genotypes examined in this study. Conclusion IL1RL1 rs1420101 TT genotype and/or IL4RA rs8832 GG genotype may predict an excellent or optimal response to biologic therapy in each patient, particularly to anti-interleukin-5 targeted therapy. The elucidation of genetic predisposition may improve the management of severe asthma in the era of biologics.
Collapse
Affiliation(s)
- Kenta Nishi
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hironobu Sunadome
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tadao Nagasaki
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Respiratory Medicine and Allergology, Kindai University Nara Hospital, Nara, Japan
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Respiratory Medicine, Kyoto City Hospital, Kyoto, Japan
| | - Noriyuki Tashima
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Respiratory Medicine, Medical Research Institute Kitano Hospital, PIIF Tazuke-kofukai, Osaka, Japan
| | - Yusuke Hayashi
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoru Terada
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kyohei Morita
- Department of Respiratory Medicine, Osaka Red Cross Hospital, Osaka, Japan
| | - Chie Yoshimura
- Department of Respiratory Medicine, Osaka Red Cross Hospital, Osaka, Japan
| | - Yasuo Nishizaka
- Department of Respiratory Medicine, Osaka Red Cross Hospital, Osaka, Japan
| | - Akiko Sano
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takashi Iwanaga
- Center for General Medical Education and Clinical Training, Kindai University Hospital, Osaka, Japan
| | - Hiroyuki Sano
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Ryuta Haraguchi
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yuji Tohda
- Kindai University Hospital, Osaka, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Kleniewska P, Pawliczak R. The Link Between Dysbiosis, Inflammation, Oxidative Stress, and Asthma-The Role of Probiotics, Prebiotics, and Antioxidants. Nutrients 2024; 17:16. [PMID: 39796449 PMCID: PMC11722634 DOI: 10.3390/nu17010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Asthma (a chronic inflammatory disease of the airways) is characterized by a variable course, response to treatment, and prognosis. Its incidence has increased significantly in recent decades. Unfortunately, modern lifestyle and environmental factors contribute to the further increase in the incidence of this disease. Progressive industrialization and urbanization, widespread use of antibiotic therapy, excessive sterility and inappropriate, highly processed diets are some of the many risk factors that are relevant today. Over the years, a lot of evidence has been gathered showing the influence of microorganisms of the gut or airways on human health. Studies published in recent years indicate that dysbiosis (microbial imbalance) and oxidative stress (pro-oxidant-antioxidant imbalance) are important elements of the pathogenesis of this inflammatory disease. Scientists have attempted to counteract the effects of this process by using probiotics, prebiotics, and antioxidants. The use of probiotic microorganisms positively modulates the immune system by maintaining homeostasis between individual fractions of immune system cells. Moreover, recently conducted experiments have shown that probiotics have antioxidant, anti-inflammatory, and protective properties in oxidative stress (OS). The aim of this study is to present the current state of knowledge on the role of dysbiosis and OS in the pathogenesis of asthma. Conclusions: This review highlights the importance of using probiotics, prebiotics, and antioxidants as potential strategies to support the treatment and prevention of this disease.
Collapse
Affiliation(s)
- Paulina Kleniewska
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland;
| | | |
Collapse
|
19
|
Januskevicius A, Vasyle E, Rimkunas A, Palacionyte J, Kalinauskaite-Zukauske V, Malakauskas K. Serum T2-High Inflammation Mediators in Eosinophilic COPD. Biomolecules 2024; 14:1648. [PMID: 39766355 PMCID: PMC11674300 DOI: 10.3390/biom14121648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/08/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Eosinophils are central inflammatory cells in asthma; however, a portion of patients with chronic obstructive pulmonary disease (COPD) have blood or sputum eosinophilia, a condition termed eosinophilic COPD (eCOPD), which may contribute to the progression of the disease. We hypothesize that eosinophilic inflammation in eCOPD patients is related to Type 2 (T2)-high inflammation seen in asthma and that serum mediators might help us to identify T2-high inflammation in patients and choose an appropriate personalized treatment strategy. Thus, we aimed to investigate ten serum levels of T2-high inflammation mediators in eCOPD patients and compare them to severe non-allergic eosinophilic asthma (SNEA) patients. We included 8 subjects with eCOPD, 10 with SNEA, and 11 healthy subjects (HS) as a control group. The concentrations of biomarkers in serum samples were analyzed using an enzyme-linked immunosorbent assay (ELISA). In this study, we found that eCOPD patients were distinguished from SNEA patients by elevated serum levels of sIL-5Rα, MET, TRX1, ICTP, and IL-4, as well as decreased serum levels of eotaxin-1 and sFcεRI. Moreover, MET, ICTP, eotaxin-1, and sFcεRI demonstrated high sensitivity and specificity as potential biomarkers for eCOPD patients. Furthermore, serum levels of IL-5 and IL-25 in combination with sIL-5Rα, MET, and IL-4 demonstrated a high value in identifying T2-high inflammation in eCOPD patients. In conclusion, this study highlights that while T2-high inflammation drives eosinophilic inflammation in both eCOPD and SNEA through similar mechanisms, the distinct expression of its mediators reflects an imbalance between T1 and T2 inflammation pathways in eCOPD patients. A combined analysis of serum mediators may aid in identifying T2-high inflammation in eCOPD patients and in selecting an appropriate personalized treatment strategy.
Collapse
Affiliation(s)
- Andrius Januskevicius
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (E.V.); (A.R.); (K.M.)
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Egle Vasyle
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (E.V.); (A.R.); (K.M.)
| | - Airidas Rimkunas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (E.V.); (A.R.); (K.M.)
| | - Jolita Palacionyte
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (J.P.); (V.K.-Z.)
| | | | - Kestutis Malakauskas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (E.V.); (A.R.); (K.M.)
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (J.P.); (V.K.-Z.)
| |
Collapse
|
20
|
Choudhary I, Lamichhane R, Singamsetty D, Vo T, Brombacher F, Patial S, Saini Y. Cell-Specific Contribution of IL-4 Receptor α Signaling Shapes the Overall Manifestation of Allergic Airway Disease. Am J Respir Cell Mol Biol 2024; 71:702-717. [PMID: 39254378 PMCID: PMC11622633 DOI: 10.1165/rcmb.2024-0208oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/10/2024] [Indexed: 09/11/2024] Open
Abstract
IL-4 and IL-13 play a critical role in allergic asthma pathogenesis via their common receptor IL-4Rα. However, the cell-specific role of IL-4Rα in mixed allergen (MA)-induced allergic asthma has remained unclear. Therefore, we aimed to identify the cell-specific contribution of IL-4Rα signaling in the manifestation of various pathological outcomes in mice with allergic airway disease. We compared MA-induced pathological outcomes between hematopoietic progenitor cell (HPC)- or non-HPC-specific IL-4Rα-deficient chimera, myeloid cell-specific IL-4Rα-deficient (LysMcre+/+IL-4Rαfl/fl), and airway epithelial cell-specific IL-4Rα-deficient (CCSP-Cre+/IL-4Rαfl/fl) mice. Chimeric mice with systemic IL-4Rα sufficiency displayed hallmark features of allergic asthma, including eosinophilic and lymphocytic infiltration, type 2 (T-helper type 2) cytokine/chemokine production, IgE production, and lung pathology. These features were markedly reduced in chimeric mice with systemic IL-4Rα deficiency. Non-HPC-specific IL-4Rα-deficient mice displayed typical inflammatory features of allergic asthma but with markedly reduced mucous cell metaplasia (MCM). Deletion of IL-4Rα signaling on airway epithelial cells, a subpopulation within the non-HPC lineage, resulted in almost complete absence of MCM. In contrast, all features of allergic asthma except for MCM and mucin production were mitigated in HPC-specific IL-4Rα-deficient chimeric mice. Deleting IL-4Rα signaling in myeloid cells, a subpopulation within the HPC lineage, significantly alleviated MA-induced allergic airway inflammatory responses, but, similar to the HPC-specific IL-4Rα-deficient chimeric mice, these mice showed significant MCM and mucin production. Our findings demonstrate that the differential allergen responsiveness seen in mice with HPC-specific and non-HPC-specific IL-4Rα deficiency is predominantly driven by the absence of IL-4Rα in myeloid cells and airway epithelial cells, respectively. Our findings also highlight distinct and mutually exclusive roles of IL-4Rα signaling in mediating pathological outcomes within the myeloid and airway epithelial cell compartments.
Collapse
Affiliation(s)
- Ishita Choudhary
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Richa Lamichhane
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Dhruthi Singamsetty
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Thao Vo
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology and Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town, South Africa; and
| | - Sonika Patial
- National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Yogesh Saini
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
21
|
Weare-Regales N, Carr T, Holguin F, Tibbitt CA, Lockey RF. Obesity and hormonal influences on asthma: Mechanisms, management challenges, and emerging therapeutic strategies. J Allergy Clin Immunol 2024; 154:1355-1368. [PMID: 39362350 DOI: 10.1016/j.jaci.2024.09.018] [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: 06/27/2024] [Revised: 08/13/2024] [Accepted: 09/13/2024] [Indexed: 10/05/2024]
Abstract
Obesity and hormone dysregulation, common comorbidities of asthma, not only influence asthma risk and onset but can also complicate its management. The pathobiologic characteristics of obesity, such as insulin resistance and metabolism alterations, can impact lung function and airway inflammation while highlighting potential opportunities for therapeutic intervention. Likewise, obesity alters immune cell phenotypes and corticosteroid pharmacokinetics. Hormones such as sex hormones, incretins, and thyroid hormones can also affect asthma. This review highlights the mechanisms underlying obesity-related asthma and hormonal pathologies while exploring potential therapeutic strategies and the need for more research and innovative approaches in managing these comorbid conditions.
Collapse
Affiliation(s)
- Natalia Weare-Regales
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, University of South Florida, Morsani College of Medicine, and the Division of Endocrinology, Department of Internal Medicine, James A. Haley Veterans Administration, Tampa.
| | - Tara Carr
- Asthma and Airway Disease Research Center, University of Arizona, and the Section of Allergy and Immunology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Fernando Holguin
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Medical School, Aurora
| | - Christopher Andrew Tibbitt
- Department of Medicine Huddinge, Centre for Infectious Medicine, Karolinska Institutet, and the Clinical Lung and Allergy Research Medical Unit for Lung and Allergy Diseases, Karolinska University Hospital, Stockholm
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida, Morsani College of Medicine, Tampa
| |
Collapse
|
22
|
Li Y, Deng Z, Wen J, Ou C, Cen X, Liao Y, Zhang Q, Xie J. Efficacy of dupilumab and risk factors for dupilumab-induced hypereosinophilia in severe asthma: a preliminary study from China. Ann Med 2024; 56:2311843. [PMID: 38316016 PMCID: PMC10846423 DOI: 10.1080/07853890.2024.2311843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVE Dupilumab has been approved for the treatment of severe asthma with type 2 inflammation by inhibiting interleukin (IL)-4 and IL-13 signaling. However, dupilumab-induced hypereosinophilia (HE) has been reported and should not be ignored. The aim of this study was to investigate the efficacy of dupilumab in Chinese patients with severe asthma, whether HE affects its efficacy, and the possible risk factors for HE. METHODS 20 patients with severe asthma who received dupilumab treatment for at least 12 months in the First Affiliated Hospital of Guangzhou Medical University from 2019 to 2022 were included. We compared clinical data and laboratory tests results before dupilumab treatment and at 4 and 12 months after treatment. Based on whether dupilumab treatment triggers HE defined as blood eosinophil count (BEC) ≥ 1.5 × 109 cells/L, the patients were allocated into non-HE and HE groups. RESULTS The patients showed a significant increase in asthma control test (ACT) scores, a decrease in the number of exacerbations, a decrease in the proportion of patients taking an oral corticosteroid (OCS) and in the dose, and a significant improvement in the pulmonary function parameters FEV1/FVC (%) and FEV1 (% predicted) after 4 and 12 months of treatment with dupilumab. For type 2 inflammatory biomarkers, the levels of fractional concentration of exhaled nitric oxide (FeNO), sputum eosinophil count percentage (SEC%) and total immunoglobulin E (TIgE) decreased significantly, whereas BEC were higher after 4 months of treatment, but returned to baseline levels after 12 months. 8 patients (40%) developed asymptomatic HE after dupilumab, and the efficacy was not significantly different between the HE and non-HE groups. The earliest BEC elevation appeared at 1 month after treatment, but most of them declined after 6 months, and basically returned to the baseline level around 12 months of treatment. In addition, we further found that when patients had FeNO ≥ 60 ppb, food allergens positive and combined eosinophilic otitis media (EOM), their BEC increased significantly more than that of the control group after 4 months as well as 12 months of treatment. CONCLUSIONS This study demonstrated that dupilumab was efficacious in Chinese patients with severe asthma, and some patients developed asymptomatic, self-limited HE, which did not affect its efficacy. Additionally, FeNO ≥60 ppb, food allergens positive, and co-morbidities with EOM may be the risk factors for developing HE.
Collapse
Affiliation(s)
- You Li
- Department of pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, Shenzhen Guangming District People’s Hospital, Shenzhen, China
| | - Zhenan Deng
- Department of pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junjie Wen
- Department of pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changxing Ou
- Department of pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Cen
- Department of pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongkang Liao
- Department of pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingling Zhang
- Department of pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiaxing Xie
- Department of pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
23
|
Rodrigues e-Lacerda R, Barra NG, Fang H, Anhê GF, Schertzer JD. NOD2 protects against allergic lung inflammation in obese female mice. iScience 2024; 27:111130. [PMID: 39507249 PMCID: PMC11539594 DOI: 10.1016/j.isci.2024.111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 08/07/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
Obesity is associated with compartmentalized changes in immune responses that can be protective or pathogenic. It has been proposed that obesity-related changes in the microbiota influence allergic lung inflammation. We hypothesized that sensors of the bacterial cell wall influenced allergenic lung inflammation during obesity. Ovalbumin (OVA)-induced lung inflammation was similar in female Nod1-/- and wild-type mice during high-fat-diet-induced obesity, but allergic lung inflammation was higher in obese, high-fat-diet-fed female Nod2-/- mice. Obese Nod2-/- mice had higher inflammatory cell infiltration in the bronchial alveolar lavage (BAL) and lungs, pulmonary fibrosis, mucus levels, hypertrophy and hyperplasia of goblet cells, M2 alveolar macrophage infiltration, interleukin-4 (IL-4), IL-5, IL-6, and lower CXCL1 and IL-22. Therefore, Nod2 protects against excessive lung inflammation and is a bacterial sensor that relays protective responses to allergenic lung inflammation in obese female mice.
Collapse
Affiliation(s)
- Rodrigo Rodrigues e-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- Department of Translational Medicine, University of Campinas, Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, SP CEP 13083-887, Brazil
| | - Nicole G. Barra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| | - Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| | - Gabriel Forato Anhê
- Department of Translational Medicine, University of Campinas, Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, SP CEP 13083-887, Brazil
| | - Jonathan D. Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| |
Collapse
|
24
|
Sharma Y, Bala K. Multifarious Aspect of Cytokines as an Immuno-Therapeutic for Various Diseases. J Interferon Cytokine Res 2024; 44:477-485. [PMID: 39394036 DOI: 10.1089/jir.2024.0090] [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] [Indexed: 10/13/2024] Open
Abstract
Cytokines are known to be a group of growing small proteins that are majorly responsible for the transmission of signals and communication between hematopoietic cells, the cells of the human immune system, and other types of cells. Cytokines play a dominant role in different types of disorders and in perpetuating the inflammation-related disorders. The production of cytokines is a natural process inside the body of a human being against any foreign invasion or due to some pathogenic state to maintain the homeostasis. Cytokines respond in two ways; in some cases, the production and development of cytokines as a therapeutic discovery or intervention will enhance the treatment process and support the reaction given by the body against any pathogenic activity, and in some cases, overproduction of these cytokines responds in the opposite way and behaves as antagonists toward a typical therapeutic drug and its treatment. Overall, 41 articles were reviewed, and it was found that cytokines have proved to be a therapeutic approach among various diseases and can be utilized as a good candidate or a better choice for cancer therapeutics in future development.
Collapse
Affiliation(s)
- Yash Sharma
- Department of Biotechnology, IILM University, Greater Noida, India
| | - Kumud Bala
- Department of Biotechnology, IILM University, Greater Noida, India
- Therapeutics and Molecular Diagnostic Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India
| |
Collapse
|
25
|
Desai B, Adrish M, Mohan A, Lugogo NL. Biologics in Asthma: Emerging Biologics. Immunol Allergy Clin North Am 2024; 44:751-763. [PMID: 39389722 DOI: 10.1016/j.iac.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Advances in our understanding of asthma pathophysiology have led to the advent of multiple targeted asthma therapies such as biologics. However, partial response to biologics occurs, indicating residual disease activity in some patients. Hence, there exists a need for new therapies that focus on novel pathways, alongside perhaps evaluation of combination biologic therapies and modulators of downstream cytokine activation. Therefore, although our current focus is on biologics; it is critical to take a more holistic approach including consideration for nonbiologic therapies that have the potential to significantly advance asthma care.
Collapse
Affiliation(s)
- Brinda Desai
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Muhammad Adrish
- Department of Pulmonary & Critical Care, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Arjun Mohan
- Department of Medicine, University of Michigan, 300 North Ingalls Street, Suite 2d21, Ann Arbor, MI 48109, USA
| | - Njira L Lugogo
- Department of Medicine, University of Michigan, 300 North Ingalls Street, Suite 2c40, Ann Arbor, MI 48109, USA.
| |
Collapse
|
26
|
Kawahara E, Senpuku K, Kawaguchi Y, Yamamoto S, Yasuda K, Kuroda E, Ouji-Sageshima N, Ito T, Hirai T, Shibata T, Yoshioka Y. Recombinant RSV G protein vaccine induces enhanced respiratory disease via IL-13 and mucin overproduction. NPJ Vaccines 2024; 9:187. [PMID: 39394212 PMCID: PMC11470036 DOI: 10.1038/s41541-024-00987-w] [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: 03/17/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
The G protein expressed on the surface of respiratory syncytial virus (RSV) is important for adhesion to host cells and as a vaccine target antigen. The corresponding vaccines can effectively eliminate RSV. However, they exacerbate pulmonary immunopathology including eosinophilic infiltration in the lungs after an RSV challenge in animal models, raising concerns about enhanced respiratory disease (ERD); thus, approaches that mitigate these effects are urgently needed. Herein, we aimed to examine the mechanisms of G protein vaccine-induced ERD in mice, using recombinant G protein as a vaccine antigen. After the RSV challenge, G protein-vaccinated mice exhibited lung weight gain, lung tissue damage, and increased infiltration of eosinophils, neutrophils, and CD4+ T cells into the lungs. We set lung weight gain as the endpoint for ERD and examined the impact of each infiltrating cell on lung weight gain. We observed that CD4+ T cells, but not eosinophils or neutrophils, that infiltrate the lungs are responsible for lung weight gain. In addition, T helper 2 cell-mediated IL-13 induced mucin hypersecretion and lung weight gain. Mucin hypersecretion may contribute to weight gain in the lungs. In conclusion, our results indicate a novel mechanism of G protein vaccine-induced ERD via IL-13 and mucin hypersecretion, which could lead to the development of safe G protein vaccines and the elucidation of the causes of ERD associated with other vaccines.
Collapse
Affiliation(s)
- Eigo Kawahara
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kota Senpuku
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoshino Kawaguchi
- Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shinya Yamamoto
- The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Koubun Yasuda
- Department of Immunology, Hyogo College of Medicine, Hyogo, Japan
| | - Etsushi Kuroda
- Department of Immunology, Hyogo College of Medicine, Hyogo, Japan
| | | | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Nara, Japan
| | - Toshiro Hirai
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Takehiko Shibata
- Department of Microbiology, Tokyo Medical University, Tokyo, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
- Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan.
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan.
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan.
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
| |
Collapse
|
27
|
Zhu M, Ma L, Zhong P, Huang J, Gai J, Li G, Li Y, Qiao P, Gu H, Li X, Yin Y, Zhang L, Deng Z, Sun B, Chen Z, Ding Y, Wan Y. A novel inhalable nanobody targeting IL-4Rα for the treatment of asthma. J Allergy Clin Immunol 2024; 154:1008-1021. [PMID: 38871183 DOI: 10.1016/j.jaci.2024.05.027] [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: 11/20/2023] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Inhalable biologics represent a promising approach to improve the efficacy and safety of asthma treatment. Although several mAbs targeting IL-4 receptor α chain (IL-4Rα) have been approved or are undergoing clinical trials, the development of inhalable mAbs targeting IL-4Rα presents significant challenges. OBJECTIVE Capitalizing on the distinctive advantages of nanobodies (Nbs) in maintaining efficacy during storage and administration, we sought to develop a novel inhalable IL-4Rα Nb for effectively treating asthma. METHODS Three IL-4Rα immunized Nb libraries were used to generate specific and functional IL-4Rα Nbs. LQ036, a bivalent Nb comprising 2 HuNb103 units, was constructed with a high affinity and specificity for human IL-4Rα. The efficacy, pharmacokinetics, and safety of inhaled LQ036 were evaluated in B-hIL4/hIL4RA humanized mice. RESULTS LQ036 inhibited secreted embryonic alkaline phosphatase reporter activity, inhibited TF-1 cell proliferation, and suppressed phosphorylated signal transducer and activator of transduction 6 in T cells from patients with asthma. Crystal structure analysis revealed a binding region similar to dupilumab but with higher affinity, leading to better efficacy in blocking the signaling pathway. HuNb103 competed with IL-4 and IL-13 for IL-4Rα binding. Additionally, LQ036 significantly inhibited ovalbumin-specific IgE levels in serum, CCL17 levels in bronchoalveolar lavage fluid, bronchial mucous cell hyperplasia, and airway goblet cell hyperplasia in B-hIL4/hIL4RA humanized mice. Inhaled LQ036 exhibited favorable pharmacokinetics, safety, and tissue distribution, with higher concentrations observed in the lungs and bronchi. CONCLUSIONS These findings from preclinical studies establish the safety and efficacy of inhaled LQ036, underscoring its potential as a pioneering inhalable biologic therapy for asthma.
Collapse
Affiliation(s)
- Min Zhu
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Linlin Ma
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Peiyu Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Huang
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Junwei Gai
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Guanghui Li
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Yanfei Li
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Peng Qiao
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Huaiyu Gu
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Xiaofei Li
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Yong Yin
- Department of Respiratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Children's Medical Center Pediatric Medical Complex (Pudong), Shanghai, China; Pediatric AI Clinical Application and Research Center, Shanghai Children's Medical Center, Shanghai, China
| | - Lei Zhang
- Department of Respiratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenzhen Deng
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Baihe Sun
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine of Zhongshan Hospital, Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China.
| | - Yu Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| | - Yakun Wan
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China.
| |
Collapse
|
28
|
Jung JH, Kang SA, Park JH, Kim SD, Yu HS, Mun SJ, Cho KS. Immunomodulatory Effect of Adipose Stem Cell-Derived Extra-Cellular Vesicles on Cytokine Expression and Regulatory T Cells in Patients with Asthma. Int J Mol Sci 2024; 25:10524. [PMID: 39408853 PMCID: PMC11477288 DOI: 10.3390/ijms251910524] [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: 08/16/2024] [Revised: 09/18/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Although mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) are as effective as MSCs in the suppression of allergic airway inflammation, few studies have evaluated the immunomodulatory capacity of MSC-derived EVs in patients with asthma. Thus, we assessed the effects of adipose stem cell (ASC)-derived EVs on cytokine expression and regulatory T cells (Tregs) in peripheral blood mononuclear cells (PBMCs) of asthmatic patients. PBMCs (1 × 106 cells/mL) were isolated from asthmatic patient and healthy controls and co-cultured with 1 μg/mL of ASC-derived EVs. Th (T helper) 1-, Th2-, and Treg-related cytokine expression, fluorescence-activated cell sorting analysis of CD4+CD25+FOXP3+ T cells, and co-stimulatory molecules were analyzed before and after ASC-derived EV treatment. The expression levels of IL-4 and costimulatory molecules such as CD83 and CD86 were significantly higher in PBMCs of asthmatic patients than in control PBMCs. However, ASC-derived EV treatment significantly decreased the levels of interleukin (IL)-4 and co-stimulatory molecules such as CD83 and CD86 in the phytohemagglutinin (PHA)-stimulated PBMC of asthmatic patients. Furthermore, ASC-derived EVs remarkably increased the transforming growth factor-β (TGF-β) levels and expression of Tregs in the PBMC of asthmatic patients. ASC-derived EVs induce Treg expansion and have immunomodulatory effects by downregulating IL-4 and upregulating TGF-β in PBMCs of asthmatic patients.
Collapse
Affiliation(s)
- Jae Hoon Jung
- Department of Otorhinolaryngology, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Shin Ae Kang
- Department of Environmental Medical Biology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea;
| | - Ji-Hwan Park
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| | - Sung-Dong Kim
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| | - Hak Sun Yu
- Department of Parasitology and Tropical medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Sue Jean Mun
- Department of Otorhinolaryngology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea;
| | - Kyu-Sup Cho
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea; (J.-H.P.); (S.-D.K.)
| |
Collapse
|
29
|
Gohal G, Moni SS, Bakkari MA, Elmobark ME. A Review on Asthma and Allergy: Current Understanding on Molecular Perspectives. J Clin Med 2024; 13:5775. [PMID: 39407835 PMCID: PMC11476424 DOI: 10.3390/jcm13195775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Asthma, a complex disease characterized by persistent airway inflammation, remains an urgent global health concern. We explored the critical role of allergic biomarkers and dysregulated immune system in asthma through an extensive literature review in databases such as Web of Science, PubMed, EMBASE, Scopus, and Google Scholar. This review summarizes the growing data on the pivotal role of allergic biomarkers and dysregulated immune system in the development and evolution of asthma. Recent studies have uncovered several biomarkers that elucidate intrinsic allergic mechanisms in individuals with asthma. This article highlights these biomarkers' potential in predicting asthma onset, assessing its intensity, guiding therapeutic interventions, and tracking disease progression. We also explore the innovative therapeutic prospects arising from the convergence of allergy and dysregulated immune system in asthma and emphasize the potential for precision medicine approaches. Understanding allergic biomarkers intertwined with a dysregulated immune system heralds a new era in asthma treatment and points to improved and individualized treatment modalities.
Collapse
Affiliation(s)
- Gassem Gohal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Sivakumar S. Moni
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
| | - Mohammed Ali Bakkari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
30
|
Riccardi E, Guida G, Garino S, Bertolini F, Carriero V, Brusamento M, Pizzimenti S, Giannoccaro F, Falzone E, Arrigo E, Levra S, Ricciardolo FLM. Biologics in T2 Severe Asthma: Unveiling Different Effectiveness by Real-World Indirect Comparison. J Clin Med 2024; 13:4750. [PMID: 39200892 PMCID: PMC11355661 DOI: 10.3390/jcm13164750] [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: 07/07/2024] [Revised: 08/02/2024] [Accepted: 08/11/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Indirect comparison among biologics in severe asthma (SA) is a challenging but desirable goal for clinicians in real life. The aim of the study is to define characteristics of a biologic-treated T2-driven-SA population and to evaluate the effectiveness of biologic treatments in a real-world setting by variation in intra/inter-biologic parameters in an up to 4-year follow-up. Methods: Demographic, clinical, functional, and biological characteristics were evaluated retrospectively in 104 patients recruited until July 2022 at baseline (T0) and over a maximum of 4 years (T4) of biologic therapy (omalizumab/OmaG = 41, from T0 to T4, mepolizumab/MepoG = 26, from T0 to T4, benralizumab/BenraG = 18, from T0 to T2, and dupilumab/DupiG = 19, from T0 to T1). Variations of parameters using means of paired Delta were assessed. Results: At baseline, patients had high prevalence of T2-driven comorbidities, low asthma control test (ACT mean 17.65 ± 4.41), impaired pulmonary function (FEV1 65 ± 18 %pred), frequent exacerbations/year (AEs 3.5 ± 3), and OCS dependence (60%). DupiG had lower T2 biomarkers/comorbidities and AEs, and worse FEV1 (57 ± 19 %pred) compared to other biologics (p < 0.05). All biologics improved ACT, FEV1%, FVC%, AEs rate, and OCS use. FEV1% improved in MepoG and BenraG over the minimal clinically important difference and was sustained over 4 years in OmaG and MepoG. A significant RV reduction in OmaG (T4) and DupiG (T1), and BenraG normalization (T2) of airflow limitation were found. We observed through inter-biologic parameters pair delta variation comparison a significant nocturnal awakenings reduction in BenraG vs. OmaG/MepoG, and neutrophils reduction in BenraG/DupiG vs. OmaG. Conclusions: Indirect comparison among biologics unveils clinical and functional improvements that may mark a different effectiveness. These results may highlight the preference of a single biologic compared to another with regard to specific treatable traits.
Collapse
Affiliation(s)
- Elisa Riccardi
- Regional Hospital Parini, Pulmonology Unit, Aosta, 11100 Aosta, Italy;
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
| | - Giuseppe Guida
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
- Severe Asthma, Rare Lung Disease and Respiratory Pathophysiology, San Luigi Gonzaga University Hospital, Orbassano, 10043 Turin, Italy; (S.P.); (F.G.)
| | - Sonia Garino
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
| | - Francesca Bertolini
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
| | - Vitina Carriero
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
| | | | - Stefano Pizzimenti
- Severe Asthma, Rare Lung Disease and Respiratory Pathophysiology, San Luigi Gonzaga University Hospital, Orbassano, 10043 Turin, Italy; (S.P.); (F.G.)
| | - Fabiana Giannoccaro
- Severe Asthma, Rare Lung Disease and Respiratory Pathophysiology, San Luigi Gonzaga University Hospital, Orbassano, 10043 Turin, Italy; (S.P.); (F.G.)
| | - Erica Falzone
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
| | - Elisa Arrigo
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
| | - Stefano Levra
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
| | - Fabio Luigi Massimo Ricciardolo
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Turin, Italy; (S.G.); (F.B.); (V.C.); (E.F.); (E.A.); (S.L.); (F.L.M.R.)
- Severe Asthma, Rare Lung Disease and Respiratory Pathophysiology, San Luigi Gonzaga University Hospital, Orbassano, 10043 Turin, Italy; (S.P.); (F.G.)
- Institute of Translational Pharmacology, National Research Council (IFT-CNR), Section of Palermo, 90146 Palermo, Italy
| |
Collapse
|
31
|
Pelaia C, Zannoni E, Paoletti G, Marzio V, Heffler E, Carrón-Herrero A. Clinical remission in severe asthma: lights and shadows on an ambitious goal. Curr Opin Allergy Clin Immunol 2024; 24:230-236. [PMID: 38713864 DOI: 10.1097/aci.0000000000000991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2024]
Abstract
PURPOSE OF REVIEW The aim of this study was Describe the latest evidence related to the concept of clinical remission in patients with severe asthma, focusing on the lights and shadows of this concept. RECENT FINDINGS The idea of clinical remission in severe asthma patients brings about a significant shift in the way asthma is treated and managed. Although there has yet to be unanimous agreement among various scientific societies on the precise definition, this concept can be extremely useful in advancing the treatment of the disease. SUMMARY Asthma is a common respiratory condition that affects more than 300 million people globally. It has variable symptoms and severity levels, with about 10% of patients experiencing severe asthma. While there have been advancements in treatment, severe asthma poses significant challenges. Recent approaches have focused on achieving clinical remission, which goes beyond symptom control to address underlying inflammation and biological processes. Clinical remission criteria include the absence of symptoms, reduced medication usage, and normalized inflammatory markers. Various biologic therapies show promise, with some patients achieving remission. However, remission's definition varies globally, hindering standardization and a valid comparison. Standardizing remission criteria and refining predictive factors are crucial for effective asthma management. Overall, achieving clinical remission offers hope for improved long-term outcomes in severe asthma patients.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Medical and Surgical Sciences, "Magna Graecia" University of Catanzaro, Catanzaro
| | - Eleonora Zannoni
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Milan
| | - Giovanni Paoletti
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Milan
- Department of Biomedical Sciences, Humanitas University, Milan
| | - Valentina Marzio
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Milan
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Enrico Heffler
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Milan
- Department of Biomedical Sciences, Humanitas University, Milan
| | | |
Collapse
|
32
|
Goleij P, Rahimi M, Pourshahroudi M, Tabari MAK, Muhammad S, Suteja RC, Daglia M, Majma Sanaye P, Hadipour M, Khan H, Sadeghi P. The role of IL-2 cytokine family in asthma. Cytokine 2024; 180:156638. [PMID: 38761716 DOI: 10.1016/j.cyto.2024.156638] [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: 03/09/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND The interleukin-2 (IL-2) family of cytokines, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, are pivotal regulators of the immune response, impacting both innate and adaptive immunity. Understanding their molecular characteristics, receptor interactions, and signalling pathways is essential for elucidating their roles in health and disease. OBJECTIVES This review provides a comprehensive overview of the IL-2 family of cytokines, highlighting their molecular biology, receptor interactions, and signalling mechanisms. Furthermore, it explores the involvement of IL-2 family cytokines in the pathogenesis of chronic respiratory diseases, with a specific focus on chronic obstructive pulmonary disease (COPD) and asthma. METHODS A thorough literature review was conducted to gather insights into the molecular biology, receptor interactions, and signalling pathways of IL-2 family cytokines. Additionally, studies investigating the roles of these cytokines in chronic respiratory diseases, particularly COPD and asthma, were analysed to discern their implications in wider pathophysiology of disease. RESULTS IL-2 family cytokines exert pleiotropic effects on immune cells, modulating cellular proliferation, differentiation, and survival. Dysregulation of IL-2 family cytokines has been implicated in the pathogenesis of chronic respiratory illnesses, including COPD and asthma. Elevated levels of IL-2 and IL-9 have been associated with disease severity in COPD, while IL-4 and IL-9 play crucial roles in asthma pathogenesis by promoting airway inflammation and remodelling. CONCLUSION Understanding the intricate roles of IL-2 family cytokines in chronic respiratory diseases provides valuable insights into potential therapeutic targets for these conditions. Targeting specific cytokines or their receptors may offer novel treatment modalities to attenuate disease progression and improve clinical outcomes in patients with COPD and asthma.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Mohammad Rahimi
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran.
| | - Motahareh Pourshahroudi
- Department of Public Health, Faculty of Health, Education and Life Sciences, Birmingham City University, Birmingham, United Kingdom.
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran; Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Syed Muhammad
- Farooqia College of Pharmacy, Mysuru, Karnataka, India.
| | | | - Maria Daglia
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China.
| | | | - Mahboube Hadipour
- Department of Biochemistry, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan.
| | - Parniyan Sadeghi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Samaha MM, El-Desoky MM, Hisham FA. AdipoRon, an adiponectin receptor agonist, modulates AMPK signaling pathway and alleviates ovalbumin-induced airway inflammation in a murine model of asthma. Int Immunopharmacol 2024; 136:112395. [PMID: 38833845 DOI: 10.1016/j.intimp.2024.112395] [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: 03/28/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/06/2024]
Abstract
Asthma is a long-term disease that causes airways swelling and inflammation and in turn airway narrowing. AdipoRonis an orally active synthetic small molecule that acts as a selective agonist at theadiponectin receptor 1 and 2. The aim of the current study is to delineate the protective effect and the potential underlying mechanism ofadipoRon inairway inflammationinduced byovalbumin (OVA) in comparison withdexamethasone. Adult maleSwiss Albino micewere sensitized to OVA on days 0 and 7, then challenged with OVA on days 14, 15 and 16. AdipoRon was administered orally for 6 days starting from the 11th day till the 16th and 1 h prior to OVA in the challenge days. Obtained results from asthmatic control group showed a significant decrease in serum adiponectin concentration, an increase in inflammatory cell counts inthe bronchoalveolar lavage fluid(BALF), CD68 protein expression, inflammatory cytokine concentration and oxidative stress as well. Administration of adipoRon enhanced antioxidant mechanisms limiting oxidative stress by significantly increasing reduced glutathione (GSH) pulmonary content, decreasing serum lactate dehydrogenase (LDH) together with malondialdehyde (MDA) significant reduction in lung tissue. In addition, it modulated the levels of serum immunoglobulin E (IgE), pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-4, IL-13, nuclear factor kappa B (NF-κB) and the anti-inflammatory one IL-10 improving lung inflammation as revealed by histopathological evaluation. Furthermore, lung tissue expression of nuclear factor erythroid 2-related factor (Nrf2) and 5'AMP-activated protein kinase (AMPK) were significantly increased adipoRon. Notably, results of adipoRon received group were comparable to those of dexamethasone group. In conclusion, our study demonstrates that adipoRon can positively modulate adiponectin expression with activation of AMPK pathway and subsequent improvement in inflammatory and oxidative signaling.
Collapse
Affiliation(s)
- Mahmoud M Samaha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Manal M El-Desoky
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Fatma A Hisham
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
34
|
Tajiri T, Suzuki M, Nishiyama H, Ozawa Y, Kurokawa R, Takeda N, Ito K, Fukumitsu K, Kanemitsu Y, Mori Y, Fukuda S, Uemura T, Ohkubo H, Takemura M, Maeno K, Ito Y, Oguri T, Izuhara K, Niimi A. Efficacy of dupilumab for airway hypersecretion and airway wall thickening in patients with moderate-to-severe asthma: A prospective, observational study. Allergol Int 2024; 73:406-415. [PMID: 38472036 DOI: 10.1016/j.alit.2024.02.002] [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: 11/12/2023] [Revised: 01/18/2024] [Accepted: 02/03/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Dupilumab has clinical effects in patients with moderate-to-severe asthma. When considering interleukin (IL)-4 and IL-13 signaling, effects of dupilumab on airway mucus hypersecretion and airway remodeling are expected, but they have been reported in only a few short-term studies. Its efficacy for airway hyperresponsiveness (AHR) remains unknown. We comprehensively assessed the efficacy of dupilumab, especially for subjective and objective measures of airway mucus hypersecretion and airway dimensions in moderate-to-severe asthmatic patients. METHODS In 28 adult patients with moderate-to-severe uncontrolled asthma, the comprehensive efficacy of 48-week dupilumab treatment, including the Cough and Sputum Assessment Questionnaire (CASA-Q), radiological mucus scores and airway dimensions on computed tomography (CT), was assessed prospectively. Treatment responsiveness to dupilumab was analyzed. RESULTS With 48-week dupilumab treatment, all four cough and sputum domain scores of CASA-Q improved significantly. Radiological mucus scores and airway wall thickening on CT were significantly decreased. The decreases in mucus scores were significantly associated with improvements in Asthma Control Questionnaire scores, Asthma Quality of Life Questionnaire (AQLQ) overall scores, airway obstruction, and airway type 2 inflammation. When defined by > 0.5 improvement in AQLQ overall scores, 18 patients (64%) were identified as responders. CONCLUSIONS Dupilumab reversed subjective and objective measures of airway mucus hypersecretion and some aspects of airway remodeling in patients with moderate-to-severe uncontrolled asthma.
Collapse
Affiliation(s)
- Tomoko Tajiri
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan.
| | - Motohiko Suzuki
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan; Department of Otorhinolaryngology, Nagoya City University Midori Municipal Hospital, Aichi, Japan
| | - Hirono Nishiyama
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Yoshiyuki Ozawa
- Department of Radiology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Ryota Kurokawa
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Norihisa Takeda
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Keima Ito
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Kensuke Fukumitsu
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Yoshihiro Kanemitsu
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Yuta Mori
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Satoshi Fukuda
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Takehiro Uemura
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Hirotsugu Ohkubo
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Masaya Takemura
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Ken Maeno
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Yutaka Ito
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Tetsuya Oguri
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Akio Niimi
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Aichi, Japan
| |
Collapse
|
35
|
Pelaia C, Melhorn J, Hinks TS, Couillard S, Vatrella A, Pelaia G, Pavord ID. Type 2 severe asthma: pathophysiology and treatment with biologics. Expert Rev Respir Med 2024; 18:485-498. [PMID: 38994712 DOI: 10.1080/17476348.2024.2380072] [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: 05/05/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION The hallmark of most patients with severe asthma is type 2 inflammation, driven by innate and adaptive immune responses leading to either allergic or non-allergic eosinophilic infiltration of airways. The cellular and molecular pathways underlying severe type 2 asthma can be successfully targeted by specific monoclonal antibodies. AREAS COVERED This review article provides a concise overview of the pathophysiology of type 2 asthma, followed by an updated appraisal of the mechanisms of action and therapeutic efficacy of currently available biologic treatments used for management of severe type 2 asthma. Therefore, all reported information arises from a wide literature search performed on PubMed. EXPERT OPINION The main result of the recent advances in the field of anti-asthma biologic therapies is the implementation of a personalized medicine approach, aimed to achieve clinical remission of severe asthma. Today this accomplishment is made possible by the right choice of the most beneficial biologic drug for the pathologic traits characterizing each patient, including type 2 severe asthma and its comorbidities.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Calabria, Italy
| | - James Melhorn
- Nuffield Department of Medicine, Respiratory Medicine Unit, University of Oxford, Oxford, UK
| | - Timothy Sc Hinks
- Nuffield Department of Medicine, Respiratory Medicine Unit, University of Oxford, Oxford, UK
| | - Simon Couillard
- Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Ian D Pavord
- Nuffield Department of Medicine, Respiratory Medicine Unit, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Pilkington AW, Buragamadagu B, Johnston RA. Weighted Breaths: Exploring Biologic and Non-Biologic Therapies for Co-Existing Asthma and Obesity. Curr Allergy Asthma Rep 2024; 24:381-393. [PMID: 38878250 PMCID: PMC11233394 DOI: 10.1007/s11882-024-01153-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 07/10/2024]
Abstract
PURPOSE OF REVIEW To discuss the effectiveness of biologics, some of which comprise the newest class of asthma controller medications, and non-biologics in the treatment of asthma co-existing with obesity. RECENT FINDINGS Our review of recent preliminary and published data from clinical trials revealed that obese asthmatics respond favorably to dupilumab, mepolizumab, omalizumab, and tezepelumab, which are biologics currently indicated as add-on maintenance therapy for severe asthma. Furthermore, clinical trials are ongoing to assess the efficacy of non-biologics in the treatment of obese asthma, including a glucagon-like peptide-1 receptor agonist, a Janus kinase inhibitor, and probiotics. Although many biologics presently indicated as add-on maintenance therapy for severe asthma exhibit efficacy in obese asthmatics, other phenotypes of asthma co-existing with obesity may be refractory to these medications. Thus, to improve quality of life and asthma control, it is imperative to identify therapeutic options for all existing phenotypes of obese asthma.
Collapse
Affiliation(s)
- Albert W Pilkington
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, United States Department of Health and Human Services, 1000 Frederick Lane, Morgantown, WV, 26508-5402, USA
| | - Bhanusowmya Buragamadagu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Richard A Johnston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, United States Department of Health and Human Services, 1000 Frederick Lane, Morgantown, WV, 26508-5402, USA.
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA.
- Department of Physiology, Pharmacology, and Toxicology, School of Medicine, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
37
|
Sévoz-Couche C, Liao W, Foo HYC, Bonne I, Lu TB, Tan Qi Hui C, Azhar SH, Peh WYX, Yen SC, Wong WSF. Direct vagus nerve stimulation: A new tool to control allergic airway inflammation through α7 nicotinic acetylcholine receptor. Br J Pharmacol 2024; 181:1916-1934. [PMID: 38430056 DOI: 10.1111/bph.16334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND AND PURPOSE Asthma is characterized by airway inflammation, mucus hypersecretion, and airway hyperresponsiveness. The use of nicotinic agents to mimic the cholinergic anti-inflammatory pathway (CAP) controls experimental asthma. Yet, the effects of vagus nerve stimulation (VNS)-induced CAP on allergic inflammation remain unknown. EXPERIMENTAL APPROACH BALB/c mice were sensitized and challenged with house dust mite (HDM) extract and treated with active VNS (5 Hz, 0.5 ms, 0.05-1 mA). Bronchoalveolar lavage (BAL) fluid was assessed for total and differential cell counts and cytokine levels. Lungs were examined by histopathology and electron microscopy. KEY RESULTS In the HDM mouse asthma model, VNS at intensities equal to or above 0.1 mA (VNS 0.1) but not sham VNS reduced BAL fluid differential cell counts and alveolar macrophages expressing α7 nicotinic receptors (α7nAChR), goblet cell hyperplasia, and collagen deposition. Besides, VNS 0.1 also abated HDM-induced elevation of type 2 cytokines IL-4 and IL-5 and was found to block the phosphorylation of transcription factor STAT6 and expression level of IRF4 in total lung lysates. Finally, VNS 0.1 abrogated methacholine-induced hyperresponsiveness in asthma mice. Prior administration of α-bungarotoxin, a specific inhibitor of α7nAChR, but not propranolol, a specific inhibitor of β2-adrenoceptors, abolished the therapeutic effects of VNS 0.1. CONCLUSION AND IMPLICATIONS Our data revealed the protective effects of VNS on various clinical features in allergic airway inflammation model. VNS, a clinically approved therapy for depression and epilepsy, appears to be a promising new strategy for controlling allergic asthma.
Collapse
Affiliation(s)
- Caroline Sévoz-Couche
- INSERM, UMRS1158 Neurophysiologie Respiratoire et Clinique, Sorbonne Université, Paris, France
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore
| | - Hazel Y C Foo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore
| | - Isabelle Bonne
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Thong Beng Lu
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Caris Tan Qi Hui
- Advanced imaging and Histology Core, Immunology Program, Life Science Institute, National University of Singapore, Singapore
| | - Syaza Hazwany Azhar
- Advanced imaging and Histology Core, Immunology Program, Life Science Institute, National University of Singapore, Singapore
| | - Wendy Yen Xian Peh
- The N.1 Institute for Health, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
| | - Shih-Cheng Yen
- The N.1 Institute for Health, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
38
|
Palacionyte J, Januskevicius A, Vasyle E, Rimkunas A, Miliauskas S, Malakauskas K. Clinical Remission Criteria and Serum Levels of Type 2 Inflammation Mediators during 24 Weeks of Treatment with the Anti-IL-5 Drug Mepolizumab in Patients with T2-High Severe Asthma. Diagnostics (Basel) 2024; 14:1345. [PMID: 39001236 PMCID: PMC11240777 DOI: 10.3390/diagnostics14131345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
Anti-interleukin (IL) 5 is an effective treatment modality for inhibiting eosinophilic inflammation in patients with T2-high severe asthma. The aim of this study was to determine the clinical efficacy and serum levels of type 2 inflammatory mediators during 24 weeks of mepolizumab treatment in patients with T2-high severe asthma. Eighteen patients with T2-high severe asthma were enrolled in this study. All patients received 100 mg of mepolizumab subcutaneously every 4 weeks and were retested at 4, 12, and 24 weeks. A clinical examination, asthma control test (ACT), and spirometry were performed; fractional exhaled nitric oxide (FeNO) levels were evaluated; and blood samples were drawn at every visit. Type 2 inflammation mediator levels were measured using enzyme-linked immunosorbent assay (ELISA). The blood eosinophil level significantly decreased, the ACT score and FEV1 increased after 4 weeks of mepolizumab treatment with the same tendency after 12 and 24 weeks (p < 0.05), and the FeNO level did not change (p > 0.05). A total of 27.8% of patients reached clinical remission criteria after 24 weeks of mepolizumab treatment. IL-33 and eotaxin significantly increased (p < 0.05) while IL-5, IL-13, thymic stromal lymphopoietin (TSLP), soluble IL-5 receptor subunit alpha (sIL-5Rα), and soluble high-affinity immunoglobulin E receptor (sFcεRI) decreased, with the same tendency after 12 and 24 weeks (p < 0.05). The serum levels of immunoglobulin (Ig) E and IL-4 and IL-25 levels did not change during mepolizumab treatment compared to baseline (p > 0.05). In conclusion, treatment with mepolizumab over 24 weeks improved lung function and asthma control in T2-high severe asthma patients, with nearly one-third achieving clinical remission criteria, and affected the balance of type 2 inflammatory mediators.
Collapse
Affiliation(s)
- Jolita Palacionyte
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (S.M.); (K.M.)
| | - Andrius Januskevicius
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (E.V.); (A.R.)
| | - Egle Vasyle
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (E.V.); (A.R.)
| | - Airidas Rimkunas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (E.V.); (A.R.)
| | - Skaidrius Miliauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (S.M.); (K.M.)
| | - Kestutis Malakauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (S.M.); (K.M.)
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (E.V.); (A.R.)
| |
Collapse
|
39
|
Xu C, Song Y, Liu W, Liu R, Bai Q, Li L, Wang C, Yan G. IL-4 activates ULK1/Atg9a/Rab9 in asthma, NLRP3 inflammasomes, and Golgi fragmentation by increasing autophagy flux and mitochondrial oxidative stress. Redox Biol 2024; 71:103090. [PMID: 38373380 PMCID: PMC10878789 DOI: 10.1016/j.redox.2024.103090] [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: 01/18/2024] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024] Open
Abstract
During asthma, there is an intensification of pulmonary epithelial inflammation, mitochondrial oxidative stress, and Golgi apparatus fragmentation. However, the underlying mechanism remains largely unknown. Therefore, this study investigated the roles of ULK1, Atg9a, and Rab9 in epithelial inflammation, mitochondrial oxidative stress, and Golgi apparatus fragmentation. We found that ULK1 gene knockout reduced the infiltration of inflammatory cells, restored the imbalance of the Th1/Th2 ratio, and inhibited the formation of inflammatory bodies in the lung tissue of house dust mite-induced asthma mice. Moreover, we demonstrated that Atg9a interacted with ULK1 at S467. ULK1 phosphorylated Atg9a at S14. Treatment with ULK1 activator (LYN-1604) and ULK1 inhibitor (ULK-101) respectively promoted and inhibited inflammasome activation, indicating that the activation of inflammasome induced by house dust mite in asthma mice is dependent on ULK1. For validation of the in vivo results, we then used a lentivirus containing ULK1 wild type and ULK1-S467A genes to infect Beas-2b-ULK1-knockout cells and establish a stable cell line. The results suggest that the ULK1 S467 site is crucial for IL-4-induced inflammation and oxidative stress. Experimental verification confirmed that Atg9a was the superior signaling pathway of Rab9. Interestingly, we found for the first time that Rab9 played a very important role in inflammation-induced fragmentation of the Golgi apparatus. Inhibiting the activation of the ULK1/Atg9a/Rab9 signaling pathways can inhibit Golgi apparatus fragmentation and mitochondrial oxidative stress in asthma while reducing the production of NLRP3-mediated pulmonary epithelial inflammation.
Collapse
Affiliation(s)
- Chang Xu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, 133002, PR China
| | - Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, 133002, PR China
| | - Wanting Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, 133002, PR China
| | - Ruobai Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, 133002, PR China
| | - Qiaoyun Bai
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, 133002, PR China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, 133002, PR China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, 133002, PR China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, 133002, PR China.
| |
Collapse
|
40
|
AlBloushi S, Al-Ahmad M. Exploring the immunopathology of type 2 inflammatory airway diseases. Front Immunol 2024; 15:1285598. [PMID: 38680486 PMCID: PMC11045947 DOI: 10.3389/fimmu.2024.1285598] [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: 08/30/2023] [Accepted: 03/22/2024] [Indexed: 05/01/2024] Open
Abstract
Significant advancements have been achieved in understanding the roles of different immune cells, as well as cytokines and chemokines, in the pathogenesis of eosinophilic airway conditions. This review examines the pathogenesis of Chronic Rhinosinusitis with Nasal Polyps (CRSwNP), marked by complex immune dysregulation, with major contributions from type 2 inflammation and dysfunctional airway epithelium. The presence of eosinophils and the role of T-cell subsets, particularly an imbalance between Treg and Th17 cells, are crucial to the disease's pathogenesis. The review also investigates the pathogenesis of eosinophilic asthma, a unique asthma subtype. It is characterized by inflammation and high eosinophil levels, with eosinophils playing a pivotal role in triggering type 2 inflammation. The immune response involves Th2 cells, eosinophils, and IgE, among others, all activated by genetic and environmental factors. The intricate interplay among these elements, chemokines, and innate lymphoid cells results in airway inflammation and hyper-responsiveness, contributing to the pathogenesis of eosinophilic asthma. Another scope of this review is the pathogenesis of Eosinophilic Granulomatosis with Polyangiitis (EGPA); a complex inflammatory disease that commonly affects the respiratory tract and small to medium-sized blood vessels. It is characterized by elevated eosinophil levels in blood and tissues. The pathogenesis involves the activation of adaptive immune responses by antigens leading to T and B cell activation and eosinophil stimulation, which causes tissue and vessel damage. On the other hand, Allergic Bronchopulmonary Aspergillosis (ABPA) is a hypersensitive response that occurs when the airways become colonized by aspergillus fungus, with the pathogenesis involving activation of Th2 immune responses, production of IgE antibodies, and eosinophilic action leading to bronchial inflammation and subsequent lung damage. This analysis scrutinizes how an imbalanced immune system contributes to these eosinophilic diseases. The understanding derived from this assessment can steer researchers toward designing new potential therapeutic targets for efficient control of these disorders.
Collapse
Affiliation(s)
| | - Mona Al-Ahmad
- Al-Rashed Allergy Center, Ministry of Health, Kuwait City, Kuwait
- Microbiology Department, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
41
|
Pham DD, Lee JH, Kwon HS, Song WJ, Cho YS, Kim H, Kwon JW, Park SY, Kim S, Hur GY, Kim BK, Nam YH, Yang MS, Kim MY, Kim SH, Lee BJ, Lee T, Park SY, Kim MH, Cho YJ, Park C, Jung JW, Park HK, Kim JH, Moon JY, Adcock I, Bhavsar P, Chung KF, Kim TB. Prospective direct comparison of biologic treatments for severe eosinophilic asthma: Findings from the PRISM study. Ann Allergy Asthma Immunol 2024; 132:457-462.e2. [PMID: 37977324 DOI: 10.1016/j.anai.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Although various monoclonal antibodies have been used as add-on therapy for severe eosinophilic asthma (SEA), to the best of our knowledge, no direct head-to-head comparative study has evaluated their efficacy. OBJECTIVE To compare the efficacy of reslizumab, mepolizumab, and dupilumab in patients with SEA. METHODS This was a multicenter, prospective observational study in patients with SEA who had received 1 of these biologic agents for at least 6 months. Cox proportional hazard models were used to compare the risk of the first exacerbation event, adjusting for sputum or blood eosinophils and common asthma-related covariates. The annual exacerbation rate was analyzed using a negative binomial model, and a mixed-effect model was used to analyze changes in forced expiratory volume in 1 second and asthma control test score over time. RESULTS A total of 141 patients with SEA were included in the analysis; 71 (50%) received dupilumab; 40 (28%) received reslizumab, and 30 (21%) received mepolizumab. During the 12-month follow-up, 27.5%, 43.3%, and 38.0% of patients in the reslizumab, mepolizumab, and dupilumab groups, respectively, experienced at least 1 exacerbation. However, after adjusting for confounding factors, the dupilumab and mepolizumab groups showed similar outcomes in time-to-first exacerbation, exacerbation rate, forced expiratory volume in 1 second, and asthma control test score to those of the reslizumab group. CONCLUSION In patients with SEA, treatment with reslizumab, mepolizumab, and dupilumab resulted in comparable clinical outcomes within a 12-month period. TRIAL REGISTRATION The cohort protocol was sanctioned by the Institutional Review Board of each study center (clinicaltrial.gov identifier NCT05164939).
Collapse
Affiliation(s)
- Duong Duc Pham
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji-Hyang Lee
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyouk-Soo Kwon
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Woo-Jung Song
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - You Sook Cho
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyunkyoung Kim
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae-Woo Kwon
- Department of Allergy and Clinical Immunology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - So-Young Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Chung-Ang University Gwangmyeong Hospital, Korea
| | - Sujeong Kim
- School of Medicine, Department of Internal Medicine, Kyungpook National University, Daegu, Korea
| | - Gyu Young Hur
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Byung Keun Kim
- Department of Internal Medicine, Korea University Medical Center Anam Hospital, Seoul, Korea
| | - Young-Hee Nam
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Min-Suk Yang
- Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Mi-Yeong Kim
- Department of Internal Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Sae-Hoon Kim
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Byung-Jae Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Taehoon Lee
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - So Young Park
- Department of Internal Medicine, Eulji University School of Medicine, Seoul, Korea
| | - Min-Hye Kim
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Young-Joo Cho
- Department of Allergy and Clinical Immunology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Korea
| | - ChanSun Park
- Department of Internal Medicine, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Jae-Woo Jung
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Han Ki Park
- Department of Allergy and Clinical Immunology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Joo-Hee Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Ji-Yong Moon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Ian Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pankaj Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Tae-Bum Kim
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
42
|
Li N, Shakib S, Qian W, Yao X, Li P, Nip TK, Bai X, Shen K. Safety, pharmacokinetics, and pharmacodynamics of anti-IL-4Rα antibody SHR-1819 in healthy subjects: A randomized, controlled phase I study. Clin Transl Sci 2024; 17:e13763. [PMID: 38545854 PMCID: PMC10974694 DOI: 10.1111/cts.13763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 04/19/2024] Open
Abstract
SHR-1819 is a novel anti-IL-4Rα monoclonal antibody currently under clinical development for use in patients with type 2 inflammatory diseases. In this randomized, double-blind, placebo-controlled, single-dose escalation phase I trial, we evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics of SHR-1819 in healthy subjects. Subjects received a single subcutaneous injection of SHR-1819 or placebo, with dose escalation starting at 60 mg and subsequently increasing to 120, 240, 360, and 720 mg. A total of 42 eligible subjects were randomized, and 33 received SHR-1819 (1 subject in the 60 mg cohort and 8 subjects each in the 120, 240, 360 , and 720 mg cohorts) and 9 received placebo. SHR-1819 was well-tolerated, with the majority of adverse events being mild in severity. The exposure of SHR-1819 increased in a manner greater than proportionally with a dose range of 120 to 720 mg. The median Tmax was within 4-7 days (60-720 mg), and the mean half-life ranged from 2.88 to 5.97 days (120-720 mg). The clearance rate of SHR-1819 exhibited a decrease with increasing dose level. Administration of SHR-1819 resulted in a certain degree of reduction in the percentage change from baseline in concentrations of inflammatory biomarkers TARC/CCL17 and IgE, while the reduction of TARC/CCL17 concentrations showed a dose-dependent trend. More than half of the total subjects treated with SHR-1819 were reported antidrug antibody-negative. The preliminary data from this phase I study support further development of SHR-1819 for the treatment of type 2 inflammatory diseases.
Collapse
Affiliation(s)
- Na Li
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Sepehr Shakib
- CMAX Clinical ResearchAdelaideSouth AustraliaAustralia
- Department of Clinical PharmacologyUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Weilin Qian
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Xiaoyan Yao
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Puyuan Li
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Tsz Keung Nip
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Xiaoyan Bai
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| | - Kai Shen
- Jiangsu Hengrui Pharmaceuticals Co., Ltd.ShanghaiChina
| |
Collapse
|
43
|
Pelaia C, Giacalone A, Ippolito G, Pastore D, Maglio A, Piazzetta GL, Lobello N, Lombardo N, Vatrella A, Pelaia G. Difficult-To-Treat and Severe Asthma: Can Real-World Studies On Effectiveness of Biological Treatments Change the Lives of Patients? Pragmat Obs Res 2024; 15:45-51. [PMID: 38495680 PMCID: PMC10941791 DOI: 10.2147/por.s396799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Many different phenotypes that characterize severe asthma are supported by intricate pathomechanisms called endotypes. The latter are driven by molecular interactions, mediated by intercellular networks. With regard to the biological treatments of either allergic or non-allergic eosinophilic type 2 asthma, real-world studies have confirmed the positive effects of currently available antibodies directed against immunoglobulins E (IgE), interleukin-5 (IL-5) and its receptor, as well as the receptors of interleukins-4 (IL-4) and 13 (IL-13). The best way to treat severe asthma should be chosen based on the peculiar phenotypic and endotypic traits of each patient. This will lead to relevant improvements in both clinical and functional outcomes. In particular, biological therapies can change the lives of asthma patients with a strong impact on quality of life. Unfortunately, patients with severe non-type-2 asthma, who continue to have pertinent unmet needs, are not receiving satisfactory advances within the context of biological treatments. It is also hopeful that in the next future new therapeutic strategies will be specifically implemented for these people, perhaps offering them the opportunity to improve their current, mostly inadequate asthma management.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Antonio Giacalone
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Gianluca Ippolito
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Daniela Pastore
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Angelantonio Maglio
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Giovanna Lucia Piazzetta
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Nadia Lobello
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Nicola Lombardo
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
44
|
Goetzl EJ. Th2 cells in rapid immune responses and protective avoidance reactions. FASEB J 2024; 38:e23485. [PMID: 38372961 DOI: 10.1096/fj.202302584rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/20/2024]
Abstract
Type 2 helper cells (Th2 cells) differentiate from CD4 helper T cells under the influence of IL-4 and conventional or monocyte-derived CD11b+ dendritic cells. Th2 cells are capable of generating IL-4, IL-5, and IL-13, as well as evoking immunoglobulin class-switch to IgE. Three types of rapid immune responses are Th2 cell-dependent: (1) mast cell-IgE mediated allergic reactions, (2) Th2 cell-derived cytokine-mediated reactions that complement allergic reactions and protect the host from toxins, xenobiotics, environmental irritants, and helminthic parasites, and (3) IgE-stimulated mast cell-derived cysteinyl-leukotriene mediated avoidance of toxins. The contributions of Th2 cell-derived cytokines to eosinophilia (IL-5), IgE class-switch, and epithelial barrier activation, mucous secretion, and metaplasia (IL-4 and IL-13) in asthma, allergic rhinitis with polyps and atopic dermatitis have led to anti-cytokine monoclonal antibody treatments. Anti-IL-5 neutralizing monoclonal antibody in asthma and anti-IL-4/IL-13 receptor neutralizing monoclonal antibody in asthma and atopic dermatitis are proven successful therapies in appropriately selected patients who are not sufficiently improved by conventional treatments.
Collapse
Affiliation(s)
- Edward J Goetzl
- Research Department, Geriatric Research Center, University of California Medical Center, San Francisco, San Francisco, California, USA
| |
Collapse
|
45
|
Moon C, Sahakijpijarn S, Maier EY, Taft DR, Jara MO, Praphawatvet T, Manandhar R, Shetty N, Lubach J, Narang A, Nagapudi K, Williams RO. Inhaled JAK Inhibitor GDC-0214 Nanoaggregate Powder Exhibits Improved Pharmacokinetic Profile in Rats Compared to the Micronized Form: Benefits of Thin Film Freezing. Mol Pharm 2024; 21:564-580. [PMID: 38215042 DOI: 10.1021/acs.molpharmaceut.3c00719] [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] [Indexed: 01/14/2024]
Abstract
Asthma is a common chronic disease affecting the airways in the lungs. The receptors of allergic cytokines, including interleukin (IL)-4, IL-5, and IL-13, trigger the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, which involves the pathogenesis of asthma. GDC-0214 is a JAK inhibitor that was developed as a potent and selective target for the treatment of asthma, specifically targeting the lungs. While inhaled GDC-0214 is a promising novel treatment option against asthma, improvement is still needed to achieve increased potency of the powder formulation and a reduced number of capsules containing powder to be inhaled. In this study, high-potency amorphous powder formulations containing GDC-0214 nanoaggregates for dry powder inhalation were developed using particle engineering technology, thin film freezing (TFF). A high dose per capsule was successfully achieved by enhancing the solubility of GDC-0214 and powder conditioning. Lactose and/or leucine as excipients exhibited optimum stability and aerosolization of GDC-0214 nanoaggregates, and aerosolization of the dose was independent of air flow through the device between 2 and 6 kPa pressure drops. In the rat PK study, formulation F20, which contains 80% GDC-0214 and 20% lactose, resulted in the highest AUC0-24h in the lungs with the lowest AUC0-24h in the plasma that corresponds to a 4.8-fold higher ratio of the lung-to-plasma exposures compared to micronized crystalline GDC-0214 powder administered by dry powder inhalation. Therefore, GDC-0214 nanoaggregates produced by TFF provided an improved dry powder for inhalation that can lead to enhanced therapeutic efficacy with a lower risk of systemic toxicity.
Collapse
Affiliation(s)
- Chaeho Moon
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Sawittree Sahakijpijarn
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
- TFF Pharmaceuticals, Inc., Austin, Texas 78753, United States
| | - Esther Y Maier
- Drug Dynamics Institute, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78723, United States
| | - David R Taft
- Division of Pharmaceutical Sciences, Long Island University, Brooklyn, New York 11201, United States
| | - Miguel O Jara
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Tuangrat Praphawatvet
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | | | - Nivedita Shetty
- Genentech, Inc., South San Francisco, California 94080, United States
| | - Joseph Lubach
- Genentech, Inc., South San Francisco, California 94080, United States
| | - Ajit Narang
- Genentech, Inc., South San Francisco, California 94080, United States
| | - Karthik Nagapudi
- Genentech, Inc., South San Francisco, California 94080, United States
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
46
|
Arentoft NS, Fialla AD, Krohn PS, Patursson MT, Thudium RF, Suarez-Zdunek MA, Høgh J, Lauridsen EHE, Hansen JB, Jensen JUS, Perch M, Møller DL, Pommergaard HC, Aagaard NK, Davidsen JR, Lange P, Çolak Y, Afzal S, Nordestgaard BG, Rasmussen A, Nielsen SD. Fraction of exhaled nitric oxide is higher in liver transplant recipients than in controls from the general population: a cohort study. Front Immunol 2024; 15:1330923. [PMID: 38361926 PMCID: PMC10867152 DOI: 10.3389/fimmu.2024.1330923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/02/2024] [Indexed: 02/17/2024] Open
Abstract
Background Fraction of exhaled nitric oxide with an expiratory flow of 50 mL/s (FENO50) is a biomarker of eosinophilic airway inflammation. Liver transplant recipients have an increased risk of pulmonary infections, but little is known about the burden of chronic pulmonary diseases in this group. We aimed to assess the prevalence of elevated FENO50 in liver transplant recipients and compare it to controls from the general population. Methods FENO50 was measured in 271 liver transplant recipients from The Danish Comorbidity in Liver Transplant Recipients (DACOLT) study and 1,018 age- and sex-matched controls from The Copenhagen General Population Study (CGPS). Elevated FENO50 was defined as ≥25 or ≥50 parts per billion (ppb). The analyses were adjusted for known and suspected confounders. Results The median age of the liver transplant recipients was 55 years (interquartile range (IQR) 46-64), and 58% were men. The liver transplant recipients had a higher median FENO50 than the controls [16 ppb (IQR 10-26) vs. 13 ppb (IQR 8-18.), p < 0.001]. Furthermore, the liver transplant recipients had a higher prevalence of elevated FENO50 (for FENO50 ≥25 ppb 27% vs. 11%, p < 0.001 and ≥50 ppb 4% vs. 2%, p = 0.02). The results were similar after adjusting for age, sex, smoking status, use of airway medication, and blood eosinophil counts [the adjusted odds ratio (OR) for FENO50 ≥25 ppb was 3.58 (95% CI: 2.50-5.15, p < 0.0001) and the adjusted OR for FENO50 ≥50 ppb was 3.14 (95% CI: 1.37-7.20, p = 0.007)]. Conclusion The liver transplant recipients had elevated FENO50, implying increased eosinophilic airway inflammation. The clinical impact of this finding needs further investigation.
Collapse
Affiliation(s)
- Nicoline S. Arentoft
- Department of Infectious Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Annette D. Fialla
- Department of Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Paul S. Krohn
- Department of Surgery and Transplantation, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Magda T. Patursson
- Department of Infectious Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Rebekka F. Thudium
- Department of Infectious Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Moises A. Suarez-Zdunek
- Department of Infectious Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Julie Høgh
- Department of Infectious Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Emilie H. E. Lauridsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper B. Hansen
- Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
| | - Jens-Ulrik S. Jensen
- Department of Respiratory Medicine, Copenhagen University Hospital - Herlev and Gentofte, Gentofte, Denmark
| | - Michael Perch
- Department of Cardiology, Heart and Lung Transplant Unit, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dina L. Møller
- Department of Infectious Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Hans-Christian Pommergaard
- Department of Surgery and Transplantation, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels K. Aagaard
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper R. Davidsen
- South Danish Center for Interstitial Lung Diseases (SCILS), Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark
| | - Peter Lange
- Department of Respiratory Medicine, Copenhagen University Hospital - Herlev and Gentofte, Gentofte, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Yunus Çolak
- Department of Respiratory Medicine, Copenhagen University Hospital - Herlev and Gentofte, Gentofte, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Shoaib Afzal
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Børge G. Nordestgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Allan Rasmussen
- Department of Surgery and Transplantation, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Susanne D. Nielsen
- Department of Infectious Diseases, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Giombi F, Pace GM, Pirola F, Cerasuolo M, Ferreli F, Mercante G, Spriano G, Canonica GW, Heffler E, Ferri S, Puggioni F, Paoletti G, Malvezzi L. Airways Type-2 Related Disorders: Multiorgan, Systemic or Syndemic Disease? Int J Mol Sci 2024; 25:730. [PMID: 38255804 PMCID: PMC10815382 DOI: 10.3390/ijms25020730] [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: 12/14/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Chronic rhinosinusitis (CRS) has recently undergone a significant paradigm shift, moving from a phenotypical classification towards an "endotype-based" definition that places more emphasis on clinical and therapeutic aspects. Similar to other airway diseases, like asthma, most cases of CRS in developed countries exhibit a dysregulated type-2 immune response and related cytokines. Consequently, the traditional distinction between upper and lower airways has been replaced by a "united airway" perspective. Additionally, type-2 related disorders extend beyond respiratory boundaries, encompassing conditions beyond the airways, such as atopic dermatitis. This necessitates a multidisciplinary approach. Moreover, consideration of possible systemic implications is crucial, particularly in relation to sleep-related breathing diseases like Obstructive Sleep Apnoea Syndrome (OSAS) and the alteration of systemic inflammatory mediators such as nitric oxide. The trends in epidemiological, economic, and social burden are progressively increasing worldwide, indicating syndemic characteristics. In light of these insights, this narrative review aims to present the latest evidence on respiratory type-2 related disorders, with a specific focus on CRS while promoting a comprehensive perspective on the "united airways". It also introduces a novel concept: viewing these conditions as a multiorgan, systemic, and syndemic disease.
Collapse
Affiliation(s)
- Francesco Giombi
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Gian Marco Pace
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Francesca Pirola
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
| | - Michele Cerasuolo
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Otorhinolaryngology Head & Neck Surgery Unit, Casa di Cura Humanitas San Pio X, Via Francesco Nava 31, 20159 Milan, Italy
| | - Fabio Ferreli
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Giuseppe Mercante
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Giuseppe Spriano
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Sebastian Ferri
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Francesca Puggioni
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Giovanni Paoletti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Luca Malvezzi
- Otorhinolaryngology Head & Neck Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (F.G.); (F.P.); (M.C.); (F.F.); (G.M.); (G.S.); (L.M.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (G.W.C.); (E.H.); (S.F.); (F.P.); (G.P.)
- Otorhinolaryngology Head & Neck Surgery Unit, Casa di Cura Humanitas San Pio X, Via Francesco Nava 31, 20159 Milan, Italy
| |
Collapse
|
48
|
Ferri S, Montagna C, Casini M, Malvezzi L, Pirola F, Russo E, Racca F, Messina MR, Puggioni F, Nappi E, Costanzo G, Del Moro L, Mercante G, Spriano G, Canonica GW, Paoletti G, Heffler E. Sleep quality burden in chronic rhinosinusitis with nasal polyps and its modulation by dupilumab. Ann Allergy Asthma Immunol 2024; 132:69-75. [PMID: 37652235 DOI: 10.1016/j.anai.2023.08.594] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps (CRSwNP) is associated with a substantial burden on patients' quality of life and impaired sleep quality. The most common CRSwNP endotype is characterized by type 2 inflammation, with enhanced production of interleukin (IL)-4, IL-5, and IL-13. Dupilumab is a monoclonal antibody against IL-4 receptor-α, which inhibits both IL-4 and IL-13 signaling, and was recently approved for treatment of CRSwNP. OBJECTIVE We investigated the effect of dupilumab on the sleep quality of patients with CRSwNP in a real-life setting. METHODS Patients were evaluated at baseline and after 1 and 3 months of dupilumab treatment by means of the Epworth sleepiness scale (ESS), insomnia severity index (ISI), Pittsburgh sleep quality index (PSQI), and sinonasal outcome test 22 (SNOT-22) sleep domain. RESULTS A total of 29 consecutive patients were enrolled, and their baseline sleep quality assessment were as follows: ESS of 7.9 (± 4.5); ISI of 13.1 (± 6.2); PSQI of 9.2 (± 3.7); and SNOT-22 sleep domain of 12.1 (± 4.2). Excessive daily sleepiness, insomnia, and globally impaired sleep quality were present in 24.1%, 79.3%, and 93.1% respectively. Treatment with dupilumab was associated with significant improvement in ESS, ISI, PSQI, and SNOT-22 sleep domain with concomitant reduction of the proportion of patients with insomnia and globally impaired sleep quality. CONCLUSION CRSwNP was associated with a significant impact on global sleep quality, in particular, insomnia, and treatment with dupilumab induced a rapid improvement (after 1 single month of treatment) in all the sleep quality parameters, suggesting that sleep disturbances should be more carefully evaluated as an additional outcome in these patients.
Collapse
Affiliation(s)
- Sebastian Ferri
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Carlo Montagna
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marta Casini
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luca Malvezzi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Otorhinolaryngology, Head and Neck Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Pirola
- Otorhinolaryngology, Head and Neck Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Elena Russo
- Otorhinolaryngology, Head and Neck Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Racca
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Maria Rita Messina
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Puggioni
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Emanuele Nappi
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giovanni Costanzo
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Lorenzo Del Moro
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giuseppe Mercante
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Otorhinolaryngology, Head and Neck Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giuseppe Spriano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Otorhinolaryngology, Head and Neck Surgery Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Giovanni Paoletti
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Enrico Heffler
- Personalized Medicine, Asthma, and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
| |
Collapse
|
49
|
Madeira Gerardo A, da Silva Alves C, Gomes M, Pardal C, Sokolova A, Liberato H, Mendes A, Tonin FS, Duarte-Ramos F, Lopes C. The Effects of Benralizumab on Lung Volumes and Airway Resistance in Severe Eosinophilic Asthma: A Real-World Study. Cureus 2024; 16:e52452. [PMID: 38371150 PMCID: PMC10871543 DOI: 10.7759/cureus.52452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
INTRODUCTION Add-on biological monoclonal antibodies such as benralizumab (anti-IL-5Ra) are recommended by international guidelines to reduce exacerbations in severe eosinophilic asthma (SEA). However, few studies have assessed the impact of these therapies on lung function-related outcomes. Our goal was to evaluate the effectiveness of benralizumab on lung function, including lung volumes and airway resistance, in SEA patients in Portugal. METHODS This was a real-world, observational, prospective, multicentric study including adult patients diagnosed with SEA (January-June 2023). Spirometry and plethysmography were performed at baseline (T0) and after six months of treatment (T6) with benralizumab to assess: total lung capacity (TLC), residual volume (RV), forced expiratory volume in one second (FEV1), forced vital capacity (FVC), mean forced expiratory flow between 25% and 75% of FVC (mFEF-25/75), intrathoracic gas volume (ITGV), and respiratory airway resistance (Raw). Descriptive statistics (with categorical variables described as frequencies and continuous values as mean and standard deviation (SD)) and paired t-test and Cohen's d effect size were calculated (analyses performed in StataCorp v.15.1; StataCorp LLC, TX, USA). RESULTS Overall, 30 SEA patients were evaluated, mostly women (n=18, 60.0%), with atopy (n=22, 73.3%), a mean age of 58.4 years (SD 11.7), and assisted by pulmonology (n=19, 63.3%) or immunology-allergology (n=11, 36.7%) services. Mean eosinophilia at baseline was 1103.57 cells/mcL (SD 604.88; minimum-maximum 460-2400); after the use of benralizumab, the count dropped to zero. After six months of treatment, a significant increase (p<0.0001) in FVC (15.3%), FEV1 (22.6%), and mFEF-25/75 (17.7%) were observed from baseline (Cohen's d between 0.78 and 1.11). ITGV, RV, RV/TLC, and Raw significantly decreased (p<0.0001) during the study period (-17.3%, -29.7%, -8.9%, and -100.6%, respectively) (Cohen's d between -0.79 and -1.06). No differences in TLC were obtained (p=0.173). No differences between sexes were observed for any measure. Patients with more significant eosinophilia (>900 cells/mcL count; n=15) presented better responses in FEV1 (p=0.001) and mFEF-25/75 (p=0.007). CONCLUSIONS A notable eosinophil depletion with add-on benralizumab led to significant improvements in SEA patients' respiratory function (static lung volumes and airway resistance) in real-world settings after six months. The significant deflating effect of benralizumab on patients' hyperinflated lungs led to enhanced expiratory flow (increased FEV1 and mFEF-25/75) and air trapping (decreased RV/TLC), suggesting this antibody improves bronchial obstruction, lung hyperinflation, and airway resistance. Further studies in a larger population are required to confirm these findings.
Collapse
Affiliation(s)
| | | | - Margarida Gomes
- Allergy and Immunology, Hospital de Santa Maria, Unidade Multidisciplinar de Asma Grave, Lisboa, PRT
| | - Cecília Pardal
- Pulmonology, Hospital Professor Doutor Fernando Fonseca, Lisboa, PRT
| | - Anna Sokolova
- Pulmonology, Hospital Professor Doutor Fernando Fonseca, Lisboa, PRT
| | - Hedi Liberato
- Pulmonology, Hospital Professor Doutor Fernando Fonseca, Lisboa, PRT
| | - Ana Mendes
- Allergy and Immunology, Hospital de Santa Maria, Unidade Multidisciplinar de Asma Grave, Lisboa, PRT
| | - Fernanda S Tonin
- Health and Technology Research Center, Escola Superior de Tecnologia da Saúde de Lisboa (ESTeSL) Instituto Politécnico de Lisboa (IPL), Lisbon, PRT
| | | | - Carlos Lopes
- Pulmonology, Hospital de Santa Maria, Unidade Multidisciplinar de Asma Grave, Lisboa, PRT
| |
Collapse
|
50
|
Cottam A, Manneh IL, Gindeh A, Sillah AK, Cham O, Mendy J, Barry A, Coker EG, Daffeh GK, Badjie S, Barry S, Owolabi O, Winter J, Walzl G, Sutherland JS. The impact of prior SARS-CoV-2 infection on host inflammatory cytokine profiles in patients with TB or other respiratory diseases. Front Immunol 2023; 14:1292486. [PMID: 38179046 PMCID: PMC10764540 DOI: 10.3389/fimmu.2023.1292486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Background Tuberculosis (TB) and COVID-19 are the two leading causes of infectious disease mortality worldwide, and their overlap is likely frequent and inevitable. Previous research has shown increased mortality in TB/COVID-coinfected individuals, and emerging evidence suggests that COVID-19 may increase susceptibility to TB. However, the immunological mechanisms underlying these interactions remain unclear. In this study, we aimed to elucidate the impact of prior or concurrent COVID-19 infection on immune profiles of TB patients and those with other respiratory diseases (ORD). Methods Serum and nasopharyngeal samples were collected from 161 Gambian adolescents and adults with either TB or an ORD. Concurrent COVID-19 infection was determined by PCR, while prior COVID-19 was defined by antibody seropositivity. Multiplex cytokine immunoassays were used to quantify 27 cytokines and chemokines in patient serum samples at baseline, and throughout treatment in TB patients. Results Strikingly, TB and ORD patients with prior COVID-19 infection were found to have significantly reduced expression of several cytokines, including IL-1β, TNF-α and IL-7, compared to those without (p<0.035). Moreover, at month-six of anti-TB treatment, seropositive patients had lower serum Basic FGF (p=0.0115), IL-1β (p=0.0326) and IL-8 (p=0.0021) than seronegative. TB patients with acute COVID-19 coinfection had lower levels of IL-8, IL-13, TNF-α and IP-10 than TB-only patients, though these trends did not reach significance (p>0.035). Conclusions Our findings demonstrate that COVID-19 infection alters the subsequent response to TB and ORDs, potentially contributing to pathogenesis. Further work is necessary to determine whether COVID-19 infection accelerates TB disease progression, though our results experimentally support this hypothesis.
Collapse
Affiliation(s)
- Annabelle Cottam
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ismaila L. Manneh
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Awa Gindeh
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Abdou K. Sillah
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Ousainou Cham
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Joseph Mendy
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Amadou Barry
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Edward G. Coker
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Georgetta K. Daffeh
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Simon Badjie
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Salieu Barry
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Olumuyiwa Owolabi
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Jill Winter
- Catalysis Foundation for Health, Emeryville, CA, United States
| | - Gerhard Walzl
- Department of Molecular Biology and Human Genetics, University of Stellenbosch, Tygerberg, South Africa
| | - Jayne S. Sutherland
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia
| |
Collapse
|