1
|
Jacobs I, Deleu S, Cremer J, De Hertogh G, Vermeire S, Breynaert C, Vanuytsel T, Verstockt B. Eosinophil Depletion as a Potential Therapeutic Strategy in Acute and Chronic Intestinal Inflammation Based on a Dextran Sulfate Sodium Colitis Model. Inflamm Bowel Dis 2025; 31:169-177. [PMID: 39107256 DOI: 10.1093/ibd/izae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND A role for eosinophils in intestinal inflammation and fibrosis in the context of inflammatory bowel disease has been suggested, yet the precise nature, whether causal or secondary remains debated. Hence, it remains unclear whether targeting eosinophils should be further explored as a treatment option in inflammatory bowel disease. METHODS Acute and chronic dextran sulfate sodium colitis was induced in wild-type C57BL/6 mice. Eosinophils were depleted by anti-CCR3 injections before colitis induction in a chronic model and after colitis onset in an acute model in order to investigate the impact of eosinophil depletion on pre-existing colitis. Inflammation was assessed using the disease activity index, macroscopic damage, and histological disease activity score. In the chronic model, fibrosis was assessed by examining colon weight/length ratio, collagen deposition through Martius Scarlet Blue staining, hydroxyproline assay, and COL1A1 expression. Protein and gene expression were assessed using the Meso Scale Discovery platform and real-time quantitative polymerase chain reaction. RESULTS In the acute and chronic colitis model, eosinophil depletion resulted in reduced disease activity and faster recovery, as observed via the total area under the curve of the disease activity index (P = .004 and P = .02, respectively), macroscopic damage score (P = .009 and P = .08, respectively), and histological disease activity score (P = .09 and P = .002, respectively). In the acute model, the accelerated recovery was accompanied by an increase in interleukin (IL)-10 (P = .03) and a decrease in IL-4 (P = .03) and IL-6 (P = .009). Colon weight/length ratio and collagen deposition were not affected by eosinophil depletion. CONCLUSIONS Eosinophil depletion prevents and decreases intestinal inflammation in a preclinical dextran sulfate sodium model without affecting fibrosis. These results pave the way for exploring eosinophil depletion as a novel treatment modality in addressing intestinal inflammation.
Collapse
Affiliation(s)
- Inge Jacobs
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Sara Deleu
- Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Jonathan Cremer
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Gert De Hertogh
- Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Christine Breynaert
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of General Internal Medicine, UZ Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Bram Verstockt
- Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Rubin L, Talmon A, Ribak Y, Lavie D, Nechushtan H, Caplan N, Lotem M, Shamriz O, Adini I, Tal Y. Targeted inhibition of the IL5 axis for immune checkpoint inhibitors eosinophilic-induced adverse events. J Immunother Cancer 2024; 12:e009658. [PMID: 39395838 PMCID: PMC11474678 DOI: 10.1136/jitc-2024-009658] [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: 05/17/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
Given the broad implementation of immune checkpoint inhibitors (ICI) for cancer therapy, we encounter a variety of immune-related adverse events (irAE) including immune-related blood eosinophilia. Eosinophilia demonstrated a potential positive predictive marker for a beneficial clinical response to ICI. However, there are reports of eosinophil-induced adverse events (Eo-irAE) with organ dysfunction requiring initiation of oral glucocorticoid therapy and discontinuation of ICI.We aim to assess the efficacy and safety of interleukin (IL) 5-axis inhibition in Eo-irAE secondary to ICI therapy.We present three cases of Eo-irAE referred to our allergy and clinical immunology unit at Hadassah Hebrew University Medical Center following therapy with pembrolizumab and nivolumab, monoclonal antibodies that target the programmed cell death 1 (PD-1) receptor, for two cases of melanoma and one metastatic non-small cell lung carcinoma. Following informed consent and committee approval, two patients were treated with 1-3 doses of mepolizumab, 100 mg, monoclonal IgG1 kappa anti-IL-5 antibody, and one patient received up-to-date 9 doses of benralizumab, 30 mg, monoclonal IgG1 kappa antibody directed against the alpha chain of the interleukin-5 receptor, both administered subcutaneously. Patients were carefully followed and treatment response was assessed by physical examinations and laboratory tests.Hypereosinophilia at the level of 2300-8000 K/UL was observed 8-12 months following therapy accompanied by symptoms of dyspnea, arthralgia, myalgia, fasciitis, 'morphea'-like lesions, fatigue, abdominal discomfort, pruritus, and chest pain. ICI discontinuation did not improve symptoms, two patients were resistant to glucocorticoids and therefore biological treatment was initiated to inhibit the IL5 axis. Patients demonstrated rapid clinical response and a decrease in peripheral blood eosinophil levels with long-term symptoms remission. There were no signals of negative impacts, such as tumor progression following IL5 axis inhibition.Eosinophilia secondary to ICI therapy can lead to organ dysfunction. Discontinuation of ICI might not be effective and symptoms may be refractory to steroid therapy hence targeted inhibition of the IL5 axis should be considered.
Collapse
Affiliation(s)
- Limor Rubin
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah University Medical Center Division of Internal Medicine, Jerusalem, Israel
| | - Aviv Talmon
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah University Medical Center Division of Internal Medicine, Jerusalem, Israel
| | - Yaarit Ribak
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah University Medical Center Division of Internal Medicine, Jerusalem, Israel
| | - David Lavie
- Department of Hematology, Hadassah University Medical Center, Jerusalem, Israel
| | - Hovav Nechushtan
- Oncology, Hadassah Medical Center, Jerusalem, Israel
- Hebrew University, Jerusalem, Israel
| | - Nadia Caplan
- Department of Radiology, Hadassah University Medical Center, Jerusalem, Israel
| | - Michal Lotem
- Oncology, Hadassah Medical Center, Jerusalem, Israel
| | - Oded Shamriz
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah University Medical Center, Jerusalem, Israel
- Institute of Medical Research Israel-Canada, Faculty of Medicine, Hebrew University The Lautenberg Center for Immunology and Cancer, Jerusalem, Israel
| | - Irit Adini
- Department of Surgery, Center for Engineering in Medicine and Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuval Tal
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah University Medical Center Division of Internal Medicine, Jerusalem, Israel
| |
Collapse
|
3
|
Arima M, Ito K, Abe T, Oguma T, Asano K, Mukherjee M, Ueki S. Eosinophilic mucus diseases. Allergol Int 2024; 73:362-374. [PMID: 38594175 DOI: 10.1016/j.alit.2024.03.002] [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: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
Eosinophilic inflammation is primarily characterized by type 2 immune responses against parasitic organisms. In the contemporary human being especially in developed countries, eosinophilic inflammation is strongly associated with allergic/sterile inflammation, and constitutes an undesired immune reaction. This situation is in stark contrast to neutrophilic inflammation, which is indispensable for the host defense against bacterial infections. Among eosinophilic inflammatory disorders, massive accumulation of eosinophils within mucus is observed in certain cases, and is often linked to the distinctive clinical finding of mucus with high viscosity. Eosinophilic mucus is found in a variety of diseases, including chronic allergic keratoconjunctivitis, chronic rhinosinusitis encompassing allergic fungal sinusitis, eosinophilic otitis media, eosinophilic sialodochitis, allergic bronchopulmonary aspergillosis/mycosis, eosinophilic plastic bronchitis, and eosinophilic asthma. In these pathological conditions, chronic inflammation and tissue remodeling coupled with irreversible organ damage due to persistent adhesion of toxic substances and luminal obstruction may impose a significant burden on the body. Eosinophils aggregate in the hyperconcentrated mucus together with cell-derived crystals, macromolecules, and polymers, thereby affecting the biophysical properties of the mucus. This review focuses on the clinically significant challenges of mucus and discusses the consequences of activated eosinophils on the mucosal surface that impact mucus and persistent inflammation.
Collapse
Affiliation(s)
- Misaki Arima
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Keisuke Ito
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Tomoe Abe
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Tsuyoshi Oguma
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Manali Mukherjee
- Department of Medicine, McMaster University & St Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Shigeharu Ueki
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita, Japan.
| |
Collapse
|
4
|
Jesenak M, Diamant Z, Simon D, Tufvesson E, Seys SF, Mukherjee M, Lacy P, Vijverberg S, Slisz T, Sediva A, Simon HU, Striz I, Plevkova J, Schwarze J, Kosturiak R, Alexis NE, Untersmayr E, Vasakova MK, Knol E, Koenderman L. Eosinophils-from cradle to grave: An EAACI task force paper on new molecular insights and clinical functions of eosinophils and the clinical effects of targeted eosinophil depletion. Allergy 2023; 78:3077-3102. [PMID: 37702095 DOI: 10.1111/all.15884] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/21/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023]
Abstract
Over the past years, eosinophils have become a focus of scientific interest, especially in the context of their recently uncovered functions (e.g. antiviral, anti-inflammatory, regulatory). These versatile cells display both beneficial and detrimental activities under various physiological and pathological conditions. Eosinophils are involved in the pathogenesis of many diseases which can be classified into primary (clonal) and secondary (reactive) disorders and idiopathic (hyper)eosinophilic syndromes. Depending on the biological specimen, the eosinophil count in different body compartments may serve as a biomarker reflecting the underlying pathophysiology and/or activity of distinct diseases and as a therapy-driving (predictive) and monitoring tool. Personalized selection of an appropriate therapeutic strategy directly or indirectly targeting the increased number and/or activity of eosinophils should be based on the understanding of eosinophil homeostasis including their interactions with other immune and non-immune cells within different body compartments. Hence, restoring as well as maintaining homeostasis within an individual's eosinophil pool is a goal of both specific and non-specific eosinophil-targeting therapies. Despite the overall favourable safety profile of the currently available anti-eosinophil biologics, the effect of eosinophil depletion should be monitored from the perspective of possible unwanted consequences.
Collapse
Affiliation(s)
- Milos Jesenak
- Department of Clinical Immunology and Allergology, University Teaching Hospital in Martin, Martin, Slovak Republic
- Department of Paediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic
| | - Zuzana Diamant
- Department of Clinical Sciences Lund, Respiratory Medicine, Allergology and Palliative Medicine, Lund University, Lund, Sweden
- Department Microbiology Immunology & Transplantation, KU Leuven, Catholic University of Leuven, Leuven, Belgium
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ellen Tufvesson
- Department of Clinical Sciences Lund, Respiratory Medicine, Allergology and Palliative Medicine, Lund University, Lund, Sweden
| | - Sven F Seys
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Manali Mukherjee
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- The Firestone Institute for Respiratory Health, Research Institute of St. Joe's Hamilton, Hamilton, Ontario, Canada
| | - Paige Lacy
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Susanne Vijverberg
- Amsterdam UMC Location University of Amsterdam, Pulmonary Diseases, Amsterdam, The Netherlands
| | - Tomas Slisz
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Anna Sediva
- Department of Immunology, 2nd Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jana Plevkova
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - Jurgen Schwarze
- Child Life and Health and Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Radovan Kosturiak
- Department of Paediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Teaching Hospital in Martin, Martin, Slovak Republic
- Outpatient Clinic for Clinical Immunology and Allergology, Nitra, Slovak Republic
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, Department of Paediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Martina Koziar Vasakova
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Edward Knol
- Department Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leo Koenderman
- Department Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department Pulmonary Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
5
|
Mormile M, Mormile I, Fuschillo S, Rossi FW, Lamagna L, Ambrosino P, de Paulis A, Maniscalco M. Eosinophilic Airway Diseases: From Pathophysiological Mechanisms to Clinical Practice. Int J Mol Sci 2023; 24:ijms24087254. [PMID: 37108417 PMCID: PMC10138384 DOI: 10.3390/ijms24087254] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Eosinophils play a key role in airway inflammation in many diseases, such as allergic and non-allergic asthma, chronic rhinosinusitis with nasal polyps, and chronic obstructive pulmonary disease. In these chronic disabling conditions, eosinophils contribute to tissue damage, repair, remodeling, and disease persistence through the production a variety of mediators. With the introduction of biological drugs for the treatment of these respiratory diseases, the classification of patients based on clinical characteristics (phenotype) and pathobiological mechanisms (endotype) has become mandatory. This need is particularly evident in severe asthma, where, despite the great scientific efforts to understand the immunological pathways underlying clinical phenotypes, the identification of specific biomarkers defining endotypes or predicting pharmacological response remains unsatisfied. In addition, a significant heterogeneity also exists among patients with other airway diseases. In this review, we describe some of the immunological differences in eosinophilic airway inflammation associated with severe asthma and other airway diseases and how these factors might influence the clinical presentation, with the aim of clarifying when eosinophils play a key pathogenic role and, therefore, represent the preferred therapeutic target.
Collapse
Affiliation(s)
- Mauro Mormile
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Ilaria Mormile
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Salvatore Fuschillo
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy
| | - Francesca Wanda Rossi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Laura Lamagna
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Directorate of Telese Terme Institute, 82037 Telese Terme, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Mauro Maniscalco
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy
| |
Collapse
|
6
|
Lin J, Chen D, Guan L, Chang K, Li D, Sun B, Yang P, Liu Z. House dust mite exposure enhances immune responses to ovalbumin-induced intestinal allergy. Sci Rep 2022; 12:5216. [PMID: 35338219 PMCID: PMC8956666 DOI: 10.1038/s41598-022-09196-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/16/2022] [Indexed: 11/24/2022] Open
Abstract
House dust mites (HDM) are one of the important factors of airway allergic diseases, HDM allergens can be detected in the human gut mucosa, which induces local inflammation and increases intestinal epithelial permeability. This study tests a hypothesis that HDM contribute to the development of OVA (ovalbumin)-induced intestinal allergy. The serum levels of IgE against HDM in patients with food allergy were detected with UniCAP100 (Pharmacia, Uppsala, Sweden); a mouse model of food allergy was developed with OVA and HDM as the specific antigens. Compared to healthy controls, patients with food allergy have higher levels of serum HDM-specific IgE. Compared to food allergy alone groups, the levels of HDM-specific IgE in patients with food allergy and asthma or allergic rhinitis were significantly higher. In mouse models, we found that HDM/OVA induced allergy-like symptoms, lower body temperature, and lower body weight. The levels of IgE, IgG1, mMCP-1 (mouse mast cell protease-1), IL-4 and IL-5 in the HDM and HDM + CT (cholera toxin) groups were higher than the control groups, and the levels of IgE, IgG1, IL-4 and IL-5 in the HDM, OVA and HDM + OVA groups were higher than the control groups. The pathological changes of intestinal tissues in the HDM and HDM + CT/the HDM, OVA and HDM + OVA groups were more severe, more eosinophil infiltration than the control groups. Moreover, exposure to HDM induced intestinal barrier dysfunction, and facilitated the development of intestinal allergy in mice. In conclusion, HDM exposure enhances immune responses to OVA-induced food allergy.
Collapse
Affiliation(s)
- Jianli Lin
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Room 509 of A7 Bldgn, 1066 Xueyuan Blvd, Shenzhen, 518055, China.,State Key Laboratory of Respiratory Disease, Department of Allergy and Clinical Immunology, National Clinical Center for Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, First Affiliated Hospital, Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, Guangdong Province, China
| | - Desheng Chen
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Room 509 of A7 Bldgn, 1066 Xueyuan Blvd, Shenzhen, 518055, China.,Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Wujin road 85, Shanghai, 200080, China
| | - Lvxin Guan
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Room 509 of A7 Bldgn, 1066 Xueyuan Blvd, Shenzhen, 518055, China
| | - Kexin Chang
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Room 509 of A7 Bldgn, 1066 Xueyuan Blvd, Shenzhen, 518055, China
| | - Dan Li
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Room 509 of A7 Bldgn, 1066 Xueyuan Blvd, Shenzhen, 518055, China
| | - Baoqing Sun
- State Key Laboratory of Respiratory Disease, Department of Allergy and Clinical Immunology, National Clinical Center for Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, First Affiliated Hospital, Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, Guangdong Province, China.
| | - Pingchang Yang
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Room 509 of A7 Bldgn, 1066 Xueyuan Blvd, Shenzhen, 518055, China.
| | - Zhigang Liu
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Room 509 of A7 Bldgn, 1066 Xueyuan Blvd, Shenzhen, 518055, China. .,State Key Laboratory of Respiratory Disease, Department of Allergy and Clinical Immunology, National Clinical Center for Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, First Affiliated Hospital, Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, Guangdong Province, China.
| |
Collapse
|
7
|
Zou H, Yang Y, Dai H, Xiong Y, Wang JQ, Lin L, Chen ZS. Recent Updates in Experimental Research and Clinical Evaluation on Drugs for COVID-19 Treatment. Front Pharmacol 2021; 12:732403. [PMID: 34880750 PMCID: PMC8646041 DOI: 10.3389/fphar.2021.732403] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022] Open
Abstract
Since the outbreak of corona virus disease 2019 (COVID-19) in Wuhan (China) in December 2019, the epidemic has rapidly spread to many countries around the world, posing a huge threat to global public health. In response to the pandemic, a number of clinical studies have been initiated to evaluate the effect of various treatments against COVID-19, combining medical strategies and clinical trial data from around the globe. Herein, we summarize the clinical evaluation about the drugs mentioned in this review for COVID-19 treatment. This review discusses the recent data regarding the efficacy of various treatments in COVID-19 patients, to control and prevent the outbreak.
Collapse
Affiliation(s)
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Huiqiang Dai
- Cell Research Center, Shenzhen Bolun Institute of Biotechnology, Shenzhen, China
| | - Yunchuang Xiong
- Cell Research Center, Shenzhen Bolun Institute of Biotechnology, Shenzhen, China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Lusheng Lin
- Cell Research Center, Shenzhen Bolun Institute of Biotechnology, Shenzhen, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| |
Collapse
|
8
|
Rhyou HI, Han SH, Nam YH. Successful induction treatment of bullous pemphigoid using reslizumab: a case report. Allergy Asthma Clin Immunol 2021; 17:117. [PMID: 34784964 PMCID: PMC8596938 DOI: 10.1186/s13223-021-00619-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/21/2021] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Bullous pemphigoid (BP) is a potentially life-threatening autoimmune blistering disease which is characterized by autoantibodies against hemidesmosomal proteins of the skin and mucous membranes. In recent years, the role of eosinophil and immunoglobulin E autoantibodies have been further elucidated in BP, and have been considered potential therapeutic targets.
Case presentations
A 67-year-old male presented with erythematous bullous eruption. The skin eruption was located on whole body, and suggested BP. Peripheral blood eosinophil count and total immunoglobulin E markedly elevated in initial laboratory findings. Topical and systemic steroid (methylprednisolone 2 mg/kg/day) treatment was started, and his skin symptoms worsened repeatedly, whenever systemic steroid were reduced. On admission day 29, reslizumab (anti-interleukin-5) 3.5 mg/kg was administered intravenously to the patients. The bullous skin lesion began to improve rapidly, and methylprednisolone (8 mg/day) was reduced without any worsening of symptoms during two doses of reslizumab.
Conclusions
We report a case of successful treatment response to reslizumab administration in a patient with BP. Further studies are needed to confirm the role of anti-interleukin-5 as a treatment for BP in the future.
Collapse
|
9
|
Jacobs I, Ceulemans M, Wauters L, Breynaert C, Vermeire S, Verstockt B, Vanuytsel T. Role of Eosinophils in Intestinal Inflammation and Fibrosis in Inflammatory Bowel Disease: An Overlooked Villain? Front Immunol 2021; 12:754413. [PMID: 34737752 PMCID: PMC8560962 DOI: 10.3389/fimmu.2021.754413] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/30/2021] [Indexed: 12/20/2022] Open
Abstract
Eosinophils are leukocytes which reside in the gastrointestinal tract under homeostatic conditions, except for the esophagus which is normally devoid of eosinophils. Research on eosinophils has primarily focused on anti-helminth responses and type 2 immune disorders. In contrast, the search for a role of eosinophils in chronic intestinal inflammation and fibrosis has been limited. With a shift in research focus from adaptive to innate immunity and the fact that the eosinophilic granules are filled with inflammatory mediators, eosinophils are becoming a point of interest in inflammatory bowel diseases. In the current review we summarize eosinophil characteristics and recruitment as well as the current knowledge on presence, inflammatory and pro-fibrotic functions of eosinophils in inflammatory bowel disease and other chronic inflammatory conditions, and we identify research gaps which should be covered in the future.
Collapse
Affiliation(s)
- Inge Jacobs
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Matthias Ceulemans
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lucas Wauters
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Christine Breynaert
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Katholieke Universiteit Leuven, Leuven, Belgium
- Department of General Internal Medicine, Allergy and Clinical Immunology, University Hospitals Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
10
|
The Cycling of Intracellular Calcium Released in Response to Fluid Shear Stress Is Critical for Migration-Associated Actin Reorganization in Eosinophils. Cells 2021; 10:cells10010157. [PMID: 33467432 PMCID: PMC7829934 DOI: 10.3390/cells10010157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 01/08/2023] Open
Abstract
The magnitude of eosinophil mobilization into respiratory tissues drives the severity of inflammation in several airway diseases. In classical models of leukocyte extravasation, surface integrins undergo conformational switches to high-affinity states via chemokine binding activation. Recently, we learned that eosinophil integrins possess mechanosensitive properties that detect fluid shear stress, which alone was sufficient to induce activation. This mechanical stimulus triggered intracellular calcium release and hallmark migration-associated cytoskeletal reorganization including flattening for increased cell–substratum contact area and pseudopodia formation. The present study utilized confocal fluorescence microscopy to investigate the effects of pharmacological inhibitors to calcium signaling and actin polymerization pathways on shear stress-induced migration in vitro. Morphological changes (cell elongation, membrane protrusions) succeeded the calcium flux in untreated eosinophils within 2 min, suggesting that calcium signaling was upstream of actin cytoskeleton rearrangement. The inhibition of ryanodine receptors and endomembrane Ca2+-ATPases corroborated this idea, indicated by a significant increase in time between the calcium spike and actin polymerization. The impact of the temporal link is evident as the capacity of treated eosinophils to move across fibronectin-coated surfaces was significantly hampered relative to untreated eosinophils. Furthermore, we determined that the nature of cellular motility in response to fluid shear stress was nondirectional.
Collapse
|
11
|
Low-dose chlorine exposure impairs lung function, inflammation and oxidative stress in mice. Life Sci 2020; 267:118912. [PMID: 33338503 DOI: 10.1016/j.lfs.2020.118912] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022]
Abstract
AIM To explore the different consequences of acute and chronic exposure to chlorine gas (Cl2) on the functional and histological parameters of health mice. MAIN METHODS Firstly, male BALB/c mice were acute exposed to 3.3 or 33.3 or 70.5 mg/m3 Cl2. We analyzed the lung function, the inflammatory cells in the bronchoalveolar lavage, cell influx in the peribrochoalveolar space and mucus production. In a second phase, mice were chronic exposed to 70.5 mg/m3 Cl2. Besides the first phase analyses, we also evaluated the epithelial cells thickness, collagen deposition in the airways, immunohistochemistry stain for IL-1β, iNOS, IL-17 and ROCK-2 and the levels of IL-5, IL-13, IL-17, IL-1β and TNF-α in lung homogenate. KEY FINDINGS Acute exposure to chlorine impaired the lung function, increased the number of inflammatory cells in the BALF and in the airways, also increased the mucus production. Furthermore, when chlorine was exposed chronically, increased the airway remodeling with collagen deposition and epithelial cells thickness, positive cells for IL-1β, iNOS, IL-17 in the airways and in the alveolar walls and ROCK-2 in the alveolar walls, lung inflammation with increased levels of IL-5, IL-13, IL-1β and TNF-α in the lung homogenate, and also, induced the acid mucus production by the nasal epithelium. SIGNIFICANCE Acute and chronic exposure to low dose of chlorine gas worsens lung function, induces oxidative stress activation and mucus production and contributes to augmenting inflammation in health mice.
Collapse
|
12
|
Knight V, Long T, Meng QH, Linden MA, Rhoads DD. Variability in the Laboratory Measurement of Cytokines. Arch Pathol Lab Med 2020; 144:1230-1233. [PMID: 32401053 DOI: 10.5858/arpa.2019-0519-cp] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— The measurement of cytokines in clinical laboratories is becoming an increasingly routine part of immune monitoring when administering biologic and cell-based immunotherapies and also for clinical assessment of inflammatory conditions. While a number of commercial assays and platforms are available for cytokine measurement, there is currently little standardization among these analytical methods. OBJECTIVE.— To characterize the variability and comparability among cytokine testing platforms that are commonly used in clinical laboratories. DESIGN.— We analyzed data for 4 cytokines (interleukin [IL]-1, IL-6, IL-8, and tumor necrosis factor-alpha [TNF-α]) from 6 College of American Pathologists cytokine surveys administered from 2015 to 2018. Analyses interrogated variability between testing methods and variability within each laboratory across the mailings. RESULTS.— Significant variability was noted across methods with analysis of IL-1 showing the least variability and IL-6, IL-8, and TNF-α varying between methods to a greater extent. Intralab variability was also significant with TNF-α measurements again showing the greatest variability. CONCLUSIONS.— This retrospective analysis of College of American Pathologists proficiency testing data for cytokine measurement is the largest method comparison to date, and this study provides a description of the variation of cytokine measurement across methods, across laboratories, and within laboratories. Serial monitoring of cytokines should preferentially be performed by the same method within the same laboratory.
Collapse
Affiliation(s)
- Vijaya Knight
- From the Department of Pediatrics, Section of Allergy and Immunology, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora (Knight)
| | - Thomas Long
- Department of Biostatistics, College of American Pathologists, Northfield, Illinois (Long)
| | - Qing H Meng
- Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston (Meng)
| | - Michael A Linden
- Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (Linden)
| | - Daniel D Rhoads
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio (Rhoads)
| |
Collapse
|
13
|
Mukherjee M, Forero DF, Tran S, Boulay ME, Bertrand M, Bhalla A, Cherukat J, Al-Hayyan H, Ayoub A, Revill SD, Javkar T, Radford K, Kjarsgaard M, Huang C, Dvorkin-Gheva A, Ask K, Olivenstein R, Dendukuri N, Lemiere C, Boulet LP, Martin JG, Nair P. Suboptimal treatment response to anti-IL-5 monoclonal antibodies in severe eosinophilic asthmatics with airway autoimmune phenomena. Eur Respir J 2020; 56:13993003.00117-2020. [PMID: 32444405 DOI: 10.1183/13993003.00117-2020] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/08/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND In clinical trials, the two anti-interleukin (IL)-5 monoclonal antibodies (mAbs: mepolizumab and reslizumab) approved to treat severe eosinophilic asthma reduce exacerbations by ∼50-60%. OBJECTIVE To observe response to anti-IL-5 mAbs in a real-life clinical setting, and to evaluate predictors of suboptimal response. METHODS In four Canadian academic centres, predefined clinical end-points in 250 carefully characterised moderate-to-severe asthmatic patients were collected prospectively to assess response to the two anti-IL-5 mAbs. Suboptimal response was determined based on failure to reduce maintenance corticosteroid (MCS) or asthma symptoms scores (Asthma Control Questionnaire (ACQ)) or exacerbations, in addition to persistence of sputum/blood eosinophils. Worsening in suboptimal responders was assessed based on reduced lung function by 25% or increase in MCS/ACQ. A representative subset of 39 patients was evaluated for inflammatory mediators, autoantibodies and complement activation in sputum (by ELISA) and for immune-complex deposition by immunostaining formalin-fixed paraffin-embedded sputum plugs. RESULTS Suboptimal responses were observed in 42.8% (107 out of 250) patients treated with either mepolizumab or reslizumab. Daily prednisone requirement, sinus disease and late-onset asthma diagnoses were the strongest predictors of suboptimal response. Asthma worsened in 13.6% (34 out of 250) of these patients. The majority (79%) of them were prednisone-dependent. Presence of sputum anti-eosinophil peroxidase immunoglobulin (Ig)G was a predictor of suboptimal response to an anti-IL-5 mAb. An increase in sputum C3c (marker of complement activation) and deposition of C1q-bound/IL-5-bound IgG were observed in the sputa of those patients who worsened on therapy, suggesting an underlying autoimmune-mediated pathology. CONCLUSION A significant number of patients who meet currently approved indications for anti-IL5 mAbs show suboptimal response to them in real-life clinical practice, particularly if they are on high doses of prednisone. Monitoring blood eosinophil count is not helpful to identify these patients. The concern of worsening of symptoms associated with immune-complex mediated complement activation in a small proportion of these patients highlights the relevance of recognising airway autoimmune phenomena and this requires further evaluation.
Collapse
Affiliation(s)
- Manali Mukherjee
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada.,Supervision of this work was shared by M. Mukherjee and P. Nair, and both take overall guarantee of the manuscript
| | - David Felipe Forero
- Technology Assessment Unit, McGill University Health Centre, Montreal, QC, Canada
| | - Stephanie Tran
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Marie-Eve Boulay
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Mylène Bertrand
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Anurag Bhalla
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Jayant Cherukat
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Hajar Al-Hayyan
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Anmar Ayoub
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Spencer D Revill
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Tanvi Javkar
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Katherine Radford
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Melanie Kjarsgaard
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Chynna Huang
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Anna Dvorkin-Gheva
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Ronald Olivenstein
- Dept of Medicine, McGill University, Montreal, QC, Canada.,Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nandini Dendukuri
- Centre for Outcomes Research, Department of Medicine, McGill University, Montreal, QC, Canada
| | | | - Louis-Philippe Boulet
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - James G Martin
- Dept of Medicine, McGill University, Montreal, QC, Canada.,Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Parameswaran Nair
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada.,Supervision of this work was shared by M. Mukherjee and P. Nair, and both take overall guarantee of the manuscript
| |
Collapse
|
14
|
Chiarella E, Lombardo N, Lobello N, Aloisio A, Aragona T, Pelaia C, Scicchitano S, Bond HM, Mesuraca M. Nasal Polyposis: Insights in Epithelial-Mesenchymal Transition and Differentiation of Polyp Mesenchymal Stem Cells. Int J Mol Sci 2020; 21:E6878. [PMID: 32961745 PMCID: PMC7555226 DOI: 10.3390/ijms21186878] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic rhinosinusitis is a common inflammatory disease of paranasal sinuses, which causes rhinorrhea, nasal congestion, and hyposmia. The genetic predisposition or the exposure to irritants can sustain the inflammatory response and the development of nasal polyposis. Nasal polyps are benign and teardrop-shaped growths that project in the nasal cavities, and originate from the ethmoid sinuses. This inflammatory process is associated with high expression of IL-4, IL-5 and IL-13 and IgE. Antibodies targeting these cytokines or receptors represent a therapeutic strategy in the treatment of nasal polyposis in combination with corticosteroids. The molecular pathogenesis of nasal polyps in chronic rhinosinusitis (CRS) patients is associated with remodeling transition, a process in which epithelial cells lose their typical phenotype, acquiring a mesenchymal-like aspect. TGFβ/SMAD, ERK, and Wnt/β-catenin pathways are altered during the nasal tissue remodeling. miRNA and inhibitor molecules targeting these signaling pathways are able to interfere with the process; which could lead to alternative therapies. Nasal polyps are an alternative source of mesenchymal stem cells, which can be isolated from surgical biopsies. A molecular understanding of the biology of PO-MSCs will contribute to the delineating inflammatory process underlying the development of nasal polyps.
Collapse
Affiliation(s)
- Emanuela Chiarella
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University “Magna Græcia”, 88100 Catanzaro, Italy; (E.C.); (A.A.); (S.S.)
| | - Nicola Lombardo
- Otolaryngology Head and Neck Surgery, Department Medical and Surgical Sciences, University “Magna Græcia”, 88100 Catanzaro, Italy; (N.L.); (N.L.)
| | - Nadia Lobello
- Otolaryngology Head and Neck Surgery, Department Medical and Surgical Sciences, University “Magna Græcia”, 88100 Catanzaro, Italy; (N.L.); (N.L.)
| | - Annamaria Aloisio
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University “Magna Græcia”, 88100 Catanzaro, Italy; (E.C.); (A.A.); (S.S.)
| | - Teodoro Aragona
- Otolaryngology, A.O.U. Ospedali Riuniti, 60123 Ancona, Italy;
| | - Corrado Pelaia
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Stefania Scicchitano
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University “Magna Græcia”, 88100 Catanzaro, Italy; (E.C.); (A.A.); (S.S.)
| | - Heather Mandy Bond
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University “Magna Græcia”, 88100 Catanzaro, Italy; (E.C.); (A.A.); (S.S.)
| | - Maria Mesuraca
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University “Magna Græcia”, 88100 Catanzaro, Italy; (E.C.); (A.A.); (S.S.)
| |
Collapse
|
15
|
Calzetta L, Ritondo BL, Matera MG, Facciolo F, Rogliani P. Targeting IL-5 pathway against airway hyperresponsiveness: A comparison between benralizumab and mepolizumab. Br J Pharmacol 2020; 177:4750-4765. [PMID: 32857420 DOI: 10.1111/bph.15240] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/14/2020] [Accepted: 08/10/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND AND PURPOSE Airway hyperresponsiveness (AHR) is a central abnormality in asthma. IL-5 may modulate AHR in animal models of asthma, but the available data is inconsistent on the impact of targeting IL-5 pathway against AHR. The difference between targeting IL-5 or the IL-5 receptor, α subunit (IL-5Rα) in modulating AHR remains to be investigated in human airways. The aim of this study was to compare the role of the anti-IL-5Rα benralizumab and the anti-IL-5 mepolizumab against AHR and to assess whether these agents influence the levels of cAMP. EXPERIMENTAL APPROACH Passively sensitized human airways were treated with benralizumab and mepolizumab. The primary endpoint was the inhibition of AHR to histamine. The secondary endpoints were the protective effect against AHR to parasympathetic activation and mechanical stress, and the tissue modulation of cAMP. KEY RESULTS Benralizumab and mepolizumab significantly inhibited the AHR to histamine (maximal effect -134.14 ± 14.93% and -108.29 ± 32.16%, respectively), with benralizumab being 0.73 ± 0.10 logarithm significantly more potent than mepolizumab. Benralizumab and mepolizumab significantly inhibited the AHR to transmural stimulation and mechanical stress. Benralizumab was 0.45 ± 0.16 logarithm significantly more potent than mepolizumab against AHR to parasympathetic activation. The effect of these agents was significantly correlated with increased levels of cAMP. CONCLUSION AND IMPLICATIONS Targeting the IL-5/IL-5Rα axis is an effective strategy to prevent the AHR. Benralizumab was more potent than the mepolizumab and the concentration-dependent beneficial effects of both these monoclonal antibodies were related to improved levels of cAMP in hyperresponsive airways.
Collapse
Affiliation(s)
- Luigino Calzetta
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Beatrice Ludovica Ritondo
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Facciolo
- Thoracic Surgery Unit, Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
16
|
Kisling A, Jones J, Hixson C, Hostler D, Hostler J. Mepolizumab: an alternative therapy for idiopathic chronic eosinophilic pneumonia with glucocorticoid intolerance. Drugs Context 2020; 9:dic-2020-5-3. [PMID: 32884574 PMCID: PMC7440848 DOI: 10.7573/dic.2020-5-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 11/21/2022] Open
Abstract
A 55-year-old woman with cough-variant asthma presented for 1 month of worsening wheezing, cough, and dyspnea refractory to treatment. Initial laboratory findings revealed profound peripheral eosinophilia, and a chest computed tomography showed bi-apical consolidation. Bronchio-alveolar lavage demonstrated alveolar eosinophilia. She was diagnosed with idiopathic chronic eosinophilic pneumonia (ICEP). Her peripheral eosinophilia and respiratory symptoms improved rapidly with high-dose systemic corticosteroid therapy. However, she was intolerant to corticosteroid monotherapy due to non-compliance and psychological adverse effects. Mepolizumab was initiated as a steroid-sparing agent, resulting in successful therapy for 2 years without relapse or adverse effects. Mepolizumab is an interleukin-5 (IL-5) antagonist monoclonal antibody, which is a targeted therapy for diseases mediated by eosinophil activity and eosinophil proliferation. Mepolizumab is typically used in ICEP refractory to steroids, but this case supports its use in cases of glucocorticoid intolerance. Further study of IL-5 antagonist therapies for ICEP may identify an alternative treatment modality for patients in whom the adverse effects of corticosteroids pose a challenge.
Collapse
Affiliation(s)
- Adam Kisling
- Department of Medicine, Tripler Army Medical Center, Honolulu, HI, USA
| | - Jason Jones
- Department of Medicine, Tripler Army Medical Center, Honolulu, HI, USA
| | - Caleb Hixson
- Department of Pathology, Tripler Army Medical Center, Honolulu, HI, USA
| | - David Hostler
- Department of Pulmonology, Tripler Army Medical Center, Honolulu, HI, USA
| | - Jordanna Hostler
- Department of Pulmonology, Tripler Army Medical Center, Honolulu, HI, USA
| |
Collapse
|
17
|
Xie X, Xu D, Zhuang L, Liu H, Tan S, Lu Y, Su M, Chen J, Pan H, Lu L, Xu Y, Liao M, Xu Z, He J. Sanfu herbal patch applied at acupoints in patients with bronchial asthma: study protocol for a randomized controlled trial. Trials 2020; 21:684. [PMID: 32727619 PMCID: PMC7389380 DOI: 10.1186/s13063-020-04604-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/12/2020] [Indexed: 11/29/2022] Open
Abstract
Background Bronchial asthma is one of the most common inflammatory airway disorders. As one of the main non-drug therapies, the Sanfu herbal patch (SHP) has been widely used to treat bronchial asthma, although the evidence for its efficacy and associated mechanism are inconclusive. The objective of this trial is to clarify the clinical efficacy and safety of the SHP in the treatment of bronchial asthma in the chronic persistent or clinical remission stage and to provide high-quality data for further research. Methods We propose a multicentre, double-blinded, parallel, randomized, placebo-controlled clinical trial involving 4 study hospitals in China. A total of 72 eligible participants will be randomized into an SHP group and a placebo group. They will receive an SHP for 3 treatment sessions. The primary outcome will be changes in forced expiratory volume in 1 s after 3 treatment sessions. Secondary outcomes will include the following: (1) the Asthma Quality of Life Questionnaire, Asthma Control Test, and Asthma Long-term Follow-up Scale; (2) levels of Metallothionein-2 and Transgelin-2 in blood and urine; and (3) levels of IL-5, IL-13, IL-23, IL-25, and thymic stromal lymphopoietin in blood. Analysis of the data will be performed at baseline, at the end of the 2nd and 3rd treatment sessions, and at the 24-week follow-up. The safety of the SHP will be evaluated at each treatment session. Discussion The aims of this trial are to determine whether the SHP is more effective than placebo in the treatment of patients with bronchial asthma, as well as whether the SHP works by reducing airway inflammation and reversing bronchoconstriction. Trial registration Chinese Clinical Trial Registry (http://www.chictr.org.cn), ChiCTR1900024616. Registered on 19 July 2019.
Collapse
Affiliation(s)
- Xiaoyan Xie
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.,Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Danghan Xu
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Lixing Zhuang
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Hui Liu
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Sui Tan
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yanqing Lu
- The Fifth Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510095, China
| | - Meiyi Su
- The Fifth Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510095, China
| | - Jie Chen
- Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Haihua Pan
- Pingshan District Peoples' Hospital of Shenzhen, Shenzhen, 518118, China
| | - Lu Lu
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yiming Xu
- Guangzhou Medical University, Guangzhou, 515000, China
| | - Muxi Liao
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhanqiong Xu
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jun He
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
18
|
Venegas C, Zhao N, Ho T, Nair P. Sputum Inflammometry to Manage Chronic Obstructive Pulmonary Disease Exacerbations: Beyond Guidelines. Tuberc Respir Dis (Seoul) 2020; 83:175-184. [PMID: 32610835 PMCID: PMC7362747 DOI: 10.4046/trd.2020.0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 05/11/2020] [Indexed: 11/24/2022] Open
Abstract
Quantitative sputum cytometry facilitates in assessing the nature of bronchitis associated with exacerbations of chronic obstructive pulmonary disease (COPD). This is not assessed in most clinical trials that evaluate the effectiveness of strategies to prevent or to treat exacerbations. While up to a quarter of exacerbations may be associated with raised eosinophil numbers, the vast majority of exacerbations are associated with neutrophilic bronchitis that may indicate airway infections. While eosinophilia may be a predictor of response to corticosteroids (oral and inhaled), the limited efficacy of anti-interleukin 5 therapies would suggest that eosinophils may not directly contribute to those exacerbations. However, they may contribute to airspace enlargement in patients with COPD through various mechanisms involving the interleukin 13 and matrix metalloprotease pathways. The absence of eosinophils may facilitate in limiting the unnecessary use of corticosteroids. The presence of neutrophiia could prompt an investigation for the specific pathogens in the airway. Additionally, sputum measurements may also provide insight into the mechanisms of susceptibility to airway infections. Iron within sputum macrophages, identified by hemosiderin staining (and by more direct quantification) may impair macrophage functions while the low levels of immunoglobulins in sputum may also contribute to airway infections. The assessment of sputum at the time of exacerbations thus would facilitate in customizing treatment and treat current exacerbations and reduce future risk of exacerbations.
Collapse
Affiliation(s)
- Carmen Venegas
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Nan Zhao
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Terence Ho
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Parameswaran Nair
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare Hamilton and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
19
|
Lo Verso L, Matte JJ, Lapointe J, Talbot G, Bissonnette N, Blais M, Guay F, Lessard M. Impact of birth weight and neonatal nutritional interventions with micronutrients and bovine colostrum on the development of piglet immune response during the peri-weaning period. Vet Immunol Immunopathol 2020; 226:110072. [PMID: 32540688 DOI: 10.1016/j.vetimm.2020.110072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/19/2019] [Accepted: 05/17/2020] [Indexed: 10/24/2022]
Abstract
Immune system development of piglets is influenced by birth weight and colostrum and milk intake. Moreover, the dam transfer to piglets of vitamins A and D and copper, which play important role in immunity, is limited during lactation. In this study, we evaluated the potential of maternal and neonatal supplementations with vitamins A and D and copper, with or without neonatal supplementation of bovine colostrum (BC), to modulate the immune system development of low birth weight (LBW) and high birth weight (HBW) piglets during the peri-weaning period. Litters from 23 control sows (CONT) were assigned to one of the following treatments: 1) control (C); 2) oral administration at 2 and 8 days (d) of age of retinol-acetate, 25-hydroxyvitamin D and CuSO4 and exposure to UVB light for 15 min every second day from d 5 to d 21 (ADCu); 3) oral administration of dehydrated BC (4 g/d) from d 5 to d 10 (BC); 4) ADCu + BC. This experimental design was repeated with 24 sows fed extra daily supplements of 25-hydroxyvitamin D (4,000 IU), β-carotene (30,000 IU) and Cu-yeast (equivalent 45 mg of Cu) from 90 d of gestation until weaning at d 21 (SUPPL). Within each litter, 2 LBW and 2 HBW piglets were euthanized at d 16 and d 23 in order to characterize leukocyte subsets in mesenteric lymph nodes (MLN) and blood by flow cytometry, and to measure gene expression in the MLN and jejunal mucosa by qPCR. At d 16, results revealed that the percentages of γδ and cytotoxic T lymphocytes were significantly reduced in LBW compared to HBW piglets. The jejunal expression of interleukin (IL) 22 was also up-regulated, along with MLN expression of C-C Motif Chemokine Ligand 23, bone morphogenetic protein 2 and secreted phosphoprotein 1 (SPP1), whereas jejunal expression of tumor necrosis factor α was decreased in LBW piglets. At d 23, LBW piglets showed lower amounts of γδ T lymphocytes, higher percentages of CD3- and CD3-CD8α+CD16+ leukocytes (which include Natural killer cells) and lower jejunal expression of IL18. Furthermore, supplementation with BC increased the blood percentage of CD3-CD16+ leukocytes and reduced jejunal IL5 and MLN IL15 expression whereas supplementation with ADCu + BC increased jejunal TNF superfamily 13B and MLN SPP1 expression. Our results suggest that immune system development after birth differed between LBW and HBW piglets and that early dietary supplementation with BC and ADCu has the potential to modulate development of immune functions.
Collapse
Affiliation(s)
- Luca Lo Verso
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, 2000 College Street, Sherbrooke, QC, J1M 0C3 Canada; Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada.
| | - J Jacques Matte
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, 2000 College Street, Sherbrooke, QC, J1M 0C3 Canada
| | - Jérôme Lapointe
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, 2000 College Street, Sherbrooke, QC, J1M 0C3 Canada
| | - Guylaine Talbot
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, 2000 College Street, Sherbrooke, QC, J1M 0C3 Canada; Centre de recherche en infectiologie porcine et avicole (CRIPA-FQRNT), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, J2S 2M2 Canada
| | - Nathalie Bissonnette
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, 2000 College Street, Sherbrooke, QC, J1M 0C3 Canada
| | - Mylène Blais
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, 2000 College Street, Sherbrooke, QC, J1M 0C3 Canada; Centre de recherche en infectiologie porcine et avicole (CRIPA-FQRNT), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, J2S 2M2 Canada
| | - Frédéric Guay
- Faculté des Sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Université Laval, Québec, QC, G1V 0A6 Canada; Centre de recherche en infectiologie porcine et avicole (CRIPA-FQRNT), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, J2S 2M2 Canada
| | - Martin Lessard
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, 2000 College Street, Sherbrooke, QC, J1M 0C3 Canada; Centre de recherche en infectiologie porcine et avicole (CRIPA-FQRNT), Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, J2S 2M2 Canada
| |
Collapse
|
20
|
Jackson DJ, Korn S, Mathur SK, Barker P, Meka VG, Martin UJ, Zangrilli JG. Safety of Eosinophil-Depleting Therapy for Severe, Eosinophilic Asthma: Focus on Benralizumab. Drug Saf 2020; 43:409-425. [PMID: 32242310 PMCID: PMC7165132 DOI: 10.1007/s40264-020-00926-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Eosinophils play a pivotal role in the inflammatory pathology of asthma and have been the target of new biologic treatments for patients with eosinophilic asthma. Given the central role of interleukin (IL)-5 in the eosinophil lifecycle, several therapies directed against the IL-5 pathway have been developed, including the anti-IL-5 antibodies mepolizumab and reslizumab and the IL-5 receptor α (IL-5Rα)-directed cytolytic antibody benralizumab. Eosinophil-depleting therapies represent a relatively new class of asthma treatment, and it is important to understand their long-term efficacy and safety. Eosinophils have been associated with host protection and tumor growth, raising potential concerns about the consequences of long-term therapies that deplete eosinophils. However, evidence for these associations in humans is conflicting and largely indirect or based on mouse models. Substantial prospective clinical trial and postmarketing data have accrued, providing insight into the potential risks associated with eosinophil depletion. In this review, we explore the current safety profile of eosinophil-reducing therapies, with particular attention to the potential risks of malignancies and severe infections and a focus on benralizumab. Benralizumab is an IL-5Rα-directed cytolytic monoclonal antibody that targets and efficiently depletes blood and tissue eosinophils through antibody-dependent cell-mediated cytotoxicity. Benralizumab is intended to treat patients with severe, uncontrolled asthma with eosinophilic inflammation. The integrated analyses of benralizumab safety data from the phase III SIROCCO and CALIMA trials and subsequent BORA extension trial for patients with asthma, and the phase III GALATHEA and TERRANOVA trials for patients with chronic obstructive pulmonary disease, form the principal basis for this review.
Collapse
Affiliation(s)
- David J Jackson
- Guy's Severe Asthma Centre, Guy's & St Thomas' NHS Trust, London, UK
- Asthma UK Centre, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Stephanie Korn
- Universitätsmedizin Mainz, Langenbeckstr, Mainz, Germany
| | - Sameer K Mathur
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peter Barker
- Research and Development, AstraZeneca, Gaithersburg, MD, USA
| | | | - Ubaldo J Martin
- Research and Development, AstraZeneca, Gaithersburg, MD, USA
| | - James G Zangrilli
- Global Medical Affairs, BioPharmaceuticals Medical, AstraZeneca, One MedImmune Way, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
21
|
Licari A, Castagnoli R, De Filippo M, Foiadelli T, Tosca MA, Marseglia GL, Ciprandi G. Current and emerging biologic therapies for allergic rhinitis and chronic rhinosinusitis. Expert Opin Biol Ther 2020; 20:609-619. [PMID: 32053391 DOI: 10.1080/14712598.2020.1729350] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Allergic rhinitis and chronic rhinosinusitis, with and without nasal polyps, are the most common chronic inflammatory diseases of the upper airways. They both cause relevant respiratory symptoms and a substantial detriment to patients' quality of life, mainly in uncontrolled and severe patients.Areas covered: This review aims to present the most recent evidence on current and emerging biologic therapies for allergic rhinitis and chronic rhinosinusitis and discuss their potential implementation in clinical practice. To select relevant literature for inclusion in this review, we conducted a literature search using the PubMed database, using terms 'biologics OR biological agents', 'allergic rhinitis' and 'chronic rhinosinusitis'. The literature review was performed for publication years 2009-2019, restricting the articles to humans and English language publications.Expert opinion: Biological therapies represent a potential step forward in providing individualized care for all patients with uncontrolled severe upper airway diseases. Biologics recently showed promising results for the treatment of severe uncontrolled allergic rhinitis and chronic rhinosinusitis with nasal polyps with or without associated asthma. Endotyping inflammatory pathways and identifying related biomarkers remain the major challenge for positioning biologics in the care pathway of chronic respiratory diseases.
Collapse
Affiliation(s)
- Amelia Licari
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Riccardo Castagnoli
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Maria De Filippo
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Thomas Foiadelli
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | | | - Gian Luigi Marseglia
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | | |
Collapse
|
22
|
Hillas G, Fouka E, Papaioannou AI. Antibodies targeting the interleukin-5 signaling pathway used as add-on therapy for patients with severe eosinophilic asthma: a review of the mechanism of action, efficacy, and safety of the subcutaneously administered agents, mepolizumab and benralizumab. Expert Rev Respir Med 2020; 14:353-365. [PMID: 31958239 DOI: 10.1080/17476348.2020.1718495] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Since the discovery of eosinophils in the sputum of asthmatic patients, several studies have offered evidence on their prominent role in the pathology and severity of asthma. Blood eosinophils, are a useful biomarker for therapy selection in severe asthma patients. IL-5 plays crucial role on maturation, activation, recruitment, and survival of eosinophils and constitutes an important therapeutic target for patients with severe uncontrolled eosinophilic asthma.Areas covered: This review focuses on the similarities and differences on mechanisms of action, efficacy, and safety, of two subcutaneously(SC) administered agents, the anti-interleukin(IL)-5 monoclonal antibody mepolizumab and the IL-5 receptor-α(IL-5Rα)-directed cytolytic monoclonal antibody benralizumab. All information used was collected from PubMed using keywords such as severe asthma, eosinophils, IL-5, airway inflammation, asthma exacerbations, mepolizumab, benralizumab, anti-IL5, and anti-IL5R either as single terms or in several combinations.Expert opinion: Both mepolizumab and benralizumab are promising for the treatment of severe eosinophilic asthma resulting in asthma control improvement and exacerbations reduction and can serve as steroid-sparing agents. However, since no head-to-head comparisons exist, it is unknown whether their different mechanisms of action might be related to different efficacy in specific patients' sub-phenotypes. Long-term clinical observations will provide real-world evidence regarding their lasting effectiveness and safety.
Collapse
Affiliation(s)
- Georgios Hillas
- 5th Pulmonary Department, Sotiria Chest Diseases Hospital, Athens, Greece
| | - Evangelia Fouka
- Pulmonary Department of Aristotele University of Thessaloniki, G Papanikolaou Hospital, Thessaloniki, Greece
| | - Andriana I Papaioannou
- 2nd Respiratory Medicine Department, University of Athens, Attikon Hospital, Athens, Greece
| |
Collapse
|
23
|
Baseline blood eosinophil count as a predictor of treatment response to the licensed dose of mepolizumab in severe eosinophilic asthma. Respir Med 2019; 159:105806. [PMID: 31751853 DOI: 10.1016/j.rmed.2019.105806] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Previous analyses examining the relationship between blood eosinophil count and mepolizumab treatment effects in severe eosinophilic asthma have used a range of doses and administration routes. METHODS This post hoc meta-analysis included data from the MENSA (MEA115588/NCT01691521) and MUSCA (200862/NCT02281318) trials. Patients (≥12 years) with severe eosinophilic asthma who experienced ≥2 exacerbations in the prior year received either mepolizumab 100 mg subcutaneously (SC) or 75 mg intravenously, or placebo plus standard of care every 4 weeks. This meta-analysis reports data from patients receiving the licensed dose of mepolizumab (100 mg SC) or placebo only. The primary endpoint was the annual rate of clinically significant exacerbations; secondary endpoints included rate of exacerbations requiring hospitalization/emergency room (ER) visit, proportion of patients with no clinically significant exacerbations, and changes from baseline in forced expiratory volume in 1 s, Asthma Control Questionnaire-5 and St George's Respiratory Questionnaire scores. Analyses were stratified by baseline blood eosinophil count (<150, ≥150, ≥300, ≥400, ≥500, ≥750, ≥1000, ≥150-<300, or ≥300-<500 cells/μL). RESULTS Mepolizumab reduced annual clinically significant exacerbation rates by 45%-85%, exacerbations requiring hospitalization/ER visit by 60%-70%, and increased the odds of no clinically significant exacerbations across all eosinophil threshold subgroups versus placebo, and improved all other secondary endpoints in subgroups ≥150 cells/μL. Greater treatment effects with increasing blood eosinophil count were observed. CONCLUSIONS Mepolizumab demonstrated consistent clinical benefits in patients with baseline blood eosinophil counts ≥150 cells/μL, confirming the suitability of this cut-off for identifying patients responsive to the licensed mepolizumab dose.
Collapse
|
24
|
Takemura N, Kurashima Y, Mori Y, Okada K, Ogino T, Osawa H, Matsuno H, Aayam L, Kaneto S, Park EJ, Sato S, Matsunaga K, Tamura Y, Ouchi Y, Kumagai Y, Kobayashi D, Suzuki Y, Yoshioka Y, Nishimura J, Mori M, Ishii KJ, Rothenberg ME, Kiyono H, Akira S, Uematsu S. Eosinophil depletion suppresses radiation-induced small intestinal fibrosis. Sci Transl Med 2019; 10:10/429/eaan0333. [PMID: 29467297 DOI: 10.1126/scitranslmed.aan0333] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 10/05/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022]
Abstract
Radiation-induced intestinal fibrosis (RIF) is a serious complication after abdominal radiotherapy for pelvic tumor or peritoneal metastasis. Herein, we show that RIF is mediated by eosinophil interactions with α-smooth muscle actin-positive (α-SMA+) stromal cells. Abdominal irradiation caused RIF especially in the submucosa (SM) of the small intestine, which was associated with the excessive accumulation of eosinophils in both human and mouse. Eosinophil-deficient mice showed markedly ameliorated RIF, suggesting the importance of eosinophils. After abdominal irradiation, chronic crypt cell death caused elevation of extracellular adenosine triphosphate, which in turn activated expression of C-C motif chemokine 11 (CCL11) by pericryptal α-SMA+ cells in the SM to attract eosinophils in mice. Inhibition of C-C chemokine receptor 3 (CCR3) by genetic deficiency or neutralizing antibody (Ab) treatment suppressed eosinophil accumulation in the SM after irradiation in mice, suggesting a critical role of the CCL11/CCR3 axis in the eosinophil recruitment. Activated α-SMA+ cells also expressed granulocyte-macrophage colony-stimulating factor (GM-CSF) to activate eosinophils. Transforming growth factor-β1 from GM-CSF-stimulated eosinophils promoted collagen expression by α-SMA+ cells. In translational studies, treatment with a newly developed interleukin-5 receptor α-targeting Ab, analogous to the human agent benralizumab, depleted intestinal eosinophils and suppressed RIF in mice. Collectively, we identified eosinophils as a crucial factor in the pathogenesis of RIF and showed potential therapeutic strategies for RIF by targeting eosinophils.
Collapse
Affiliation(s)
- Naoki Takemura
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yosuke Kurashima
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Institute for Global Prominent Research, Chiba University, Chiba 260-8670, Japan.,Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yuki Mori
- Laboratory of Biofunctional Imaging, World Premier Institute (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kazuki Okada
- Immunology and Allergy R&D Unit, R&D Division, Kyowa Hakko Kirin Co. Ltd., 3-6-6 Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Takayuki Ogino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.,Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, Mainz 55131, Germany
| | - Hideki Osawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hirosih Matsuno
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Lamichhane Aayam
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Satoshi Kaneto
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Eun Jeong Park
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Department of Molecular Pathobiology and Cell Adhesion Biology, Basic Medical Sciences, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Shintaro Sato
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Mucosal Vaccine Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kouta Matsunaga
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yusuke Tamura
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yasuo Ouchi
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yutaro Kumagai
- Quantitative Immunology Research Unit, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Daichi Kobayashi
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Science, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Yoshichika Yoshioka
- Laboratory of Biofunctional Imaging, World Premier Institute (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Junichi Nishimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, 7-6-8 Asagi Saito, Ibaraki, Osaka 567-0085, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Mark E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan.,Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Satoshi Uematsu
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan. .,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
25
|
Chapman KR, Albers FC, Chipps B, Muñoz X, Devouassoux G, Bergna M, Galkin D, Azmi J, Mouneimne D, Price RG, Liu MC. The clinical benefit of mepolizumab replacing omalizumab in uncontrolled severe eosinophilic asthma. Allergy 2019; 74:1716-1726. [PMID: 31049972 PMCID: PMC6790683 DOI: 10.1111/all.13850] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/19/2019] [Accepted: 04/03/2019] [Indexed: 02/03/2023]
Abstract
Background Mepolizumab and omalizumab are treatments for distinct but overlapping severe asthma phenotypes. Objective To assess if patients eligible for both biologics but not optimally controlled with omalizumab experience improved asthma control when switched directly to mepolizumab. Methods OSMO was a multicenter, open‐label, single‐arm, 32‐week trial in patients with ≥2 asthma exacerbations in the year prior to enrollment, despite receiving high‐dose inhaled corticosteroids and other controller(s), plus omalizumab (≥4 months). At baseline, patients with blood eosinophil counts ≥150 cells/µL (or ≥300 cells/µL in the prior year) and an Asthma Control Questionnaire (ACQ)‐5 score ≥1.5 discontinued omalizumab and immediately commenced mepolizumab 100 mg subcutaneously every 4 weeks. Endpoints included change from baseline in ACQ‐5 score (primary), St George's Respiratory Questionnaire (SGRQ) score and the proportions of ACQ‐5 and SGRQ responders, all at Week 32, and the annualized exacerbation rate over the study period. Results At Week 32 (intent‐to‐treat population [n = 145]), the least squares (LS) mean changes (standard error [SE]) in ACQ‐5 and SGRQ total scores were −1.45 (0.107) and −19.0 (1.64) points; with 77% and 79% of patients achieving the minimum clinically important differences (ACQ‐5: ≥0.5 points; SGRQ: ≥4 points), respectively. The annualized rate of clinically significant exacerbations was 1.18 events/year, a 64% reduction from 3.26 events/year during the previous year. Safety and immunogenicity profiles were consistent with previous trials. Conclusion After directly switching from omalizumab to mepolizumab, patients with uncontrolled severe eosinophilic asthma experienced clinically significant improvements in asthma control, health status, and exacerbation rate, with no tolerability issues reported.
Collapse
Affiliation(s)
- Kenneth R. Chapman
- Asthma and Airway Centre University Health Network, University of Toronto Toronto Ontario Canada
| | - Frank C. Albers
- Global Respiratory Medical Franchise GSK Research Triangle Park North Carolina
| | - Bradley Chipps
- Capital Allergy and Respiratory Disease Center Sacramento California
| | - Xavier Muñoz
- Pulmonology Department Hospital Universitari Vall d'Hebron Barcelona Spain
- Ciber Enfermedades Respiratorias Madrid Spain
| | - Gilles Devouassoux
- Service de Pneumologie, Hôpital de la Croix Rousse, Hospices Civils de Lyon UCB Lyon 1 Lyon France
| | - Miguel Bergna
- Respiratory Research CEMER, Vicente Lopez Buenos Aires Argentina
| | - Dmitry Galkin
- Global Respiratory Medical Franchise GSK Research Triangle Park North Carolina
| | | | | | | | - Mark C. Liu
- Divisions of Allergy and Clinical Immunology, Pulmonary and Critical Care Medicine Johns Hopkins Asthma and Allergy Center Baltimore Maryland
| |
Collapse
|
26
|
Effects of Air Pollution on Lung Innate Lymphoid Cells: Review of In Vitro and In Vivo Experimental Studies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16132347. [PMID: 31269777 PMCID: PMC6650824 DOI: 10.3390/ijerph16132347] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/28/2019] [Accepted: 06/29/2019] [Indexed: 12/21/2022]
Abstract
Outdoor air pollution is associated with respiratory infections and allergies, yet the role of innate lymphoid cells (ILCs) in pathogen containment and airway hyperresponsiveness relevant to effects of air pollutants on ILCs is poorly understood. We conducted a systematic review to evaluate the available evidence on the effect of outdoor air pollutants on the lung type 1 (ILC1) and type 2 ILCs (ILC2) subsets. We searched five electronic databases (up to Dec 2018) for studies on the effect of carbon monoxide (CO), sulfur dioxide (SO2), nitrogen dioxide (NO2), diesel exhaust particles (DEP), ozone (O3), and particulate matter (PM) on respiratory ILCs. Of 2209 identified citations, 22 full-text papers were assessed for eligibility, and 12 articles describing experimental studies performed in murine strains (9) and on human blood cells (3) were finally selected. Overall, these studies showed that exposure to PM, DEP, and high doses of O3 resulted in a reduction of interferon gamma (IFN-γ) production and cytotoxicity of ILC1. These pollutants and carbon nanotubes stimulate lung ILC2s, produce high levels of interleukin (IL)-5 and IL-13, and induce airway hyperresponsiveness. These findings highlight potential mechanisms by which human ILCs react to air pollution that increase the susceptibility to infections and allergies.
Collapse
|
27
|
Crowe M, Robinson D, Sagar M, Chen L, Ghamande S. Chronic eosinophilic pneumonia: clinical perspectives. Ther Clin Risk Manag 2019; 15:397-403. [PMID: 30936702 PMCID: PMC6420789 DOI: 10.2147/tcrm.s157882] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic eosinophilic pneumonia (CEP) is an eosinophilic lung disease that is typically diagnosed by a triad of clinical symptoms including pulmonary symptoms, eosinophilia and characteristic radiographic abnormalities. It requires a high index of suspicion given its overlap with other eosinophilic conditions and lack of a specific diagnostic test. The diagnosis is made after careful consideration of other secondary causes of eosinophilia, such as infectious, drugs, or toxic etiologies. CEP generally responds rapidly to treatment, which primarily consists of corticosteroid therapy, but relapses are common. Novel therapies are being explored as more information is being discovered about the pathophysiology of eosinophilic disease processes. Close follow-up is important given the difficulty in weaning patients from glucocorticoids with many patients developing sequelae of chronic glucocorticoid therapy. Therefore, exploring alternative treatments is of upmost importance.
Collapse
Affiliation(s)
- Matthew Crowe
- Department of Medicine, Division of Pulmonary and Critical Care, Baylor Scott and White Medical Center, Temple, TX, USA,
| | - Drew Robinson
- Department of Medicine, Baylor Scott and White Medical Center, Temple, TX, USA
| | - Malvika Sagar
- Department of Pediatrics, Baylor Scott and White McLane Children's Specialty Clinic Temple, Temple, TX, USA
| | - Li Chen
- Department of Pathology, Baylor Scott and White Medical Center Temple, Temple, TX, USA
| | - Shekhar Ghamande
- Department of Medicine, Division of Pulmonary and Critical Care, Baylor Scott and White Medical Center, Temple, TX, USA,
| |
Collapse
|
28
|
Steinfeld J, Bradford ES, Brown J, Mallett S, Yancey SW, Akuthota P, Cid MC, Gleich GJ, Jayne D, Khoury P, Langford CA, Merkel PA, Moosig F, Specks U, Weller PF, Wechsler ME. Evaluation of clinical benefit from treatment with mepolizumab for patients with eosinophilic granulomatosis with polyangiitis. J Allergy Clin Immunol 2018; 143:2170-2177. [PMID: 30578883 DOI: 10.1016/j.jaci.2018.11.041] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/02/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND In a recent phase III trial (NCT02020889) 53% of mepolizumab-treated versus 19% of placebo-treated patients with eosinophilic granulomatosis with polyangiitis (EGPA) achieved protocol-defined remission. OBJECTIVE We sought to investigate post hoc the clinical benefit of mepolizumab in patients with EGPA using a comprehensive definition of benefit encompassing remission, oral glucocorticoid (OGC) dose reduction, and EGPA relapses. METHODS The randomized, placebo-controlled, double-blind, parallel-group trial recruited patients with relapsing/refractory EGPA receiving stable OGCs (prednisolone/prednisone, ≥7.5-50 mg/d) for 4 or more weeks. Patients received 300 mg of subcutaneous mepolizumab or placebo every 4 weeks for 52 weeks. Clinical benefit was defined post hoc as follows: remission at any time (2 definitions used), 50% or greater OGC dose reduction during weeks 48 to 52, or no EGPA relapses. The 2 remission definitions were Birmingham Vasculitis Activity Score of 0 plus OGC dose of 4 mg/d or less (remission 1/clinical benefit 1) or 7.5 mg/d or less (remission 2/clinical benefit 2). Clinical benefit was assessed in all patients and among subgroups with a baseline blood eosinophil count of less than 150 cells/μL, baseline OGC dosage of greater than 20 mg/d, or weight of greater than 85 kg. RESULTS With mepolizumab versus placebo, 78% versus 32% of patients experienced clinical benefit 1, and 87% versus 53% of patients experienced clinical benefit 2 (both P < .001). Significantly more patients experienced clinical benefit 1 with mepolizumab versus placebo in the blood eosinophil count less than 150 cells/μL subgroup (72% vs 43%, P = .033) and weight greater than 85 kg subgroup (68% vs 23%, P = .005); in the OGC greater than 20 mg/d subgroup, results were not significant but favored mepolizumab (60% vs 36%, P = .395). CONCLUSION When a comprehensive definition of clinical benefit was applied to data from a randomized controlled trial, 78% to 87% of patients with EGPA experienced benefit with mepolizumab.
Collapse
Affiliation(s)
- Jonathan Steinfeld
- Respiratory TAU & Flexible Discovery Unit, GlaxoSmithKline, Philadelphia, Pa
| | - Eric S Bradford
- Respiratory Therapeutic Area, GlaxoSmithKline, Research Triangle Park, NC
| | - Judith Brown
- Research and Development, Immuno-Inflammation TAU, Uxbridge, United Kingdom
| | - Stephen Mallett
- Research & Development, Statistics, Programming and Data Standards, GlaxoSmithKline, Stockley Park West, Uxbridge, United Kingdom
| | - Steven W Yancey
- Respiratory Therapeutic Area, GlaxoSmithKline, Research Triangle Park, NC
| | - Praveen Akuthota
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, Calif
| | - Maria C Cid
- Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Gerald J Gleich
- Departments of Dermatology and Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paneez Khoury
- Human Eosinophil Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Carol A Langford
- Department of Rheumatic and Immunologic Diseases, Center for Vasculitis Care and Research, Cleveland Clinic, Cleveland, Ohio
| | - Peter A Merkel
- Division of Rheumatology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pa
| | - Frank Moosig
- Rheumazentrum, Schleswig-Holstein Mitte, Neumünster, Germany
| | - Ulrich Specks
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minn
| | - Peter F Weller
- Divisions of Allergy and Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass
| | | |
Collapse
|
29
|
Bagnasco D, Caminati M, Ferrando M, Aloè T, Testino E, Canonica GW, Passalacqua G. Anti-IL-5 and IL-5Ra: Efficacy and Safety of New Therapeutic Strategies in Severe Uncontrolled Asthma. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5698212. [PMID: 30519580 PMCID: PMC6241368 DOI: 10.1155/2018/5698212] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 09/25/2018] [Indexed: 12/25/2022]
Abstract
The current developments of the new biological drugs targeting interleukin 5 (IL-5) and IL-5 receptor allowed to expand the treatment options for severe hypereosinophilic asthma. Clinicians will then be able to choose between antibodies targeting either circulating IL-5 or its receptor expressed on eosinophils and basophils. The available clinical trials consistently reported favorable results about the reduction of exacerbations rate, improvement in quality of life, and sparing of the systemic steroid use, with a favorable safety profile. Two of these new drugs are administered subcutaneously, mepolizumab every 4 weeks and benralizumab every 8 weeks, whereas reslizumab is given intravenously monthly on a weigh-based dose. In the future, the research actions will be involved in the identification of a single biomarker or multiple biomarkers for the optimal choice of biological agents to be properly prescribed.
Collapse
Affiliation(s)
- Diego Bagnasco
- Allergy & Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Marco Caminati
- Asthma Center and Allergy Unit, Verona University and General Hospital, Verona, Italy
| | - Matteo Ferrando
- Allergy & Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Teresita Aloè
- Allergy & Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Elisa Testino
- Allergy & Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Giorgio Walter Canonica
- Allergy & Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
- Department of Internal Medicine, Respiratory Disease Clinic, IRCCS, Humanitas Clinical and Research Center, Humanitas University, Rozzano, Milan, Italy
| | - Giovanni Passalacqua
- Allergy & Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| |
Collapse
|
30
|
Eosinophils Target Therapy for Severe Asthma: Critical Points. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7582057. [PMID: 30498762 PMCID: PMC6222236 DOI: 10.1155/2018/7582057] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/24/2018] [Accepted: 10/17/2018] [Indexed: 01/27/2023]
Abstract
Asthma is a chronic and heterogeneous disease, which is defined as severe disease whenever it requires treatment with a high dose of inhaled corticosteroids plus a second controller and/or systemic corticosteroids to prevent it from becoming ‘‘uncontrolled” or if it remains ‘‘uncontrolled” despite this therapy. Severe asthma is a heterogeneous condition consisting of phenotypes such as eosinophilic asthma, which is characterized by sputum eosinophilia, associated with mild to moderate increase in blood eosinophil count, frequently adult-onset, and associated with chronic rhinosinusitis with nasal polyps in half of the cases. Eosinophilic asthma is driven by T2 inflammation, characterized, among the others, by interleukin-5 production. IL-5 plays a key role in the differentiation, survival, migration, and activation of eosinophils, and it has become an appealing therapeutic target for eosinophilic asthma. In recent years two monoclonal antibodies (mepolizumab and reslizumab) directed against IL-5 and one monoclonal antibody directed against the alpha-subunit of the IL-5 receptor (benralizumab) have been developed. All these IL-5 target drugs have been shown to reduce the number of exacerbation in patients with severe asthma selected on the basis of peripheral blood eosinophil count. There are still a number of unresolved issues related to the anti-IL5 strategy in eosinophilic asthma, which are here reviewed. These issues include the effects of such therapy on airway obstruction and asthmatic symptoms, the level of baseline eosinophils that predicts a response to treatment, the relationship between blood and airway eosinophilia, and, perhaps most importantly, how to elucidate the pathogenetic role played by eosinophils in the individual patient with severe eosinophilic asthma.
Collapse
|
31
|
Airway Eosinophilopoietic and Autoimmune Mechanisms of Eosinophilia in Severe Asthma. Immunol Allergy Clin North Am 2018; 38:639-654. [PMID: 30342585 DOI: 10.1016/j.iac.2018.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Eosinophils are critical in asthma biology, contributing to symptoms, airflow obstruction, airway hyperresponsiveness, and remodeling. In severe asthma, in addition to local maturation in bone marrow, in situ eosinophilopoiesis plays a key role in the persistence of airway eosinophilia. Local milieu of structural, epithelial and inflammatory cells contribute by generating eosinophilopoietic cytokines in response to epithelial-derived alarmins. Another mechanism of persistent airway eosinophilia is glucocorticosteroid insensitivity, which is linked to recurrent airway infections and presence of local autoantibodies. Novel molecules are being developed to target specific immune pathways as potential steroid-sparing strategies.
Collapse
|
32
|
Allen J, Wert M. Eosinophilic Pneumonias. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 6:1455-1461. [DOI: 10.1016/j.jaip.2018.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/15/2018] [Accepted: 03/30/2018] [Indexed: 10/17/2022]
|
33
|
Li Y, Chen Q, Ji W, Ding Y, Zhou W. HMGB1 Binding to the RAGE Receptor Contributes to the Pathogenesis of Asthma. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2018. [DOI: 10.1089/ped.2017.0846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Yuqin Li
- Department of Infectious Diseases, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Qiu Chen
- School of Radiation Medicine and Protection, Soochow University, Suzhou, People's Republic of China
| | - Wei Ji
- Department of Infectious Diseases, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ying Ding
- Department of Infectious Diseases, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Weifang Zhou
- Department of Infectious Diseases, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
34
|
Patel SS, Casale TB, Cardet JC. Biological therapies for eosinophilic asthma. Expert Opin Biol Ther 2018; 18:747-754. [PMID: 29938543 PMCID: PMC6317519 DOI: 10.1080/14712598.2018.1492540] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/20/2018] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Severe uncontrolled asthma is by definition refractory to traditional therapies or can be controlled only with therapies that have intolerable side effects. Monoclonal antibodies that target interleukin (IL)-5/IL-5Rα, IgE, and IL-4Rα have shown favorable results in clinical trials, including reductions in asthma exacerbations and other important clinical outcomes. These biological agents offer treatment alternatives to patients with uncontrolled severe eosinophilic asthma. AREAS COVERED This article reviews how the shifting emphasis toward identifying distinct asthma phenotypes has led to the approval of biological therapies that preferentially benefit patients with severe eosinophilic asthma. The clinical trials that led to the approval of these biologic treatments are discussed in detail. EXPERT OPINION Biologic therapies targeting the IL-5, IgE, IL-4/IL-13 signaling pathways have been successful in clinical trials in subjects with severe eosinophilic asthma. Some of these agents have also been successful regardless of peripheral blood eosinophil counts. These treatments have shown a relatively favorable safety profile in clinical trials, although long-term safety data for some of these agents are limited. Due to the high costs associated with these medications, they should be reserved for select patients where they yield a therapeutic and pharmacoeconomic advantage.
Collapse
Affiliation(s)
- Shiven S Patel
- a Division of Allergy and Immunology, Department of Internal Medicine , University of South Florida Morsani College of Medicine and James A. Haley Veterans' Affairs Hospital , Tampa , FL , USA
| | - Thomas B Casale
- a Division of Allergy and Immunology, Department of Internal Medicine , University of South Florida Morsani College of Medicine and James A. Haley Veterans' Affairs Hospital , Tampa , FL , USA
| | - Juan Carlos Cardet
- a Division of Allergy and Immunology, Department of Internal Medicine , University of South Florida Morsani College of Medicine and James A. Haley Veterans' Affairs Hospital , Tampa , FL , USA
| |
Collapse
|
35
|
Zhenjiang L, Rao M, Luo X, Valentini D, von Landenberg A, Meng Q, Sinclair G, Hoffmann N, Karbach J, Altmannsberger HM, Jäger E, Peredo IH, Dodoo E, Maeurer M. Cytokine Networks and Survivin Peptide-Specific Cellular Immune Responses Predict Improved Survival in Patients With Glioblastoma Multiforme. EBioMedicine 2018; 33:49-56. [PMID: 30049387 PMCID: PMC6085502 DOI: 10.1016/j.ebiom.2018.06.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 06/03/2018] [Accepted: 06/12/2018] [Indexed: 01/08/2023] Open
Abstract
PURPOSE We investigated serum cytokine and T-cell responses directed against tumour-associated antigens (TAAs) in association with survival of patients with glioblastoma multiforme (GBM). PATIENTS AND METHODS Peripheral blood from 205 treatment-naïve patients with glioma (GBM = 145; non-GBM = 60) was obtained on the day of surgery to measure (i) circulating T-cells reacting to viral antigens and TAAs, in the presence or absence of cytokine conditioning with IL-2/IL-15/IL-21 or IL-2/IL-7, and (ii) serum cytokine levels (IL-4, IL-5, IL-6, TNF-α, IFN-γ and IL-17A). Patients were followed-up for at least 1000 days post-surgery. Survivin protein and gene expression in resected GBM tumour tissue were confirmed by immunohistochemistry and real-time polymerase chain reaction, respectively. Antigen-specific T-cell responses were gauged by ICS (intracellular cytokine production). Associations between patient survival and immunological reactivity patterns were analysed using univariate and multivariate statistics. RESULTS Approximately 2% of patients with GBM and 18% of patients with non-GBM glioma, were alive beyond 1000 days of surgery. Univariate analysis indicated that the combination of three cytokines (IL-4/IL-5/IL-6, p = .0022; IFN-γ/TNF-α/IL-17A, p = .0083) but not a 'partial' combination of these cytokines, the IFN-γ immune response to EBV-EBNA-1 (p < .0001) as well as T-cell responses to the survivin97-111 peptide (p = .0152) correlated with longer survival among patients with GBM. Multivariate analysis identified survivin97-111-directed IFN-γ production with IL-2/IL-15/IL-21 conditioning (p = .024), and the combined presence of serum IFN-γ/TNF-α/IL-17a (p = .003) as independent predictors of survival. CONCLUSION Serum cytokine patterns and lymphocyte reactivity to survivin97-111, particularly with IL-2, IL-15 and IL-21 conditioning may be instrumental in predicting survival among patients with GBM. This has implications for clinical follow-up of patients with GBM and the targeted development of immunotherapy for patients with CNS tumours.
Collapse
Affiliation(s)
- Liu Zhenjiang
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Martin Rao
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xiaohua Luo
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Davide Valentini
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for allogeneic stem cell transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Anna von Landenberg
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Qingda Meng
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Georges Sinclair
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Stockholm, Sweden
| | - Nina Hoffmann
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Julia Karbach
- Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt/Main, Germany
| | | | - Elke Jäger
- Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt/Main, Germany
| | - Inti Harvey Peredo
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Stockholm, Sweden
| | - Ernest Dodoo
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Stockholm, Sweden
| | - Markus Maeurer
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for allogeneic stem cell transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
36
|
Fettelschoss-Gabriel A, Fettelschoss V, Thoms F, Giese C, Daniel M, Olomski F, Kamarachev J, Birkmann K, Bühler M, Kummer M, Zeltins A, Marti E, Kündig TM, Bachmann MF. Treating insect-bite hypersensitivity in horses with active vaccination against IL-5. J Allergy Clin Immunol 2018; 142:1194-1205.e3. [PMID: 29627082 DOI: 10.1016/j.jaci.2018.01.041] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/17/2018] [Accepted: 01/29/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Insect-bite hypersensitivity is the most common allergic dermatitis in horses. Excoriated skin lesions are typical symptoms of this seasonal and refractory chronic disease. On a cellular level, the skin lesions are characterized by massive eosinophil infiltration caused by an underlying allergic response. OBJECTIVE To target these cells and treat disease, we developed a therapeutic vaccine against equine IL-5 (eIL-5), the master regulator of eosinophils. METHODS The vaccine consisted of eIL-5 covalently linked to a virus-like particle derived from cucumber mosaic virus containing the tetanus toxoid universal T-cell epitope tt830-843 (CMVTT). Thirty-four Icelandic horses were recruited and immunized with 400 μg of eIL-5-CMVTT formulated in PBS without adjuvant (19 horses) or PBS alone (15 horses). RESULTS The vaccine was well tolerated and did not reveal any safety concerns but was able to induce anti-eIL-5 autoantibody titers in 17 of 19 horses. This resulted in a statistically significant reduction in clinical lesion scores when compared with previous season levels, as well as levels in placebo-treated horses. Protection required a minimal threshold of anti-eIL-5 antibodies. Clinical improvement by disease scoring showed that 47% and 21% of vaccinated horses reached 50% and 75% improvement, respectively. In the placebo group no horse reached 75% improvement, and only 13% reached 50% improvement. CONCLUSION Our therapeutic vaccine inducing autoantibodies against self IL-5 brings biologics to horses, is the first successful immunotherapeutic approach targeting a chronic disease in horses, and might facilitate development of a similar vaccine against IL-5 in human subjects.
Collapse
Affiliation(s)
- Antonia Fettelschoss-Gabriel
- Department of Dermatology, University Hospital Zurich, Schlieren, Switzerland; Evax AG, Münchwilen, Switzerland.
| | - Victoria Fettelschoss
- Department of Dermatology, University Hospital Zurich, Schlieren, Switzerland; Evax AG, Münchwilen, Switzerland
| | - Franziska Thoms
- Department of Dermatology, University Hospital Zurich, Schlieren, Switzerland
| | - Christoph Giese
- ETH Zurich, Institute of Molecular Biology & Biophysics, Zurich, Switzerland
| | - Michelle Daniel
- Department of Dermatology, University Hospital Zurich, Schlieren, Switzerland
| | - Florian Olomski
- Department of Dermatology, University Hospital Zurich, Schlieren, Switzerland
| | - Jivko Kamarachev
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | - Andris Zeltins
- Latvian Biomedical Research & Study Centre, Riga, Latvia
| | - Eliane Marti
- Department for Clinical Research VPH, Vetsuisse Faculty of the University of Bern, Clinical Immunology Group, Bern, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Martin F Bachmann
- RIA Immunology, Inselspital, University of Bern, Bern, Switzerland; Jenner Institute, Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Glucocorticosteroids (GCSs) remain the cornerstone of therapy for treating the inflammatory component of asthma. Clinical response to GCS is heterogeneous, varying both within asthma 'endotypes', as well as the same individual. Different factors and micro-environment can alter the canonical GCS-induced signalling pathways leading to reduced efficacy, collectively termed as GCS subsensitivity, which includes the entire spectrum of steroid insensitivity and steroid resistance. RECENT FINDINGS In the past, steroid subsensitivity has been associated with dysregulated expression of glucocorticoid-receptor isoforms, neutrophilic inflammation and Th17 cytokines, oxidative stress-inducing factors and their downstream effect on histone deacetylase activities and gene expression. The review highlights recent observations, such as GCS-induced dysregulation of key transcription factors involved in host defence, role of airway infections altering expression of critical regulatory elements like the noncoding microRNAs, and the importance of interleukin (IL)-10 in reinstating steroid response in key immune cells. Further, emerging concepts of autoimmunity triggered because of delayed resolution of eosinophilic inflammation (due to GCS subsensitivity) and observed lymphopenia (plausibly a side-effect of continued GCS use) are discussed. SUMMARY This review bridges concepts that have been known, and those under current investigation, providing both molecular and clinical insights to aid therapeutic strategies for optimal management of asthmatics with varying degree of steroid subsensitivity and disease severity, with particular emphasis on the PI3 kinase pathways.
Collapse
|
38
|
Bagnasco D, Ferrando M, Caminati M, Bragantini A, Puggioni F, Varricchi G, Passalacqua G, Canonica GW. Targeting Interleukin-5 or Interleukin-5Rα: Safety Considerations. Drug Saf 2018; 40:559-570. [PMID: 28321782 DOI: 10.1007/s40264-017-0522-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Asthma is a highly prevalent chronic disease of the airways; approximately 10% of patients with asthma will experience a severe form of the disease. New understanding of the pathogenesis of asthma has enabled the development of novel drugs and provided hope for patients with asthma. Interleukin (IL)-5 and IL-5 receptor subunit α (IL-5-Rα) plays a crucial role in the development, maturation, and operation of eosinophils so were the first important therapeutic target of these new drugs. While the results of early clinical trials of these drugs were not promising, results improved once researchers discovered the drugs worked best in patients with high eosinophil levels. Patients treated with both anti-IL-5 and IL-5-Rα experienced significant decreases in exacerbations. Trials have also demonstrated promising safety profiles; adverse events have been few and frequently only observed with placebo or considered unrelated to the study drug. The positive efficacy and safety profiles of these drugs has led to trials with interesting results in other diseases that are also secondary to the action of eosinophils: Churg-Strauss syndrome, hypereosinophilic syndrome, nasal polyposis, chronic obstructive pulmonary disease, atopic dermatitis, and esophagitis. In this review, we explore the main clinical trials of anti-IL-5 and IL-5-Rα, both in asthma and in other pathologies, with particular reference to the interesting safety and efficacy results.
Collapse
Affiliation(s)
- Diego Bagnasco
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Matteo Ferrando
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Marco Caminati
- Allergy Unit, University Hospital of Verona, Verona, Italy
| | - Alice Bragantini
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Francesca Puggioni
- Respiratory Disease Clinic, Department of Internal Medicine, IRCCS Humanitas Clinical and Research Center, Humanitas University, Milan, Italy
| | - Gilda Varricchi
- Division of Clinical Immunology and Allergy, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Giovanni Passalacqua
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Giorgio Walter Canonica
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy.
- Respiratory Disease Clinic, Department of Internal Medicine, IRCCS Humanitas Clinical and Research Center, Humanitas University, Milan, Italy.
| |
Collapse
|
39
|
Salter BMA, Smith SG, Mukherjee M, Plante S, Krisna S, Nusca G, Oliveria JP, Irshad A, Gauvreau GM, Chakir J, Nair P, Sehmi R. Human Bronchial Epithelial Cell-derived Factors from Severe Asthmatic Subjects Stimulate Eosinophil Differentiation. Am J Respir Cell Mol Biol 2018; 58:99-106. [PMID: 28853918 DOI: 10.1165/rcmb.2016-0262oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Activated bronchial epithelial cells (BEC) release various alarmins, including thymic stromal lymphopoietin (TSLP), that drive type 2 inflammation. We hypothesize that BEC-derived factors promote in situ eosinophil differentiation and maturation, a process that is driven by an IL-5-rich microenvironment in asthmatic airways. To assess the eosinophilopoietic potential of epithelial-derived factors, eosinophil/basophil colony forming units (Eo/B-CFU) were enumerated in 14-day methylcellulose cultures of blood-derived nonadherent mononuclear cells incubated with BEC supernatants (BECSN) from healthy nonatopic controls (n = 8), mild atopic asthmatics (n = 9), and severe asthmatics (n = 5). Receptor-blocking antibodies were used to evaluate the contribution of alarmins. Modulation of the mRNA expression of transcription factors that are crucial for eosinophil differentiation was evaluated. BECSN stimulated the clonogenic expansion of eosinophil progenitors in vitro. In the presence of IL-5, Eo/B-CFU numbers were significantly greater in cocultures of BESCN from severe asthmatics compared with other groups. This was attenuated in the presence of a TSLP (but not an IL-33) receptor-blocking antibody. Recombinant human TSLP (optimal at 100 pg/ml) stimulated Eo/B-CFU growth, which was significantly enhanced in the presence of IL-5 (1 ng/ml). Overnight culture of CD34+ cells with IL-5 and TSLP synergistically increased GATA-binding factor 2 and CCAAT/enhancer-binding protein α mRNA expression. The eosinophilopoietic potential of factors derived from BEC is increased in severe asthma. Our data suggest that TSLP is a key alarmin that is produced by BECs and promotes in situ eosinophilopoiesis in a type 2-rich microenvironment.
Collapse
Affiliation(s)
- Brittany M A Salter
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Steven G Smith
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Manali Mukherjee
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Sophie Plante
- 2 Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Sakktee Krisna
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Graeme Nusca
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - John Paul Oliveria
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Anam Irshad
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Gail M Gauvreau
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Jamila Chakir
- 2 Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Parameswaran Nair
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Roma Sehmi
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| |
Collapse
|
40
|
Sehmi R, Smith SG, Kjarsgaard M, Radford K, Boulet LP, Lemiere C, Prazma CM, Ortega H, Martin JG, Nair P. Role of local eosinophilopoietic processes in the development of airway eosinophilia in prednisone-dependent severe asthma. Clin Exp Allergy 2017; 46:793-802. [PMID: 26685004 DOI: 10.1111/cea.12695] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/07/2015] [Accepted: 12/13/2015] [Indexed: 11/27/2022]
Abstract
BACKGROUND In severe asthmatics with persistent airway eosinophilia, blockade of interleukin-5 has significant steroid-sparing effects and attenuates blood and sputum eosinophilia. The contribution of local maturational processes of progenitors within the airways relative to the recruitment of mature cells from the peripheral circulation to the development of airway eosinophilia is not known. We hypothesize that local eosinophilopoiesis may be the predominant process that drives persistent airway eosinophilia and corticosteroid requirement in severe asthmatics. OBJECTIVES In a cross-sectional study, the number and growth potential of eosinophil-lineage-committed progenitors (EoP) were assayed in 21 severe eosinophilic asthmatics, 19 mild asthmatics, eight COPD patients and eight normal subjects. The effect of anti-IL-5 treatment on mature eosinophils and EoP numbers was made in severe eosinophilic asthmatics who participated in a randomized clinical trial of mepolizumab (substudy of a larger GSK sponsored global phase III trial, MEA115575) where subjects received mepolizumab (100 mg, n = 9) or placebo (n = 8), as six monthly subcutaneous injections. RESULTS Mature eosinophil and EoP numbers were significantly greater in the sputum of severe asthmatics compared with all other subject groups. In colony-forming assays, EoP from blood of severe asthmatics demonstrated a greater response to IL-5 than mild asthmatics. Treatment of severe asthmatics with mepolizumab significantly attenuated blood eosinophils and increased EoP numbers consistent with blockade of systemic eosinophilopoiesis. There was however no significant treatment effect on mature eosinophils, sputum EoP numbers or the prednisone maintenance dose. CONCLUSIONS AND CLINICAL RELEVANCE Patients with severe eosinophilic asthma have an exaggerated eosinophilopoeitic process in their airways. Treatment with 100 mg subcutaneous mepolizumab significantly attenuated systemic differentiation of eosinophils, but did not suppress local airway eosinophil differentiation to mature cells. Targeting IL-5-driven eosinophil differentiation locally within the lung maybe of relevance for optimal control of airway eosinophilia and asthma.
Collapse
Affiliation(s)
- R Sehmi
- Department of Medicine, McMaster University and Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - S G Smith
- Department of Medicine, McMaster University and Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - M Kjarsgaard
- Department of Medicine, McMaster University and Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - K Radford
- Department of Medicine, McMaster University and Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - L-P Boulet
- Institut Universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
| | - C Lemiere
- Division of Pneumologie, University of Montreal, Montreal, QC, Canada
| | - C M Prazma
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | - H Ortega
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | - J G Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - P Nair
- Department of Medicine, McMaster University and Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| |
Collapse
|
41
|
Hwang YH, Hong SG, Mun SK, Kim SJ, Lee SJ, Kim JJ, Kang KY, Yee ST. The Protective Effects of Astaxanthin on the OVA-Induced Asthma Mice Model. Molecules 2017; 22:molecules22112019. [PMID: 29160801 PMCID: PMC6150233 DOI: 10.3390/molecules22112019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/17/2017] [Indexed: 01/10/2023] Open
Abstract
Although astaxanthin has a variety of biological activities such as anti-oxidant effects, inhibitory effects on skin deterioration and anti-inflammatory effects, its effect on asthma has not been studied. In this paper, the inhibitory effect of astaxanthin on airway inflammation in a mouse model of ovalbumin (OVA)-induced asthma was investigated. We evaluated the number of total cells, Th1/2 mediated inflammatory cytokines in bronchoalveolar lavage fluid (BALF) and airway hyperresponsiveness as well as histological structure. The level of total IgE, IgG1, IgG2a, OVA-specific IgG1, and OVA-specific IgG2a were also examined. The oral administration of 50 mg/mL astaxanthin inhibited the respiratory system resistance, elastance, newtonian resistance, tissue damping, and tissue elastance. Also, astaxanthin suppressed the total cell number, IL-4, and IL-5, and increased the IFN-γ in the BALF. In the sera, total IgE, IgG1, and OVA-specific IgG1 were reduced by astaxanthin exposure and IgG2a and OVA-specific IgG2a were enhanced via oral administration of astaxanthin. Infiltration of inflammatory cells in the lung, production of mucus, lung fibrosis, and expression of caspase-1 or caspase-3 were suppressed in OVA-induced asthmatic animal treated with astaxanthin. These results suggest that astaxanthin may have therapeutic potential for treating asthma via inhibiting Th2-mediated cytokine and enhancing Th1-mediated cytokine.
Collapse
Affiliation(s)
- Yun-Ho Hwang
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon 540-950, Korea; (Y.-H.H.); (S.-G.H.); (S.-K.M.); (S.-J.K.); (S.-J.L.)
| | - Seong-Gyeol Hong
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon 540-950, Korea; (Y.-H.H.); (S.-G.H.); (S.-K.M.); (S.-J.K.); (S.-J.L.)
| | - Seul-Ki Mun
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon 540-950, Korea; (Y.-H.H.); (S.-G.H.); (S.-K.M.); (S.-J.K.); (S.-J.L.)
| | - Su-Jin Kim
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon 540-950, Korea; (Y.-H.H.); (S.-G.H.); (S.-K.M.); (S.-J.K.); (S.-J.L.)
| | - Sung-Ju Lee
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon 540-950, Korea; (Y.-H.H.); (S.-G.H.); (S.-K.M.); (S.-J.K.); (S.-J.L.)
| | - Jong-Jin Kim
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, 11 Biopolis Way, No. 02-02 Helios, Singapore 138667, Singapore;
| | - Kyung-Yun Kang
- Suncheon Research Center for Natural Medicines, Suncheon 540-950, Korea;
| | - Sung-Tae Yee
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon 540-950, Korea; (Y.-H.H.); (S.-G.H.); (S.-K.M.); (S.-J.K.); (S.-J.L.)
- Correspondence: ; Tel.: +82-61-750-3752; Fax: +82-61-750-3708
| |
Collapse
|
42
|
Tran GT, Wilcox PL, Dent LA, Robinson CM, Carter N, Verma ND, Hall BM, Hodgkinson SJ. Interleukin-5 Mediates Parasite-Induced Protection against Experimental Autoimmune Encephalomyelitis: Association with Induction of Antigen-Specific CD4 +CD25 + T Regulatory Cells. Front Immunol 2017; 8:1453. [PMID: 29163523 PMCID: PMC5671975 DOI: 10.3389/fimmu.2017.01453] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/17/2017] [Indexed: 12/18/2022] Open
Abstract
Objective To examine if the protective effect of parasite infection on experimental autoimmune encephalomyelitis (EAE) was due to interleukin (IL)-5, a cytokine produced by a type-2 response that induces eosinophilia. We hypothesize that, in parasite infections, IL-5 also promotes expansion of antigen-specific T regulatory cells that control autoimmunity. Methods Nippostrongylus brasiliensis larvae were used to infect Lewis rats prior to induction of EAE by myelin basic protein. Animals were sham treated, or given blocking monoclonal antibodies to interleukin 4 or 5 or to deplete CD25+ T cells. Reactivity of CD4+CD25+ T regulatory cells from these animals was examined. Results Parasite-infected hosts had reduced severity and length of EAE. The beneficial effect of parasitic infection was abolished with an anti-IL-5 or an anti-CD25 monoclonal antibody (mAb), but not anti-IL-4 mAb. Parasite-infected animals with EAE developed antigen-specific CD4+CD25+ T regulatory cells earlier than EAE controls and these expressed more Il5ra than controls. Treatment with IL-5 also reduced the severity of EAE and induced Il5ra expressing CD4+CD25+ T regulatory cells. Interpretation The results of this study suggested that IL-5 produced by the type-2 inflammatory response to parasite infection promoted induction of autoantigen-specific CD25+Il5ra+ T regulatory cells that reduced the severity of autoimmunity. Such a mechanism may explain the protective effect of parasite infection in patients with multiple sclerosis where eosinophilia is induced by IL-5, produced by the immune response to parasites.
Collapse
Affiliation(s)
- Giang T Tran
- Immune Tolerance Laboratory, UNSW Australia, Department of Neurology, Liverpool Hospital, Sydney, NSW, Australia
| | - Paul L Wilcox
- Immune Tolerance Laboratory, UNSW Australia, Department of Neurology, Liverpool Hospital, Sydney, NSW, Australia
| | - Lindsay A Dent
- Immune Tolerance Laboratory, UNSW Australia, Department of Neurology, Liverpool Hospital, Sydney, NSW, Australia
| | - Catherine M Robinson
- Immune Tolerance Laboratory, UNSW Australia, Department of Neurology, Liverpool Hospital, Sydney, NSW, Australia
| | - Nicole Carter
- Immune Tolerance Laboratory, UNSW Australia, Department of Neurology, Liverpool Hospital, Sydney, NSW, Australia
| | - Nirupama D Verma
- Immune Tolerance Laboratory, UNSW Australia, Department of Neurology, Liverpool Hospital, Sydney, NSW, Australia
| | - Bruce M Hall
- Immune Tolerance Laboratory, UNSW Australia, Department of Neurology, Liverpool Hospital, Sydney, NSW, Australia
| | - Suzanne J Hodgkinson
- Immune Tolerance Laboratory, UNSW Australia, Department of Neurology, Liverpool Hospital, Sydney, NSW, Australia
| |
Collapse
|
43
|
Licari A, Castagnoli R, Brambilla I, Marseglia A, Tosca MA, Marseglia GL, Ciprandi G. New approaches for identifying and testing potential new anti-asthma agents. Expert Opin Drug Discov 2017; 13:51-63. [PMID: 29077521 DOI: 10.1080/17460441.2018.1396315] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Asthma is a chronic disease with significant heterogeneity in clinical features, disease severity, pattern of underlying disease mechanisms, and responsiveness to specific treatments. While the majority of asthmatic patients are controlled by standard pharmacological strategies, a significant subgroup has limited therapeutic options representing a major unmet need. Ongoing asthma research aims to better characterize distinct clinical phenotypes, molecular endotypes, associated reliable biomarkers, and also to develop a series of new effective targeted treatment modalities. Areas covered: The expanding knowledge on the pathogenetic mechanisms of asthma has allowed researchers to investigate a range of new treatment options matched to patient profiles. The aim of this review is to provide a comprehensive and updated overview of the currently available, new and developing approaches for identifying and testing potential treatment options for asthma management. Expert opinion: Future therapeutic strategies for asthma require the identification of reliable biomarkers that can help with diagnosis and endotyping, in order to determine the most effective drug for the right patient phenotype. Furthermore, in addition to the identification of clinical and inflammatory phenotypes, it is expected that a better understanding of the mechanisms of airway remodeling will likely optimize asthma targeted treatment.
Collapse
Affiliation(s)
- Amelia Licari
- a Pediatric Clinic , Fondazione IRCCS San Matteo , Pavia , Italy
| | | | - Ilaria Brambilla
- a Pediatric Clinic , Fondazione IRCCS San Matteo , Pavia , Italy
| | | | - Maria Angela Tosca
- b Pediatric Pulmonology and Allergy , IRCCS Istituto Giannina Gaslini , Genoa , Italy
| | | | - Giorgio Ciprandi
- b Pediatric Pulmonology and Allergy , IRCCS Istituto Giannina Gaslini , Genoa , Italy.,c Internal Medicine , Ospedale Policlinico San Martino , Genoa , Italy
| |
Collapse
|
44
|
Svenningsen S, Nair P. Asthma Endotypes and an Overview of Targeted Therapy for Asthma. Front Med (Lausanne) 2017; 4:158. [PMID: 29018800 PMCID: PMC5622943 DOI: 10.3389/fmed.2017.00158] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/12/2017] [Indexed: 12/04/2022] Open
Abstract
Guidelines for the management of severe asthma do not emphasize the measurement of the inflammatory component of airway disease to indicate appropriate treatments or to monitor response to treatment. Inflammation is a central component of asthma and contributes to symptoms, physiological, and structural abnormalities. It can be assessed by a number of endotyping strategies based on “omics” technology such as proteomics, transcriptomics, and metabolomics. It can also be assessed using simple cellular responses by quantitative cytometry in sputum. Bronchitis may be eosinophilic, neutrophilic, mixed-granulocytic, or paucigranulocytic (eosinophils and neutrophils not elevated). Eosinophilic bronchitis is usually a Type 2 (T2)-driven process and therefore a sputum eosinophilia of greater than 3% usually indicates a response to treatment with corticosteroids or novel therapies directed against T2 cytokines such as IL-4, IL-5, and IL-13. Neutrophilic bronchitis represents a non-T2-driven disease, which is generally a predictor of response to antibiotics and may be a predictor to therapies targeted at pathways that lead to neutrophil recruitment such as TNF, IL-1, IL-6, IL-8, IL-23, and IL-17. Paucigranulocytic disease may not warrant anti-inflammatory therapy. These patients, whose symptoms may be driven largely by airway hyper-responsiveness may benefit from smooth muscle-directed therapies such as bronchial thermoplasty or mast-cell directed therapies. This review will briefly summarize the current knowledge regarding “omics-based signatures” and cellular endotyping of severe asthma and give an overview of segmentation of asthma therapeutics guided by the endotype.
Collapse
Affiliation(s)
| | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,St Joseph's Healthcare, Hamilton, ON, Canada
| |
Collapse
|
45
|
Mitchell PD, El-Gammal AI, O'Byrne PM. Anti-IgE and Biologic Approaches for the Treatment of Asthma. Handb Exp Pharmacol 2017; 237:131-152. [PMID: 27864676 DOI: 10.1007/164_2016_65] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Current asthma treatments are effective for the majority of patients with mild-to-moderate disease. However, in those with more severe refractory asthma, agents other than inhaled corticosteroids and beta-agonists are needed both to better manage this group of patients and to avoid the side effects of high-dose corticosteroids and the social and personal hardship endured. Several biological pathways have been targeted over the last 20 years, and this research has resulted in pharmacological approaches to attempt to better treat patients with severe refractory asthma. The flagship of the biologics, the anti-IgE monoclonal antibody, omalizumab, has proven efficacious in selected subgroups of asthma patients. Tailoring asthma treatments to suit specific subtypes of asthma patients is in keeping with ideals of personalized medicine. Research in the complex interplay of allergens, epithelial host defenses, cytokines, and innate and adaptive immunity interactions has allowed better understanding of the mechanics of allergy and inflammation in asthma. As a result, new biologic treatments have been developed that target several different phenotypes and endotypes in asthma. As knowledge of the efficacy of these biological agents in asthma emerges, as well as the type of patients in whom they are most beneficial, the movement toward personalized asthma treatment will follow.
Collapse
Affiliation(s)
- Patrick D Mitchell
- Firestone Institute for Respiratory Health and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amani I El-Gammal
- Firestone Institute for Respiratory Health and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Paul M O'Byrne
- Firestone Institute for Respiratory Health and the Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
46
|
Bagnasco D, Ferrando M, Varricchi G, Puggioni F, Passalacqua G, Canonica GW. Anti-Interleukin 5 (IL-5) and IL-5Ra Biological Drugs: Efficacy, Safety, and Future Perspectives in Severe Eosinophilic Asthma. Front Med (Lausanne) 2017; 4:135. [PMID: 28913336 PMCID: PMC5583162 DOI: 10.3389/fmed.2017.00135] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 07/25/2017] [Indexed: 12/16/2022] Open
Abstract
The definition of asthma has changed considerably in recent years, to the extent that asthma is no longer considered a single disease but a heterogeneous disorder that includes several phenotypes and, possibly, endotypes. A more detailed analysis of the immunological mechanisms underlying the pathogenesis of asthma shows interleukin 5 (IL-5) to be a crucial cytokine in several asthma phenotypes. In fact, IL-5 exerts selective action on eosinophils, which, in turn, sustain airway inflammation and worsen asthma symptoms and control. Clinical trials have shown drugs targeting IL-5 or its receptor alpha subunit (IL-5Ra) to be a promising therapeutic approach to severe asthma, whose characteristics render standard therapy of little use: systemic corticosteroids only partially control the disease and have well-known adverse effects, and omalizumab is used for allergic subtypes. Analysis of the design process of clinical trials reveals the importance of patient selection, taking into account both clinical data (e.g., exacerbations, lung function, and quality of life) and biomarkers (e.g., eosinophils, which are predictive of therapeutic response).
Collapse
Affiliation(s)
- Diego Bagnasco
- Allergy and Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Matteo Ferrando
- Allergy and Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Division of Clinical Immunology and Allergy, University of Naples Federico II, Naples, Italy
| | - Francesca Puggioni
- Department of Internal Medicine, Respiratory Disease Clinic, IRCCS Humanitas Clinical and Research Center, Humanitas University, Milan, Italy
| | - Giovanni Passalacqua
- Allergy and Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Giorgio Walter Canonica
- Allergy and Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, IRCCS AOU San Martino-IST, Genoa, Italy.,Department of Internal Medicine, Respiratory Disease Clinic, IRCCS Humanitas Clinical and Research Center, Humanitas University, Milan, Italy
| |
Collapse
|
47
|
Máspero J. Reslizumab in the treatment of inadequately controlled asthma in adults and adolescents with elevated blood eosinophils: clinical trial evidence and future prospects. Ther Adv Respir Dis 2017; 11:311-325. [PMID: 28683596 PMCID: PMC5933654 DOI: 10.1177/1753465817717134] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/02/2017] [Indexed: 12/22/2022] Open
Abstract
Eosinophils have long been implicated as playing a central role in the pathophysiology of asthma in many patients, and eosinophilic asthma is now recognized as an important asthma endotype. Eosinophil differentiation, maturation, migration, and survival are primarily under the control of interleukin-5 (IL-5). Reslizumab is a humanized monoclonal (immunoglobulin G4/κ) antibody that binds with high affinity to circulating human IL-5 and downregulates the IL-5 signaling pathway, potentially disrupting the maturation and survival of eosinophils. In 2016, an intravenous formulation of reslizumab was approved in the USA, Canada, and Europe as add-on maintenance treatment for patients aged ⩾18 years with severe asthma and with an eosinophilic phenotype. The efficacy of reslizumab as add-on intravenous therapy has been reported in several phase III studies in patients with inadequately controlled moderate-to-severe asthma and elevated blood eosinophil counts (⩾400 cells/µl). Compared with placebo, reslizumab was associated with significant improvements in clinical exacerbation rate, forced expiratory volume in 1 s, asthma symptoms and quality of life, and significant reductions in blood eosinophil counts. Reslizumab also demonstrated a favorable tolerability profile similar to that of placebo, with reported adverse events being mostly mild to moderate in severity. Ongoing studies are focusing on the evaluation of a subcutaneous formulation of reslizumab in patients with asthma and elevated eosinophil levels. This review discusses the preclinical and clinical trial data available on reslizumab, potential opportunities for predicting an early response to reslizumab, and future directions in the field of anti-IL-5 antibody therapy.
Collapse
Affiliation(s)
- Jorge Máspero
- Fundación Cidea Allergy and Respiratory Research
Unit, Paraguay 2035, 2*SS, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
48
|
Mukherjee M, Bulir DC, Radford K, Kjarsgaard M, Huang CM, Jacobsen EA, Ochkur SI, Catuneanu A, Lamothe-Kipnes H, Mahony J, Lee JJ, Lacy P, Nair PK. Sputum autoantibodies in patients with severe eosinophilic asthma. J Allergy Clin Immunol 2017; 141:1269-1279. [PMID: 28751233 DOI: 10.1016/j.jaci.2017.06.033] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 06/08/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The persistence of eosinophils in sputum despite high doses of corticosteroids indicates disease severity in asthmatic patients. Chronic inflamed airways can lose tolerance over time to immunogenic entities released on frequent eosinophil degranulation, which further contributes to disease severity and necessitates an increase in maintenance corticosteroids. OBJECTIVES We sought to investigate the possibility of a polyclonal autoimmune event in the airways of asthmatic patients and to identify associated clinical and molecular characteristics. METHODS The presence of autoantibodies against eosinophil peroxidase (EPX) and anti-nuclear antibodies was investigated in patients with eosinophilic asthma maintained on high-dose corticosteroids, prednisone, or both. The ability of sputum immunoglobulins to induce eosinophil degranulation in vitro was assessed. In addition, the associated inflammatory microenvironment in patients with detectable autoantibodies was examined. RESULTS We report a "polyclonal" autoimmune event occurring in the airways of prednisone-dependent asthmatic patients with increased eosinophil activity, recurrent pulmonary infections, or both, as evident by the concomitant presence of sputum anti-EPX and anti-nuclear antibodies of the IgG subtype. Extensive cytokine profiling of sputum revealed a TH2-dominated microenvironment (eotaxin-2, IL-5, IL-18, and IL-13) and increased signalling molecules that support the formation of ectopic lymphoid structures (B-cell activating factor and B cell-attracting chemokine 1). Immunoprecipitated sputum immunoglobulins from patients with increased autoantibody levels triggered eosinophil degranulation in vitro, with release of extensive histone-rich extracellular traps, an event unsuppressed by dexamethasone and possibly contributing to the steroid-unresponsive nature of these eosinophilic patients. CONCLUSION This study identifies an autoimmune endotype of severe asthma that can be identified by the presence of sputum autoantibodies against EPX and autologous cellular components.
Collapse
Affiliation(s)
- Manali Mukherjee
- Division of Respirology, McMaster University, Hamilton, Ontario, Canada
| | - David C Bulir
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katherine Radford
- Division of Respirology, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | - Sergei I Ochkur
- Division of Pulmonary Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Ana Catuneanu
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | - James Mahony
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - James J Lee
- Division of Pulmonary Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Paige Lacy
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
49
|
Bachert C, Sousa AR, Lund VJ, Scadding GK, Gevaert P, Nasser S, Durham SR, Cornet ME, Kariyawasam HH, Gilbert J, Austin D, Maxwell AC, Marshall RP, Fokkens WJ. Reduced need for surgery in severe nasal polyposis with mepolizumab: Randomized trial. J Allergy Clin Immunol 2017; 140:1024-1031.e14. [PMID: 28687232 DOI: 10.1016/j.jaci.2017.05.044] [Citation(s) in RCA: 326] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 05/19/2017] [Accepted: 05/31/2017] [Indexed: 01/19/2023]
Abstract
BACKGROUND Patients with eosinophilic nasal polyposis frequently require surgery, and recurrence rates are high. OBJECTIVE We sought to assess the efficacy and safety of mepolizumab versus placebo for severe bilateral nasal polyposis. METHODS This randomized, double-blind, placebo-controlled trial recruited patients aged 18 to 70 years with recurrent nasal polyposis requiring surgery. Patients received 750 mg of intravenous mepolizumab or placebo every 4 weeks for a total of 6 doses in addition to daily topical corticosteroid treatment. The primary end point was the number of patients no longer requiring surgery at Week 25 based on a composite end point of endoscopic nasal polyp score and nasal polyposis severity visual analog scale (VAS) score. Secondary end points included change in nasal polyposis severity VAS score, endoscopic nasal polyp score, improvement in individual VAS symptoms (rhinorrhea, mucus in throat, nasal blockage, and sense of smell), patient-reported outcomes, and safety. RESULTS One hundred five patients received mepolizumab (n = 54) or placebo (n = 51). A significantly greater proportion of patients in the mepolizumab group compared with the placebo group no longer required surgery at Week 25 (16 [30%] vs 5 [10%], respectively; P = .006). There was a significant improvement in nasal polyposis severity VAS score, endoscopic nasal polyp score, all individual VAS symptom scores, and Sino-Nasal Outcome Test patient-reported outcome score in the mepolizumab compared with placebo groups. Mepolizumab's safety profile was comparable with that of placebo. CONCLUSION In patients with recurrent nasal polyposis receiving topical corticosteroids who required surgery, mepolizumab treatment led to a greater reduction in the need for surgery and a greater improvement in symptoms than placebo.
Collapse
Affiliation(s)
- Claus Bachert
- Upper Airways Research Laboratory and Department of Oto-Rhino-Laryngology, Ghent University and Ghent University Hospital, Ghent, Belgium; Division of ENT Diseases, CLINTEC, Karolinska Institute, University of Stockholm, Stockholm, Sweden.
| | - Ana R Sousa
- Respiratory Therapy Unit, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom
| | - Valerie J Lund
- Royal National Throat, Nose and Ear Hospital, UCLH London, London, United Kingdom
| | - Glenis K Scadding
- Royal National Throat, Nose and Ear Hospital, UCLH London, London, United Kingdom
| | - Philippe Gevaert
- Upper Airways Research Laboratory and Department of Oto-Rhino-Laryngology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Shuaib Nasser
- Department of Allergy, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Stephen R Durham
- Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Marjolein E Cornet
- Department of Otorhinolaryngology, Academic Medical Center, Amsterdam, The Netherlands
| | - Harsha H Kariyawasam
- Royal National Throat, Nose and Ear Hospital, UCLH London, London, United Kingdom
| | - Jane Gilbert
- Clinical Statistics, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom
| | - Daren Austin
- Respiratory Therapy Unit, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom
| | - Aoife C Maxwell
- Respiratory Clinical Sciences & Operations, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom
| | - Richard P Marshall
- Respiratory Therapy Unit, GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom
| | - Wytske J Fokkens
- Department of Otorhinolaryngology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Guo XZJ, Thomas PG. New fronts emerge in the influenza cytokine storm. Semin Immunopathol 2017; 39:541-550. [PMID: 28555383 PMCID: PMC5580809 DOI: 10.1007/s00281-017-0636-y] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/23/2017] [Indexed: 12/17/2022]
Abstract
Influenza virus is a significant pathogen in humans and animals with the ability to cause extensive morbidity and mortality. Exuberant immune responses induced following infection have been described as a "cytokine storm," associated with excessive levels of proinflammatory cytokines and widespread tissue damage. Recent studies have painted a more complex picture of cytokine networks and their contributions to clinical outcomes. While many cytokines clearly inflict immunopathology, others have non-pathological delimited roles in sending alarm signals, facilitating viral clearance, and promoting tissue repair, such as the IL-33-amphiregulin axis, which plays a key role in resolving some types of lung damage. Recent literature suggests that type 2 cytokines, traditionally thought of as not involved in anti-influenza immunity, may play an important regulatory role. Here, we discuss the diverse roles played by cytokines after influenza infection and highlight new, serene features of the cytokine storm, while highlighting the specific functions of relevant cytokines that perform unique immune functions and may have applications for influenza therapy.
Collapse
Affiliation(s)
- Xi-Zhi J Guo
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Integrated Biomedical Sciences Program, Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
- Integrated Biomedical Sciences Program, Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|