1
|
Kayikci H, Damadoglu E, Cihanbeylerden M, Tuccar C, Karakaya G, Kalyoncu AF. Clinical characteristics and biological treatment responses of patients with late-onset asthma phenotype. Allergy Asthma Proc 2025; 46:109-118. [PMID: 40011985 DOI: 10.2500/aap.2025.46.240105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Background: The data on subphenotypes and treatment responses to biologicals in late-onset asthma (LOA) is limited. This study aims to compare the clinical characteristics and treatment responses in severe asthma patients receiving biological treatments, categorized into early-onset asthma (EOA) and LOA groups. Methods: Patients treated with omalizumab or mepolizumab for at least six months at a tertiary care adult allergy clinic between December 2015 and December 2023 were included. Patients with persistent respiratory symptoms starting at age ≥40 years were categorized as LOA, while those with onset <40 years were categorized as EOA. Changes in Asthma Control Questionnaire (ACQ-6) scores, forced expiratory volume in one second (FEV1) percentages, and blood eosinophil counts were assessed at baseline and 6 months. The percentage change in FEV1 (liters) at 6 months relative to baseline was measured. Clinical remission rates were evaluated in those completing one year of treatment. Results: Among 87 patients, 38 (43.7%) had LOA and 49 (56.3%) had EOA. Of these, 22 (25.3%) received omalizumab and 65 (74.7%) received mepolizumab, with a mean treatment duration of 24.7 (±19.7) months. LOA patients had higher obesity rates and tobacco consumption compared to EOA patients (p = 0.041 and p = 0.024, respectively). There were no significant differences between LOA and EOA groups in ACQ scores, FEV1 percentage, the percentage change in FEV1 in liters and eosinophil counts (p = 0.531, p = 0.219, p = 0.632, p = 0.700, respectively). Within LOA patients, ACQ scores did not significantly differ between those treated with omalizumab and mepolizumab (p = 0.801). At 6 months, eosinophil counts significantly decreased with mepolizumab but not with omalizumab (p = 0.002). Conclusion: Biological treatment responses were similar between LOA and EOA groups. Omalizumab and mepolizumab showed comparable efficacy, with the exception of eosinophil count changes in LOA patients.
Collapse
|
2
|
Jackson DJ, Wechsler ME, Brusselle G, Buhl R. Targeting the IL-5 pathway in eosinophilic asthma: A comparison of anti-IL-5 versus anti-IL-5 receptor agents. Allergy 2024; 79:2943-2952. [PMID: 39396109 DOI: 10.1111/all.16346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024]
Abstract
Eosinophilic asthma is characterized by frequent exacerbations, poor symptom control and accelerated lung function decline. It is now recognized that the immune response underlying eosinophilic asthma involves a complex network of interconnected pathways from both the adaptive and innate immune systems. Within this response, interleukin-5 (IL-5) plays a central role in eosinophil differentiation, activation and survival and has emerged as a key target for therapies treating severe asthma. The monoclonal antibodies mepolizumab and reslizumab target the ligand IL-5, preventing its interaction with eosinophils; in contrast, benralizumab binds to the IL-5 receptor (IL-5R), preventing IL-5 from binding and leading to substantially greater eosinophil reduction by enhanced antibody-dependent cell-mediated cytotoxicity. Although no direct head-to-head clinical trials of asthma have been published to formally evaluate the clinical significance of these different therapeutic approaches, the potential benefits of partial versus complete eosinophil depletion continue to remain an important area of study and debate. Here, we review the existing real-world and clinical study data of anti-IL-5/anti-IL-5R therapies in severe eosinophilic asthma.
Collapse
Affiliation(s)
- David J Jackson
- Guy's Severe Asthma Centre, Guy's and St Thomas' NHS Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | | | - Guy Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Roland Buhl
- Pulmonary Department, Mainz University Hospital, Mainz, Germany
| |
Collapse
|
3
|
Leung C, Tang M, Huang BK, Fain SB, Hoffman EA, Choi J, Dunican EM, Mauger DT, Denlinger LC, Jarjour NN, Israel E, Levy BD, Wenzel SE, Sumino K, Hastie AT, Schirm J, McCulloch CE, Peters MC, Woodruff PG, Sorkness RL, Castro M, Fahy JV. A Novel Air Trapping Segment Score Identifies Opposing Effects of Obesity and Eosinophilia on Air Trapping in Asthma. Am J Respir Crit Care Med 2024; 209:1196-1207. [PMID: 38113166 PMCID: PMC11146546 DOI: 10.1164/rccm.202305-0802oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 12/19/2023] [Indexed: 12/21/2023] Open
Abstract
Rationale: Density thresholds in computed tomography (CT) lung scans quantify air trapping (AT) at the whole-lung level but are not informative for AT in specific bronchopulmonary segments. Objectives: To apply a segment-based measure of AT in asthma to investigate the clinical determinants of AT in asthma. Methods: In each of 19 bronchopulmonary segments in CT lung scans from 199 patients with asthma, AT was categorized as present if lung attenuation was less than -856 Hounsfield units at expiration in ⩾15% of the lung area. The resulting AT segment score (0-19) was related to patient outcomes. Measurements and Main Results: AT varied at the lung segment level and tended to persist at the patient and lung segment levels over 3 years. Patients with widespread AT (⩾10 segments) had more severe asthma (P < 0.05). The mean (±SD) AT segment score in patients with a body mass index ⩾30 kg/m2 was lower than in patients with a body mass index <30 kg/m2 (3.5 ± 4.6 vs. 5.5 ± 6.3; P = 0.008), and the frequency of AT in lower lobe segments in obese patients was less than in upper and middle lobe segments (35% vs. 46%; P = 0.001). The AT segment score in patients with sputum eosinophils ⩾2% was higher than in patients without sputum eosinophilia (7.0 ± 6.1 vs. 3.3 ± 4.9; P < 0.0001). Lung segments with AT more frequently had airway mucus plugging than lung segments without AT (48% vs. 18%; P ⩽ 0.0001). Conclusions: In patients with asthma, air trapping is more severe in those with airway eosinophilia and mucus plugging, whereas those who are obese have less severe trapping because their lower lobe segments are spared.
Collapse
Affiliation(s)
- Clarus Leung
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Monica Tang
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Brendan K. Huang
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Sean B. Fain
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | - Eric A. Hoffman
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | - Jiwoong Choi
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas School of Medicine, Kansas City, Kansas
| | | | - David T. Mauger
- Division of Biostatistics and Bioinformatics, Penn State College of Medicine, The Pennsylvania State University, Hershey, Pennsylvania
| | - Loren C. Denlinger
- Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, and
| | - Nizar N. Jarjour
- Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, and
| | - Elliot Israel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Bruce D. Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Sally E. Wenzel
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kaharu Sumino
- Division of Pulmonary and Critical Care Medicine, Washington University, St. Louis, Missouri
| | - Annette T. Hastie
- Section for Pulmonary, Critical Care, Allergy and Immunology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; and
| | | | | | - Michael C. Peters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Prescott G. Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| | | | - Mario Castro
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas School of Medicine, Kansas City, Kansas
| | - John V. Fahy
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| |
Collapse
|
4
|
Loewenthal L, Busby J, McDowell R, Brown T, Burhan H, Chaudhuri R, Dennison P, Dodd JW, Doe S, Faruqi S, Gore R, Idris E, Jackson DJ, Patel M, Pantin T, Pavord I, Pfeffer PE, Price DB, Rupani H, Siddiqui S, Heaney LG, Menzies-Gow A. Impact of sex on severe asthma: a cross-sectional retrospective analysis of UK primary and specialist care. Thorax 2024; 79:403-411. [PMID: 38124220 DOI: 10.1136/thorax-2023-220512] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023]
Abstract
INTRODUCTION After puberty, females are more likely to develop asthma and in a more severe form than males. The associations between asthma and sex are complex with multiple intrinsic and external factors. AIM To evaluate the sex differences in the characteristics and treatment of patients with severe asthma (SA) in a real-world setting. METHODS Demographic, clinical and treatment characteristics for patients with SA in the UK Severe Asthma Registry (UKSAR) and Optimum Patient Care Research Database (OPCRD) were retrospectively analysed by sex using univariable and multivariable logistic regression analyses adjusted for year, age and hospital/practice. RESULTS 3679 (60.9% female) patients from UKSAR and 18 369 patients (67.9% female) from OPCRD with SA were included. Females were more likely to be symptomatic with increased Asthma Control Questionnaire-6 (UKSAR adjusted OR (aOR) 1.14, 95% CI 1.09 to 1.18) and Royal College of Physicians-3 Question scores (OPCRD aOR 1.29, 95% CI 1.13 to 1.47). However, they had a higher forced expiratory volume in 1 second per cent (FEV1%) predicted (UKSAR 68.7% vs 64.8%, p<0.001) with no significant difference in peak expiratory flow. Type 2 biomarkers IgE (UKSAR 129 IU/mL vs 208 IU/mL, p<0.001) and FeNO (UKSAR 36ppb vs 46ppb, p<0.001) were lower in females with no significant difference in blood eosinophils or biological therapy. Females were less likely to be on maintenance oral corticosteroids (UKSAR aOR 0.86, 95% CI 0.75 to 0.99) but more likely to be obese (UKSAR aOR 1.67, 95% CI 145 to 1.93; OPCRD SA aOR 1.46, 95% CI 1.34 to 1.58). CONCLUSIONS Females had increased symptoms and were more likely to be obese despite higher FEV1% predicted and lower type 2 biomarkers with consistent and clinically important differences across both datasets.
Collapse
Affiliation(s)
- Lola Loewenthal
- National Lung and Heart Institute, Imperial College London, London, UK
- Department of Asthma and Allergy, Department of Respiratory Medicine, Royal Brompton and Harefield Hospitals, London, UK
| | - John Busby
- Centre for Public Health, Queen's University Belfast School of Medicine Dentistry and Biomedical Sciences, Belfast, UK
| | - Ronald McDowell
- Queen's University Belfast, Belfast, UK
- Ulster University, Coleraine, UK
| | - Thomas Brown
- Respiratory Medicine, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | - Hassan Burhan
- Respiratory Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Rekha Chaudhuri
- Respiratory Medicine, Gartnavel General Hospital, Glasgow, UK
- University of Glasgow, Glasgow, UK
| | - Paddy Dennison
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton University Hospitals NHS Trust, Southampton, UK
| | - James William Dodd
- Academic Respiratory Unit, University of Bristol, Bristol, UK
- North Bristol Lung Centre, North Bristol NHS Trust, Westbury on Trym, UK
| | - Simon Doe
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Shoaib Faruqi
- Hull University Teaching Hospitals NHS Trust, Hull, UK
| | | | | | - David Joshua Jackson
- Guy's Severe Asthma Centre, Guy's and St Thomas' Hospitals NHS Trust, London, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London Faculty of Life Sciences and Medicine, London, UK
| | - Mitesh Patel
- University Hospitals Plymouth, University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - Thomas Pantin
- Respiratory Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Ian Pavord
- NIHR Respiratory BRC, Nuffield Department of Medicine, Oxford University, Oxford, UK
| | | | - David B Price
- Observational and Pragmatic Research Institute Pte Ltd, Singapore
- Centre of Academic Primary Care, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Hitasha Rupani
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
- University of Southampton, Southampton, UK
| | - Salman Siddiqui
- National Lung and Heart Institute, Imperial College London, London, UK
| | - Liam G Heaney
- Centre of Infection and Immunity, Queen's University Belfast, Belfast, UK
| | | |
Collapse
|
5
|
Lai K, Sun D, Dai R, Samoro R, Park HS, Åstrand A, Cohen D, Jison M, Shih VH, Werkström V, Yao Y, Zhang Y, Zheng W, Zhong N, Albert A, Jianping B, Bi C, Lijun C, Mei C, Min C, Ping C, Zhimin C, Chih-Feng C, Sook CY, Xiuhua F, Xiwen G, Wei G, Wei H, Zhihai H, Wei HX, Kewu H, Mao H, Grace Dawn IM, Inbeom J, Luning J, Mingyan J, Shanping J, Meiling J, Jian K, Woo KJ, Sang-Ha K, Jiulong K, Ping-Hung K, Jie L, Manxiang L, Minjing L, Ruoran L, Wen L, Xianhua L, Yanming L, Yong LS, Chuanhe L, Chuntao L, Jing L, Xiaoxia L, Huiyu L, Zhuang L, Shengxi M, Liangping M, Hoon MK, Lin M, Choon-Sik P, Sim PH, Hye-Kyung P, Jung-Won P, Diahn-Warng P, Ronnie S, Guochao S, Debin S, Dejun S, Chun-Hua W, Guangfa W, Limin W, Xuefen W, Yan W, Liping W, Haihong W, Yi X, Zuke X, Canmao X, Jin-Fu X, Xingxiang X, Xiyuan X, Jianping Y, Hongzhong Y, Joo YH, Wencheng Y, Jin Z, Longju Z, Min Z, Wei Z, Jianping Z, Ziwen Z, Xiaoli Z, Yingqun Z. Benralizumab efficacy and safety in severe asthma: A randomized trial in Asia. Respir Med 2024:107611. [PMID: 38570145 DOI: 10.1016/j.rmed.2024.107611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/19/2024] [Accepted: 03/24/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Benralizumab is indicated as add-on therapy in patients with uncontrolled, severe eosinophilic asthma; it has not yet been evaluated in a large Asian population with asthma in a clinical trial. OBJECTIVE To evaluate the efficacy and safety of benralizumab in patients with severe asthma in Asia. METHODS MIRACLE (NCT03186209) was a randomized, Phase 3 study in China, South Korea, and the Philippines. Patients aged 12-75 years with severe asthma receiving medium-to-high-dose inhaled corticosteroid/long-acting β2-agonists, stratified (2:1) by baseline blood eosinophil count (bEOS) (≥300/μL; <300/μL), were randomized (1:1) to benralizumab 30 mg or placebo. Endpoints included annual asthma exacerbation rate (AAER; primary endpoint), change from baseline at Week 48 in pre-bronchodilator (BD) forced expiratory volume in 1 second (pre-BD FEV1) and total asthma symptom score (TASS). Safety was evaluated ≤ Week 56. RESULTS Of 695 patients randomized, 473 had baseline bEOS ≥300/μL (benralizumab n = 236; placebo n = 237). In this population, benralizumab significantly reduced AAER by 74% (rate ratio 0.26 [95% CI 0.19, 0.36], p < 0.0001) and significantly improved pre-BD FEV1 (least squares difference [LSD] 0.25 L [95% CI 0.17, 0.34], p < 0.0001) and TASS (LSD -0.25 [-0.45, -0.05], p = 0.0126) versus placebo. In patients with baseline bEOS <300/μL, there were numerical improvements in AAER, pre-BD FEV1, and TASS with benralizumab versus placebo. The frequency of adverse events was similar for benralizumab (76%) and placebo (80%) in the overall population. CONCLUSIONS MIRACLE data reinforces the efficacy and safety of benralizumab for severe eosinophilic asthma in an Asian population, consistent with the global Phase 3 results.
Collapse
Affiliation(s)
- Kefang Lai
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dejun Sun
- Inner Mongolia People's Hospital, Hohhot, China
| | - Ranran Dai
- Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ronnie Samoro
- Healthlink Medical-Surgical-Dental Clinics and Diagnostic Center, Iloilo City, Philippines
| | - Hae-Sim Park
- Ajou University School of Medicine, Ajou University Medical Center, Suwon, Republic of Korea
| | - Annika Åstrand
- Late-stage Respiratory & Immunology, AstraZeneca, Gothenburg, Sweden
| | - David Cohen
- Late-stage Respiratory & Immunology, AstraZeneca, Gaithersburg, MD, USA
| | - Maria Jison
- Late-stage Respiratory & Immunology, AstraZeneca, Gaithersburg, MD, USA
| | - Vivian H Shih
- BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, MD, USA
| | | | - Yuhui Yao
- Respiratory & Immunology, R&D China, AstraZeneca, Shanghai, China
| | - Yajuan Zhang
- Respiratory & Immunology, R&D China, AstraZeneca, Shanghai, China
| | | | - Nanshan Zhong
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Albay Albert
- Manila Doctors Hospital, Metro Manila, Philippines
| | - Bo Jianping
- Second Hospital of Shanxi Medical University, Shanxi, China
| | - Chen Bi
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chen Lijun
- Yinchuan First People's Hospital, Yinchuan, China
| | - Chen Mei
- Chengdu Fifth People's Hospital, Sichuan, China
| | - Chen Min
- The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chen Ping
- The General Hospital of Shenyang Military, Shenyang, China
| | - Chen Zhimin
- The Children's Hospital of Zhejiang University College of Medicine, Zhejiang, China
| | | | | | - Fu Xiuhua
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Gao Xiwen
- Central Hospital of Minhang District, Shanghai, China
| | - Gu Wei
- Nanjing First Hospital, Nanjing, China
| | - Han Wei
- Qingdao Municipal Hospital, Qingdao, China
| | | | - Hu Xi Wei
- The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Huang Kewu
- Beijing Chaoyang Hospital, Beijing, China
| | - Huang Mao
- Jiangsu Province Hospital, Jiangsu, China
| | | | - Jeong Inbeom
- Konyang University Hospital, Daejeon, Republic of Korea
| | - Jiang Luning
- Affiliated Hospital of Jining Medical College, Jining, China
| | | | - Jiang Shanping
- Sun Yat-Sen Memorial Hospital Sun Yat-Sen University, Guangzhou, China
| | - Jin Meiling
- Zhongshan Hospital of Fudan University, Shangha, China
| | - Kang Jian
- The First Affiliated Hospital of China Medical University, Chongqing, China
| | - Kim Jin Woo
- The Catholic University of Korea, Seoul, Republic of Korea
| | - Kim Sang-Ha
- Yonsei University Wonju Severance Christian Hospital, Gangwon-do, Republic of Korea
| | - Kuang Jiulong
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | | | - Li Jie
- First Affiliated Hospital of Ganzhou Medical University, Guangzhou, China
| | - Li Manxiang
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Minjing
- The First People's Hospital of Foshan, Foshan, China
| | - Li Ruoran
- Xuzhou Central Hospital, Jiangsu, China
| | - Li Wen
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Li Xianhua
- The First People's Hospital of Neijiang, Neijiang, China
| | | | | | - Liu Chuanhe
- Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Liu Chuntao
- West China Hospital, Sichuan University, Sichuan, China
| | - Liu Jing
- The Fifth Affiliated Hospital Sun YAT-SEN University, China
| | | | - Lu Huiyu
- Taizhou People's Hospital, Taizhou, China
| | - Luo Zhuang
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ma Shengxi
- Xinxiang Central Hospital, Xinxiang, China
| | - Mao Liangping
- Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Min Kyung Hoon
- Korea University Guro Hospital, Seoul, Republic of Korea
| | - Mu Lin
- Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Park Choon-Sik
- Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Park Hae Sim
- Ajou University Hospital, Suwon-si, Republic of Korea
| | - Park Hye-Kyung
- Pusan National University Hospital, Busan, Republic of Korea
| | | | | | - Samoro Ronnie
- Healthlink Medical-Surgical-Dental Clinics and Diagnostic Center, Iloilo City, Philippines
| | - Shi Guochao
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sun Debin
- Lishui Central Hospital, Lishui, China
| | - Sun Dejun
- Inner Mongolia People's Hospital, Hohhot, China
| | - Wang Chun-Hua
- Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wang Guangfa
- Peking University First Hospital, Beijing, China
| | - Wang Limin
- Hangzhou First People's Hospital, Hangzhou, China
| | - Wang Xuefen
- The First Affiliated Hospital Zhejiang University, Zhejiang, China
| | - Wang Yan
- The Second Affiliated Hospital of Army Medical University, PLA, China
| | - Wei Liping
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Xiao Yi
- Yanan Hospital, Kunming City, Yunnan, China
| | - Xiao Zuke
- Jiangxi Provincial People's Hospital, Jiangxi, China
| | - Xie Canmao
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xu Jin-Fu
- Shanghai Pulmonary Hospital, Shanghai, China
| | - Xu Xingxiang
- Northern Jiangsu People's Hospital, Jiangsu, China
| | - Xu Xiyuan
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia, China
| | - Yan Jianping
- Zhejiang Provincial People's Hospital, Zhejiang, China
| | | | - Yoon Ho Joo
- Hanyang University Seoul Hospital, Seoul, Republic of Korea
| | - Yu Wencheng
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhang Jin
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhang Longju
- The First People's Hospital of Zunyi, Zunyi, China
| | - Zhang Min
- The People's Hospital of Ganzhou, Ganzhou, China
| | - Zhang Wei
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhao Jianping
- Tongji Hospital of Huazhong Science and Technology University, Wuhan, China
| | - Zhao Ziwen
- Guangzhou First People's Hospital, Guangzhou, China
| | - Zhu Xiaoli
- Zhongda Hospital Southeast University, Nanjing, China
| | - Zhu Yingqun
- The Third Hospital of Changsha, Changsha, China
| |
Collapse
|
6
|
McDonald VM, Hamada Y, Agusti A, Gibson PG. Treatable Traits in Asthma: The Importance of Extrapulmonary Traits-GERD, CRSwNP, Atopic Dermatitis, and Depression/Anxiety. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:824-837. [PMID: 38278324 DOI: 10.1016/j.jaip.2024.01.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/28/2024]
Abstract
Treatable traits is a personalized medicine approach to the management of airway disease. Assessing traits within the 3 domains of pulmonary, extrapulmonary, and behavioral/lifestyle/risk factor traits, and applying targeted treatments to effectively manage these traits, enables a holistic and personalized approach to care. Asthma is a heterogeneous and complex airway disease that is frequently complicated by several extrapulmonary traits that impact asthma outcomes and predict future outcomes. We propose that the identification of extrapulmonary and behavioral risk factor traits and the implementation of targeted therapy will lead to improved management of people with asthma. Furthermore, many extrapulmonary traits present as "connected comorbidities"; that is, they coexist with asthma, have an impact on asthma, and effective treatment improves both asthma and the comorbidity or the comorbidities may share a similar mechanism. In this review, we explore this concept and look at atopic dermatitis, chronic rhinosinusitis with nasal polyps, gastroesophageal reflux disease, anxiety, and depression as treatable traits of asthma and how these can be managed using this approach.
Collapse
Affiliation(s)
- Vanessa M McDonald
- Centre of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia.
| | - Yuto Hamada
- Centre of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia; Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan
| | - Alvar Agusti
- Respiratory Institute, Hospital Clinic, Universitat de Barcelona, IDIBAPS, CIBERES, Barcelona, Spain
| | - Peter G Gibson
- Centre of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
| |
Collapse
|
7
|
Maniscalco M, Candia C, Calabrese C, D'Amato M, Matera MG, Molino A, Cazzola M. Impact of biologics on lung hyperinflation in patients with severe asthma. Respir Med 2024; 225:107578. [PMID: 38431058 DOI: 10.1016/j.rmed.2024.107578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND In asthma, inflammation affects both the proximal and distal airways and can cause significant hyperinflation, which is thought to be a major cause of dyspnea. METHODS This is a retrospective observational study evaluating the effect of three months of treatment with different biologic drugs (benralizumab, dupilumab and omalizumab) on pulmonary hyperinflation in a cohort of patients with severe asthma already receiving regular triple inhaled therapy. Changes in RV, RV/TLC ratio, FRC and FRC/TLC ratio were the primary efficacy measures. Secondary outcomes included FEV1, FVC, FEV1/FVC ratio, IC, IC/TLC ratio, asthma control test, the percentage of eosinophils in the blood and fractional FENO. RESULTS Benralizumab led to significant changes (p < 0.001) in RV, RV/TLC, FRC, and FRC/TLC. Dupilumab demonstrated a notable reduction in RV (p = 0.017) and RV/TLC (p = 0.002), but the decreases in FRC and FRC/TLC were merely numerical and not as pronounced as those induced by benralizumab. Omalizumab's positive impact on RV (p = 0.057) and RV/TLC (p = 0.085), as well as FRC (p = 0.202) and FRC/TLC (p = 0.096), was also predominantly numerical, with a tendency towards efficacy, albeit excluding the effect on FRC. Treatment with biologics resulted in improvements in all other lung function parameters assessed and a decrease in FENO levels. CONCLUSION This study, although limited by small sample size, lack of a placebo control, and unbalanced group sizes, suggests that biological agents are effective in reducing lung hyperinflation even after a relatively short treatment.
Collapse
Affiliation(s)
- Mauro Maniscalco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, Telese Terme, Italy
| | - Claudio Candia
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, Telese Terme, Italy
| | - Cecilia Calabrese
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria D'Amato
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Maria Gabriella Matera
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonio Molino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Mario Cazzola
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
8
|
Visser E, Ten Brinke A, Sizoo D, Pepels JJS, Ten Have L, van der Wiel E, van Zutphen T, Kerstjens HAM, de Jong K. Effect of dietary interventions on markers of type 2 inflammation in asthma: A systematic review. Respir Med 2024; 221:107504. [PMID: 38141862 DOI: 10.1016/j.rmed.2023.107504] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/29/2023] [Accepted: 12/17/2023] [Indexed: 12/25/2023]
Abstract
INTRODUCTION Type 2 (T2) inflammation is a key mechanism in the pathophysiology of asthma. Diet may have immunomodulatory effects, and a role for diet in T2 inflammation has been suggested in the literature. Indeed, diet and food allergies play a role in children with atopic asthma, but less is known about diet in relation to adult asthma, which is often non-atopic. OBJECTIVE To review the effect of dietary interventions on markers of T2 inflammation in adults with asthma. METHODS The databases PubMed, Embase, Cochrane Library, and CINAHL were searched for eligible studies until December 2022. We included studies of all types of foods, nutrients, diets or supplements, either as an exposure or as an intervention, in adults and adolescents with asthma. Outcomes of interest included the T2 biomarkers FeNO, eosinophils, IL-4, IL-5, IL-13, eosinophil cationic protein and eosinophil peroxidase. The methodological quality of eligible studies was systematically evaluated, and the results were summarised according to dietary clusters. RESULTS The systematic search identified studies on the dietary clusters antioxidants (n = 14), fatty acids, (n = 14), Mediterranean-style diets (n = 5), phytotherapy (n = 7), prebiotics & probiotics (n = 8), vitamin D (n = 7), and other dietary factors (n = 5). Studies within the phytotherapy and omega-3 poly-unsaturated fatty acids (PUFA) clusters showed possible improvements in T2 inflammation. Furthermore, we found little evidence for an effect of antioxidants, prebiotics & probiotics, and Mediterranean-style diets on T2 inflammation. However, heterogeneity in study protocols, methodological shortcomings and limited power of almost all studies make it difficult to fully determine the impact of different dietary approaches on T2 inflammation in asthma. CONCLUSIONS Overall, the current evidence does not support a specific dietary intervention to improve T2 inflammation in asthma. Interventions involving phytotherapy and omega-3 PUFA currently have the best evidence and warrant further evaluation in well-designed and adequately powered studies, while taking into account T2-high phenotypes of asthma.
Collapse
Affiliation(s)
- Edith Visser
- Department of Epidemiology, Medical Centre Leeuwarden, Leeuwarden, the Netherlands; Department of Sustainable Health, Faculty Campus Fryslân, University of Groningen, Leeuwarden, the Netherlands.
| | - Anneke Ten Brinke
- Department of Pulmonary Medicine, Medical Centre Leeuwarden, Leeuwarden, the Netherlands.
| | - Dionne Sizoo
- Department of Sustainable Health, Faculty Campus Fryslân, University of Groningen, Leeuwarden, the Netherlands; Centre Obesity Northern Netherlands (CON), Department of Surgery, Medical Centre Leeuwarden, Leeuwarden, the Netherlands.
| | - Janneke J S Pepels
- Department of Epidemiology, Medical Centre Leeuwarden, Leeuwarden, the Netherlands.
| | - Lianne Ten Have
- Department of Epidemiology, Medical Centre Leeuwarden, Leeuwarden, the Netherlands.
| | - Erica van der Wiel
- Department of Pulmonary Medicine, Martini Hospital, Groningen, the Netherlands.
| | - Tim van Zutphen
- Department of Sustainable Health, Faculty Campus Fryslân, University of Groningen, Leeuwarden, the Netherlands.
| | - Huib A M Kerstjens
- Department of Pulmonary Medicine, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Kim de Jong
- Department of Epidemiology, Medical Centre Leeuwarden, Leeuwarden, the Netherlands.
| |
Collapse
|
9
|
Matsubara M, Yagi K, Minami Y, Kanda E, Sunada Y, Tao Y, Takai H, Shikata E, Hirai S, Matsubara S, Uno M. Preoperative elevated eosinophils in peripheral blood for prediction of postoperative recurrence of chronic subdural hematoma. J Neurosurg 2023; 139:708-713. [PMID: 36640094 DOI: 10.3171/2022.12.jns222432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/07/2022] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Chronic subdural hematoma (CSDH) is a common neurological disease with a significant postoperative recurrence rate. There are numerous reported studies of the development of CSDH. In recent years, fibrinolysis, angiogenesis, and inflammation have all been identified as relevant factors in the development of CSDH. While several authors have reported risk factors associated with CSDH recurrence, differential blood count of leukocytes has not yet been discussed. Therefore, in this study the authors aimed to retrospectively investigate the association between differential blood leukocyte count and the rate of CSDH recurrence. METHODS The authors retrospectively reviewed 476 patients with 529 CSDHs who underwent surgery at a single institution between January 2011 and December 2021. After exclusion of patients who had not undergone a differential blood test of leukocytes preoperatively, CSDHs in 517 cerebral hemispheres of 466 patients were included in the study. Peripheral blood eosinophil counts ≥ 100/µL were considered eosinophil rich. RESULTS CSDHs in 494 cerebral hemispheres of 445 patients were followed up postoperatively for at least 3 months or until resolution indicated by CSDH disappearance. Postoperative recurrence of CSDH was observed in 46 cerebral hemispheres (9.3%). Among the preoperative differential blood counts of all leukocytes, eosinophils alone were significantly associated with CSDH recurrence (median [IQR] 76/µL [30-155/µL] vs 119/µL [39-217/µL]; p = 0.03). Multivariable regression analysis showed thrombocytopenia (adjusted OR [aOR] 5.23, 95% CI 1.85-14.79; p = 0.002), use of anticoagulant drugs (aOR 2.51, 95% CI 1.17-5.38; p = 0.02), hematoma volume (10 mL per increase) (aOR 1.08, 95% CI 1.00-1.16; p = 0.04), and eosinophil-rich peripheral blood (aOR 2.22, 95% CI 1.17-4.23; p = 0.02) were all independent predictors for CSDH recurrence. CONCLUSIONS This study showed that preoperative peripheral blood eosinophil count was an independent risk factor for CSDH recurrence. Therefore, patients with CSDH who have elevated eosinophils preoperatively in peripheral blood require careful follow-up.
Collapse
Affiliation(s)
| | | | | | - Eiichiro Kanda
- 2Medical Science, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Quirce S, Cosío BG, España A, Blanco R, Mullol J, Santander C, del Pozo V. Management of eosinophil-associated inflammatory diseases: the importance of a multidisciplinary approach. Front Immunol 2023; 14:1192284. [PMID: 37266434 PMCID: PMC10229838 DOI: 10.3389/fimmu.2023.1192284] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/05/2023] [Indexed: 06/03/2023] Open
Abstract
Elevated eosinophil counts in blood and tissue are a feature of many pathological processes. Eosinophils can migrate and accumulate in a wide variety of tissues and, by infiltrating a target organ, can mediate the development of several inflammatory diseases. The normalization of eosinophilia is a common biomarker of a treatable trait and can also be used as a prognostic and predictive biomarker since it implies a reduction in type 2 inflammation that contributes to disease pathogenesis. Biological therapies targeting this cell type and its proinflammatory mediators have been shown to be effective in the management of a number of eosinophilic diseases, and for this reason they constitute a potential common strategy in the treatment of patients with various multimorbidities that present with type 2 inflammation. Various biological options are available that could be used to simultaneously treat multiple target organs with a single drug, bearing in mind the need to offer personalized treatments under the umbrella of precision medicine in all patients with eosinophil-associated diseases (EADs). In addition to reviewing these issues, we also discuss a series of perspectives addressing the management of EAD patients from a multidisciplinary approach, with the collaboration of health professionals from different specialties who manage the different multimorbidities that frequently occur in these patients. We examine the basic principles of care that this multidisciplinary approach must cover and present a multidisciplinary expert opinion regarding the ideal management of patients with EADs, from diagnosis to therapeutic approach and follow-up.
Collapse
Affiliation(s)
- Santiago Quirce
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Department of Allergology, Hospital Universitario La Paz, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Borja G. Cosío
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Department of Respiratory Medicine, Hospital Universitari Son Espases, Fundación Instituto de Investigación Sanitaria Islas Baleares (IdiSBa), Palma de Mallorca, Spain
| | - Agustín España
- Department of Dermatology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ricardo Blanco
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Immunology Group, Instituto de Investigación Sanitaria Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Joaquim Mullol
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Rhinology Unit and Smell Clinic, Ear, Nose and Throat (ENT) Department, Hospital Clínic de Barcelona, Universitat de Barcelona (UB) - Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Cecilio Santander
- Department of Gastroenterology and Hepatology, Hospital Universitario La Princesa, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), Madrid, Spain
- Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Victoria del Pozo
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Immunoallergy Laboratory, Immunology Department, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| |
Collapse
|
11
|
Kere M, Klevebro S, Hernandez-Pacheco N, Ödling M, Ekström S, Mogensen I, Janson C, Palmberg L, van Hage M, Georgelis A, Bergström A, Kull I, Melén E, Björkander S. Exploring proteomic plasma biomarkers in eosinophilic and neutrophilic asthma. Clin Exp Allergy 2023; 53:186-197. [PMID: 36104952 DOI: 10.1111/cea.14229] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/22/2022] [Accepted: 09/12/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Few biomarkers identify eosinophilic and neutrophilic asthma beyond cell concentrations in blood or sputum. Finding novel biomarkers for asthma endotypes could give insight about disease mechanisms and guide tailored treatment. Our aim was to investigate clinical characteristics and inflammation-related plasma proteins in relation to blood eosinophil and neutrophil concentrations in subjects with and without asthma. METHODS We included 24-26-year-old subjects (n = 2063) from the Swedish population-based cohort BAMSE. Subjects with asthma (n = 239) and without asthma (n = 1824) were subdivided based on blood eosinophil and neutrophil concentrations (cut-offs 0.3 × 109 /L and 5.0 × 109 /L, respectively). We measured the levels of 92 plasma proteins using Olink Proseek Multiplex Inflammation Panel Assay. Group statistics tests were used to analyse the data, as well as adjusted multiple logistic regression models. RESULTS Among subjects with asthma, 21.8% had eosinophilic asthma and 20.5% neutrophilic asthma. Eosinophilic asthma, but not neutrophilic asthma, was associated with a distinct clinical phenotype with, for example, higher proportions of eczema and sensitization. Most plasma proteins that associated with high eosinophil and/or neutrophil blood concentrations in subjects with asthma showed similar associations in subjects without asthma. However, out of these proteins, MMP10 levels were associated with eosinophilic asthma and were significantly higher as compared to controls with high eosinophilic concentration, while CCL4 levels associated with high neutrophil concentration only in subjects with asthma. CONCLUSIONS Eosinophilic asthma was associated with a clear clinical phenotype. With our definitions, we identified MMP10 as a possible plasma biomarker for eosinophilic asthma and CCL4 was linked to neutrophilic asthma. These proteins should be evaluated further in clinical settings and using sputum granulocytes to define the asthma endotypes.
Collapse
Affiliation(s)
- Maura Kere
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Klevebro
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Natalia Hernandez-Pacheco
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Maria Ödling
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Ekström
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden.,Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ida Mogensen
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Christer Janson
- Department of Medical Sciences, Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Lena Palmberg
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marianne van Hage
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Hospital, Stockholm, Sweden
| | - Antonios Georgelis
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden.,Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Bergström
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden.,Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Inger Kull
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Sophia Björkander
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Hizawa N. The understanding of asthma pathogenesis in the era of precision medicine. Allergol Int 2023; 72:3-10. [PMID: 36195530 DOI: 10.1016/j.alit.2022.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/30/2022] [Indexed: 01/25/2023] Open
Abstract
Asthma is a syndrome with extremely diverse clinical phenotypes in which the onset, severity, and response to treatment are defined by the complex interplay of many genetic and environmental factors. Environmental factors epigenetically affect gene expression, and the disease is driven by a multidimensional dynamic network involving RNA and protein molecules derived from gene expression, as well as various metabolic products. In other words, specific pathophysiological mechanisms or endotypes are dynamic networks that arise in response to individual genotypes and the various environmental factors to which individuals have been exposed since before birth, such as diet, infection, air pollution, smoking, antibiotic use, and the bacterial flora of the intestinal tract, skin, and lungs. A key feature of asthma genome scans is their potential to reveal the molecular pathways that lead to pathogenesis. Endotypes that drive the disease have a significant impact on the phenotypes of asthma patients, including their drug responsiveness. Understanding endotypes will lead to not only the implementation of therapies that are tailored to the specific molecular network(s) underlying the patient's condition, but also to the development of therapeutic strategies that target individual endotypes, as well as to precision health, which will enable the prediction of disease onset with high accuracy from an early stage and the implementation of preventive strategies based on endotypes. Understanding of endotypes will pave the way for the practice of precision medicine in asthma care, moving away from 'one-size-fits-all' medicine and population-based prevention approaches that do not take individuals' susceptibility into account.
Collapse
Affiliation(s)
- Nobuyuki Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
13
|
Menzies-Gow A, Gurnell M, Heaney LG, Corren J, Bel EH, Maspero J, Harrison T, Jackson DJ, Price D, Lugogo N, Kreindler J, Burden A, de Giorgio-Miller A, Faison S, Padilla K, Martin UJ, Garcia Gil E. Adrenal function recovery after durable oral corticosteroid sparing with benralizumab in the PONENTE study. Eur Respir J 2022; 60:2103226. [PMID: 35896216 PMCID: PMC9791910 DOI: 10.1183/13993003.03226-2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 07/04/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Oral corticosteroid (OCS) dependence among patients with severe eosinophilic asthma can cause adverse outcomes, including adrenal insufficiency. PONENTE's OCS reduction phase showed that, following benralizumab initiation, 91.5% of patients eliminated corticosteroids or achieved a final dosage ≤5 mg·day-1 (median (range) 0.0 (0.0-40.0) mg). METHODS The maintenance phase assessed the durability of corticosteroid reduction and further adrenal function recovery. For ∼6 months, patients continued benralizumab 30 mg every 8 weeks without corticosteroids or with the final dosage achieved during the reduction phase. Investigators could prescribe corticosteroids for asthma exacerbations or increase daily dosages for asthma control deteriorations. Outcomes included changes in daily OCS dosage, Asthma Control Questionnaire (ACQ)-6 and St George's Respiratory Questionnaire (SGRQ), as well as adrenal status, asthma exacerbations and adverse events. RESULTS 598 patients entered PONENTE; 563 (94.1%) completed the reduction phase and entered the maintenance phase. From the end of reduction to the end of maintenance, the median (range) OCS dosage was unchanged (0.0 (0.0-40.0) mg), 3.2% (n=18/563) of patients experienced daily dosage increases, the mean ACQ-6 score decreased from 1.26 to 1.18 and 84.5% (n=476/563) of patients were exacerbation free. The mean SGRQ improvement (-19.65 points) from baseline to the end of maintenance indicated substantial quality-of-life improvements. Of patients entering the maintenance phase with adrenal insufficiency, 32.4% (n=104/321) demonstrated an improvement in adrenal function. Adverse events were consistent with previous reports. CONCLUSIONS Most patients successfully maintained maximal OCS reduction while achieving improved asthma control with few exacerbations and maintaining or recovering adrenal function.
Collapse
Affiliation(s)
- Andrew Menzies-Gow
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospitals, London, UK
| | - Mark Gurnell
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Liam G Heaney
- Wellcome-Wolfson Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Jonathan Corren
- David Geffen School of Medicine at UCLA and Allergy Medical Clinic Inc., Los Angeles, CA, USA
| | - Elisabeth H Bel
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Timothy Harrison
- Nottingham Respiratory NIHR BRC, University of Nottingham, Nottingham, UK
- BioPharmaceuticals R&D Digital, AstraZeneca, Cambridge, UK
| | - David J Jackson
- Guy's Severe Asthma Centre, Guy's and St Thomas' NHS Trust, London, UK
- Asthma UK Centre, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - David Price
- Observational and Pragmatic Research Institute, Singapore, Singapore
- Centre of Academic Primary Care, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Njira Lugogo
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - James Kreindler
- Global Medical Respiratory, BioPharmaceuticals Medical, AstraZeneca, Wilmington, DE, USA
| | - Annie Burden
- BioPharmaceuticals R&D, Late Respiratory and Immunology, Biometrics, AstraZeneca, Cambridge, UK
| | | | - Sarai Faison
- Late Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Durham, NC, USA
| | - Kelly Padilla
- Late Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Durham, NC, USA
| | - Ubaldo J Martin
- Late Stage Development, Respiratory and Immunology Therapeutic Area, AstraZeneca, Gaithersburg, MD, USA
| | - Esther Garcia Gil
- Global Medical Respiratory, BioPharmaceuticals Medical, AstraZeneca, Barcelona, Spain
| |
Collapse
|
14
|
Fouka E, Domvri K, Gkakou F, Alevizaki M, Steiropoulos P, Papakosta D, Porpodis K. Recent insights in the role of biomarkers in severe asthma management. Front Med (Lausanne) 2022; 9:992565. [PMID: 36226150 PMCID: PMC9548530 DOI: 10.3389/fmed.2022.992565] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/09/2022] [Indexed: 11/28/2022] Open
Abstract
Contemporary asthma management requires a proactive and individualized approach, combining precision diagnosis and personalized treatment. The introduction of biologic therapies for severe asthma to everyday clinical practice, increases the need for specific patient selection, prediction of outcomes and monitoring of these costly and long-lasting therapies. Several biomarkers have been used in asthma in disease identification, prediction of asthma severity and prognosis, and response to treatment. Novel advances in the area of personalized medicine regarding disease phenotyping and endotyping, encompass the development and application of reliable biomarkers, accurately quantified using robust and reproducible methods. The availability of powerful omics technologies, together with integrated and network-based genome data analysis, and microbiota changes quantified in serum, body fluids and exhaled air, will lead to a better classification of distinct phenotypes or endotypes. Herein, in this review we discuss on currently used and novel biomarkers for the diagnosis and treatment of asthma.
Collapse
Affiliation(s)
- Evangelia Fouka
- G. Papanikolaou General Hospital, Thessaloniki, Greece
- Pulmonary Department of Aristotle University of Thessaloniki, Thessaloniki, Greece
- *Correspondence: Evangelia Fouka
| | - Kalliopi Domvri
- G. Papanikolaou General Hospital, Thessaloniki, Greece
- Pulmonary Department of Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Foteini Gkakou
- G. Papanikolaou General Hospital, Thessaloniki, Greece
- Pulmonary Department of Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Alevizaki
- G. Papanikolaou General Hospital, Thessaloniki, Greece
- Pulmonary Department of Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Despoina Papakosta
- G. Papanikolaou General Hospital, Thessaloniki, Greece
- Pulmonary Department of Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Porpodis
- G. Papanikolaou General Hospital, Thessaloniki, Greece
- Pulmonary Department of Aristotle University of Thessaloniki, Thessaloniki, Greece
- Konstantinos Porpodis
| |
Collapse
|
15
|
Eosinophilic inflammation: An Appealing Target for Pharmacologic Treatments in Severe Asthma. Biomedicines 2022; 10:biomedicines10092181. [PMID: 36140282 PMCID: PMC9496162 DOI: 10.3390/biomedicines10092181] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 11/19/2022] Open
Abstract
Severe asthma is characterized by different endotypes driven by complex pathologic mechanisms. In most patients with both allergic and non-allergic asthma, predominant eosinophilic airway inflammation is present. Given the central role of eosinophilic inflammation in the pathophysiology of most cases of severe asthma and considering that severe eosinophilic asthmatic patients respond partially or poorly to corticosteroids, in recent years, research has focused on the development of targeted anti-eosinophil biological therapies; this review will focus on the unique and particular biology of the eosinophil, as well as on the current knowledge about the pathobiology of eosinophilic inflammation in asthmatic airways. Finally, current and prospective anti-eosinophil therapeutic strategies will be discussed, examining the reason why eosinophilic inflammation represents an appealing target for the pharmacological treatment of patients with severe asthma.
Collapse
|
16
|
Louis R, Satia I, Ojanguren I, Schleich F, Bonini M, Tonia T, Rigau D, Ten Brinke A, Buhl R, Loukides S, Kocks JWH, Boulet LP, Bourdin A, Coleman C, Needham K, Thomas M, Idzko M, Papi A, Porsbjerg C, Schuermans D, Soriano JB, Usmani OS. European Respiratory Society guidelines for the diagnosis of asthma in adults. Eur Respir J 2022; 60:2101585. [PMID: 35169025 DOI: 10.1183/13993003.01585-2021] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 01/10/2022] [Indexed: 12/20/2022]
Abstract
Although asthma is very common, affecting 5-10% of the population, the diagnosis of asthma in adults remains a challenge in the real world, which results in both over- and under-diagnosis. A taskforce was set up by the European Respiratory Society to systematically review the literature on the diagnostic accuracy of tests used to diagnose asthma in adult patients and provide recommendations for clinical practice.The taskforce defined eight Population, Index, Comparator and Outcome questions that were assessed using the Grading of Recommendations, Assessment, Development and Evaluation approach. The taskforce utilised the outcomes to develop an evidence-based diagnostic algorithm, with recommendations for a pragmatic guideline for everyday practice that was directed by real-life patient experiences.The taskforce supports the initial use of spirometry followed by bronchodilator reversibility testing (if airway obstruction is present). If initial spirometry fails to show obstruction, further tests should be performed in the following order: exhaled nitric oxide fraction, peak expiratory flow variability, or, in secondary care, bronchial challenge. We present the thresholds for each test that are compatible with a diagnosis of asthma in the presence of current symptoms.The taskforce reinforces spirometry as a priority and recognises the value of measuring blood eosinophils and serum immunoglobulin E to phenotype the patient. Measuring gas trapping by body plethysmography in patients with preserved forced expiratory volume in 1 s/forced vital capacity ratio deserves further attention. The taskforce draws attention to the difficulty of making a correct diagnosis in patients already receiving inhaled corticosteroids; the comorbidities that may obscure diagnosis; the importance of phenotyping; and the necessity of considering the patient experience in the diagnostic process.
Collapse
Affiliation(s)
- Renaud Louis
- Dept of Pneumology, CHU Liege, GIGA I Research Group, University of Liege, Liege, Belgium
- Taskforce chair
| | - Imran Satia
- Division of Respirology, McMaster University, Hamilton, ON, Canada
- These authors contributed equally
| | - Inigo Ojanguren
- Servei de Pneumologia, Hospital Universitari Vall d'Hebron, Universitat Autònomona de Barcelona (UAB); Vall d'Hebron Institut de Recerca (VHIR); CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
- These authors contributed equally
| | - Florence Schleich
- Dept of Pulmonary Medicine, University of Liege, Liège, Belgium
- These authors contributed equally
| | - Matteo Bonini
- Sapienza University of Rome, Rome, Italy
- These authors contributed equally
| | - Thomy Tonia
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - David Rigau
- Iberoamerican Cochrane Centre, Barcelona, Spain
| | - Anne Ten Brinke
- Pulmonology, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Roland Buhl
- Pulmonary Dept, Mainz University Hospital, Mainz, Germany
| | | | | | - Louis-Philippe Boulet
- Pneumologie, Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec, QC, Canada
| | | | | | | | - Mike Thomas
- Primary Care and Population Sciences Division, University of Southampton, Southampton, UK
| | - Marco Idzko
- Dept of Respiratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Alberto Papi
- Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Celeste Porsbjerg
- Respiratory Medicine, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Daniel Schuermans
- Respiratory Division, Academic Hospital UZBrussel, Brussels, Belgium
| | - Joan B Soriano
- Universidad Autónoma de Madrid, Hospital Universitario de la Princesa, Madrid, Spain
| | - Omar S Usmani
- National Heart and Lung Institute, Imperial College London, London, UK
- Taskforce co-chair
| |
Collapse
|
17
|
Li M, Wen Ma Z, Jun Deng S, Oliver BG, Wang T, Ping Zhang H, Wang L, McDonald VM, Wang J, Liu D, Gibson PG, Ming Luo F, Min Li W, Jing Wan H, Wang G. Development and validation of a noninvasive prediction model for identifying eosinophilic asthma. Respir Med 2022; 201:106935. [DOI: 10.1016/j.rmed.2022.106935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 10/17/2022]
|
18
|
Hoda U, Pavlidis S, Bansal AT, Takahashi K, Hu S, Ng Kee Kwong F, Rossios C, Sun K, Bhavsar P, Loza M, Baribaud F, Chanez P, Fowler SJ, Horvath I, Montuschi P, Singer F, Musial J, Dahlen B, Krug N, Sandstrom T, Shaw DE, Lutter R, Fleming LJ, Howarth PH, Caruso M, Sousa AR, Corfield J, Auffray C, De Meulder B, Lefaudeux D, Dahlen SE, Djukanovic R, Sterk PJ, Guo Y, Adcock IM, Chung KF. Clinical and transcriptomic features of persistent exacerbation-prone severe asthma in U-BIOPRED cohort. Clin Transl Med 2022; 12:e816. [PMID: 35474304 PMCID: PMC9043117 DOI: 10.1002/ctm2.816] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 01/28/2023] Open
Abstract
Background Exacerbation‐prone asthma is a feature of severe disease. However, the basis for its persistency remains unclear. Objectives To determine the clinical and transcriptomic features of frequent exacerbators (FEs) and persistent FEs (PFEs) in the U‐BIOPRED cohort. Methods We compared features of FE (≥2 exacerbations in past year) to infrequent exacerbators (IE, <2 exacerbations) and of PFE with repeat ≥2 exacerbations during the following year to persistent IE (PIE). Transcriptomic data in blood, bronchial and nasal epithelial brushings, bronchial biopsies and sputum cells were analysed by gene set variation analysis for 103 gene signatures. Results Of 317 patients, 62.4% had FE, of whom 63.6% had PFE, while 37.6% had IE, of whom 61.3% had PIE. Using multivariate analysis, FE was associated with short‐acting beta‐agonist use, sinusitis and daily oral corticosteroid use, while PFE was associated with eczema, short‐acting beta‐agonist use and asthma control index. CEA cell adhesion molecule 5 (CEACAM5) was the only differentially expressed transcript in bronchial biopsies between PE and IE. There were no differentially expressed genes in the other four compartments. There were higher expression scores for type 2, T‐helper type‐17 and type 1 pathway signatures together with those associated with viral infections in bronchial biopsies from FE compared to IE, while there were higher expression scores of type 2, type 1 and steroid insensitivity pathway signatures in bronchial biopsies of PFE compared to PIE. Conclusion The FE group and its PFE subgroup are associated with poor asthma control while expressing higher type 1 and type 2 activation pathways compared to IE and PIE, respectively.
Collapse
Affiliation(s)
- Uruj Hoda
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Stelios Pavlidis
- Department of Computing & Data Science Institute, Imperial College London
| | | | - Kentaro Takahashi
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK.,Research Centre for Allergy and Clinical Immunology, Asahi General Hospital, Asahi, Japan
| | | | - Francois Ng Kee Kwong
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Christos Rossios
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | | | - Pankaj Bhavsar
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Matthew Loza
- Janssen Research and Development, High Wycombe, Buckinghamshire, UK
| | | | - Pascal Chanez
- Assistance Publique des Hôpitaux de Marseille, Clinique des Bronches, Allergies et Sommeil, Aix Marseille Université, Marseille, France
| | - Stephen J Fowler
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, and NIHR Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Ildiko Horvath
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | | | - Florian Singer
- Department of Respiratory Medicine, University Children's Hospital Zurich and Childhood Research Center, Zurich, and Department of Paediatrics, Inselspital, University of Bern, Switzerland
| | - Jacek Musial
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Barbro Dahlen
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Norbert Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Thomas Sandstrom
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | - Rene Lutter
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Louise J Fleming
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Peter H Howarth
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Massimo Caruso
- Department of Biochemical and Biotechnological Medicine, University of Catania, Catania, Italy
| | - Ana R Sousa
- Respiratory Therapeutic Unit, GSK, Stockley Park, UK
| | - Julie Corfield
- AstraZeneca R&D, Molndal, Sweden, and Areteva R&D, Nottingham, UK
| | - Charles Auffray
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Bertrand De Meulder
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Diane Lefaudeux
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Sven-Erik Dahlen
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Ratko Djukanovic
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Peter J Sterk
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | | |
Collapse
|
19
|
Hardtstock F, Krieger J, Wilke T, Lukas M, Ultsch B, Welte R, Quinzler R, Maywald U, Timmermann H. Use of Biologic Therapies in the Treatment of Asthma - A Comparative Real World Data Analysis on Healthcare Resource Utilization and Costs Before and After Therapy Initiation. J Asthma Allergy 2022; 15:407-418. [PMID: 35411151 PMCID: PMC8994596 DOI: 10.2147/jaa.s354062] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/04/2022] [Indexed: 12/24/2022] Open
Abstract
Background Asthma is one of the most common chronic diseases in Germany. While many patients achieve asthma control under standard therapies, some patients still experience exacerbations and persistent airway obstructions. Thus, further pharmacological treatment is needed, and biologics could fill this gap, as they have shown clinical benefit in patients with severe asthma. Therefore, this real-world study aimed to compare healthcare resource utilization (HCRU) and associated costs before and after biologic therapy initiation. Methods A retrospective claims data analysis has been conducted on adult asthma patients who initiated a long-term biologic therapy between January 2015 and June 2018. Patients were therapy-naïve to biologics for at least 12 months. HCRU and cost incurred by patients during 12 months before and after therapy initiation were compared. Results Overall, 571 asthma patients initiated a biologic therapy during the observational period (316 omalizumab, 232 mepolizumab, 16 benralizumab, and 7 reslizumab). Patients had a mean age of 54.86 (62.70% female), and the majority (93.70%) received at least one follow-up prescription of their index-biologic agent within one year. During baseline, patients received on average 2.75 OCS prescriptions compared to 2.17 during follow-up. Most patients received less or the same amount of OCS after therapy initiation. Moreover, hospitalizations and asthma-related sick leave days decreased significantly. The average total costs per patient were €6618.90 during baseline and €22,832.33 during follow-up. Biologics mainly drove the increase; however, hospitalization costs were reduced significantly (€2443.37 vs €1941.93; p<0.001). Conclusion Our study demonstrates an improved asthma control due to the initiation of a biologic therapy in terms of decreased hospitalization frequency, OCS consumption, and sick leave days. However, biologics are associated with high costs for healthcare providers during the first year after initiation. Therefore, short- and long-term clinical benefits and financial burden must be considered in the overall context of healthcare.
Collapse
Affiliation(s)
| | | | | | - Marco Lukas
- GlaxoSmithKline GmbH & Co. KG, München, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Melhorn J, Howell I, Pavord ID. Should we apply a treatable traits approach to asthma care? Ann Allergy Asthma Immunol 2022; 128:390-397. [PMID: 35172180 DOI: 10.1016/j.anai.2022.02.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To explore the evidence for adopting a "treatable traits" approach to asthma management. DATA SOURCES PubMed, Medline, and Google Scholar. STUDY SELECTIONS The above-mentioned databases were searched for randomized, controlled phase III or IV trials of adults containing the word "asthma" in the title published in the previous 10 years and for all articles containing the title words "treatable AND trait(s)," "asthma AND biomarker(s) OR smoking OR obesity OR laryngeal OR management" published within the previous 5 years. Articles were excluded if they were not published in English. Our search identified 257 articles for consideration. We also manually searched the reference lists of studies identified and searched the websites of the British Thoracic Society, European Respiratory Society, National Institute for Health and Care Excellence, and Global Initiative for Asthma for specific guidance related to asthma management. RESULTS The "treatable traits" are described within 3 domains of pulmonary, extrapulmonary, or behavioral and lifestyle traits. We consider whether treatment should be targeted toward these traits where they are present in asthma patients, based on currently available evidence, rather than increasing treatment in response to symptoms in line with current step-up, step-down asthma management guidelines. CONCLUSION We advocate that a treatable traits approach should be applied more broadly to the assessment and management of inadequately controlled asthma, rather than a step-up, step-down approach based on patient symptoms. This approach should be focused on the 2 treatable pulmonary traits of TH2 inflammation and airflow obstruction along with smoking cessation, in the first instance.
Collapse
Affiliation(s)
- James Melhorn
- Respiratory Medicine Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Imran Howell
- Respiratory Medicine Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ian D Pavord
- Respiratory Medicine Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
21
|
La-Crette J, Abdelhamid A, Chadwick A, Chakraborty A. Life-Threatening Bronchospasm. JRSM Open 2022; 13:20542704221086165. [PMID: 35295237 PMCID: PMC8918742 DOI: 10.1177/20542704221086165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
While Eosinophilic Asthma is frequently underdiagnosed, COPD is often misdiagnosed. This case focusses on a COPD misdiagnosis that had life-threatening consequences. The patient was a 59-year-old, male smoker, who presented to the Emergency Department with a week's history of increasing shortness of breath. On presentation, severe respiratory acidosis persisted acidotic despite Nebulisers, Oxygen, Steroids, and Magnesium. He was intubated for two weeks and had severe bronchospasm associated with type 2 respiratory failure. Eosinophils on admission were markedly elevated and remained so despite a week of intravenous steroids. As he missed the window for ECMO, we were advised to look at his diagnostic spirometry. Surprisingly, the spirometry done by his general practitioner, two years prior, showed Asthma not COPD. His blood eosinophils were elevated then, too. A revised diagnosis of Eosinophilic Asthma was given. Intravenous steroids were increased, and nebulised steroids were started. Soon thereafter, his condition improved, and he was stepped down from Intensive care. Hopefully, this case report increases physician knowledge of the different Asthma phenotypes and reduces incidences where correct treatment is only started during an avoidable life-threatening exacerbation.
Collapse
Affiliation(s)
| | | | - Andrew Chadwick
- Respiratory and Intensive Care Medicine Consultant, Oxford University Hospital Trust
| | - Arup Chakraborty
- Acute and Intensive Care Medicine Consultant, Milton Keynes University Hospital Trust
| |
Collapse
|
22
|
Pavord ID, Bel EH, Bourdin A, Chan R, Han JK, Keene ON, Liu MC, Martin N, Papi A, Roufosse F, Steinfeld J, Wechsler ME, Yancey SW. From DREAM to REALITI-A and beyond: Mepolizumab for the treatment of eosinophil-driven diseases. Allergy 2022; 77:778-797. [PMID: 34402066 PMCID: PMC9293125 DOI: 10.1111/all.15056] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022]
Abstract
Effective treatment of inflammatory diseases is often challenging owing to their heterogeneous pathophysiology. Understanding of the underlying disease mechanisms is improving and it is now clear that eosinophils play a complex pathophysiological role in a broad range of type 2 inflammatory diseases. Standard of care for these conditions often still includes oral corticosteroids (OCS) and/or cytotoxic immune therapies, which are associated with debilitating side effects. Selective, biological eosinophil‐reducing agents provide treatment options that improve clinical symptoms associated with eosinophilic inflammation and reduce OCS use. Mepolizumab is a humanized monoclonal antibody that binds to and neutralizes interleukin‐5, the major cytokine involved in eosinophil proliferation, activation, and survival. Mepolizumab is approved for the treatment of severe eosinophilic asthma, eosinophilic granulomatosis with polyangiitis and hypereosinophilic syndrome. Additionally, the efficacy of add‐on mepolizumab has been observed in patients with severe chronic rhinosinusitis with nasal polyposis and chronic obstructive pulmonary disease with an eosinophilic phenotype. Here, we review the development, approval, and real‐world effectiveness of mepolizumab for the treatment of patients with severe eosinophilic asthma, from the DREAM to REALITI‐A studies, and describe how knowledge from this journey extended to the use of mepolizumab and other biologics across a broad spectrum of eosinophilic diseases.
Collapse
Affiliation(s)
- Ian D. Pavord
- Nuffield Department of Medicine and Oxford Respiratory NIHR BRC University of Oxford Oxford UK
| | - Elisabeth H. Bel
- Department of Respiratory Medicine Amsterdam UMC University of Amsterdam Amsterdam The Netherlands
| | - Arnaud Bourdin
- INSERM 12 F‐CRIN Clinical Research Initiative In Severe Asthma: a Lever for Innovation & Science (CRISALIS) France
- Service de Pneumologie and INSERM CNRS CHU Montpellier Université de Montpellier Montpellier France
| | | | - Joseph K. Han
- Department of Otolaryngology, Head & Neck Surgery Eastern Virginia Medical School Norfolk Virginia USA
| | | | - Mark C. Liu
- Divisions of Allergy and Clinical Immunology, Pulmonary and Critical Care Medicine Johns Hopkins Asthma and Allergy Center Baltimore MD USA
| | - Neil Martin
- Global Medical Affairs GSK Brentford UK
- Institute for Lung Health University of Leicester Leicester UK
| | - Alberto Papi
- Research Center on Asthma and COPD University of Ferrara Ferrara Italy
| | - Florence Roufosse
- Department of Internal Medicine Hôpital Erasme Université Libre de Bruxelles Brussels Belgium
| | | | - Michael E. Wechsler
- Department of Medicine National Jewish Health Cohen Family Asthma Institute Denver CO USA
| | - Steven W. Yancey
- Respiratory Therapeutic Area Unit GSK Research Triangle Park NC USA
| |
Collapse
|
23
|
Ye SC, Desai S, Karlsen E, Kwong E, Wilcox PG, Quon BS. Association between elevated peripheral blood eosinophil count and respiratory outcomes in adults with cystic fibrosis. J Cyst Fibros 2022; 21:1048-1052. [DOI: 10.1016/j.jcf.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022]
|
24
|
Lombardi C, Berti A, Cottini M. The emerging roles of eosinophils: Implications for the targeted treatment of eosinophilic-associated inflammatory conditions. CURRENT RESEARCH IN IMMUNOLOGY 2022; 3:42-53. [PMID: 35496822 PMCID: PMC9040157 DOI: 10.1016/j.crimmu.2022.03.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/06/2022] [Accepted: 03/07/2022] [Indexed: 01/20/2023] Open
Abstract
Eosinophils have multiple relevant biological functions, including the maintenance of homeostasis, host defense against infectious agents, innate immunity activities, immune regulation through Th1/Th2 balance, anti-inflammatory, and anti-tumorigenic effects. Eosinophils also have a main role in tissue damage through eosinophil-derived cytotoxic mediators that are involved in eosinophilic inflammation, as documented in Th2-high asthma and other eosinophilic-associated inflammatory conditions. Recent evidence shows that these multiple and apparently conflicting functions may be attributed to the existence of different eosinophil subtypes (i.e.: tissue resident and inducible eosinophils). Therapeutic intervention with biological agents that totally deplete tissues and circulating eosinophils or, vice versa, maintain a minimal proportion of eosinophils, particularly the tissue-resident ones, could therefore have a very different impact on patients, especially when considering the administration of these therapies for prolonged time. In addition, the characterization of the predominant pathway underlying eosinophilic inflammation by surrogate biomarkers (circulating eosinophils, organ-specific eosinophils levels such as eosinophil count in sputum, bronchoalveolar lavage, tissue biopsy; total circulating IgE levels, or the use of FeNO) in the single patient with an eosinophilic-associated inflammatory condition could help in choosing the treatment. These observations are crucial in light of the increasing therapeutic armamentarium effective in modulating eosinophilic inflammation through the inhibition in different, yet complementary ways of eosinophil pathways, such as the interleukin-5 one (with mepolizumab, benralizumab, reslizumab) or the interleukin-4/13 one (with dupilumab and lebrikizumab), in severe T2-high asthma as well as in other systemic eosinophilic associated diseases, such as eosinophilic granulomatosis with polyangiitis and hypereosinophilic syndrome. Recent evidence pointed out the existence of different eosinophil subtypes, i.e. tissue resident and inducible eosinophils, with different and apparently conflicting functions. Biological therapies with different mechanisms can deplete completely tissues and circulating eosinophils or maintain a minimal proportion of eosinophils, particularly the tissue-resident ones, and this could therefore have a different impact on patients, especially when considering the administration of these therapies for prolonged time. The identification of the predominant pathway underlying eosinophilic inflammation by surrogate biomarkers (circulating eosinophils, organ-specific eosinophils levels such as eosinophil count in sputum, bronchoalveolar lavage, tissue biopsy; total circulating IgE levels, or the use of FeNO) should be sought in the single patient with an eosinophilic-associated inflammatory condition. These considerations may help in choosing the best anti-eosinophilic treatment, considering the increasing therapeutic armamentarium effective in modulating eosinophilic inflammation through the inhibition of the interleukin-5 one (with mepolizumab, benralizumab, reslizumab) or the interleukin-4/13 one (with dupilumab and lebrikizumab)
Collapse
Affiliation(s)
- Carlo Lombardi
- Departmental Unit of Allergology, Immunology & Pulmonary Diseases, Fondazione Poliambulanza, Brescia, Italy
- Corresponding author. Departmental Unit of Pneumology & Allergology, Fondazione Poliambulanza Istituto Ospedaliero, Via Bissolati, 57 Brescia, 25100, Italy.
| | - Alvise Berti
- Ospedale Santa Chiara and Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Thoracic Disease Research Unit, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
25
|
Pérez de Llano LA, Cosío BG, Lobato Astiárraga I, Soto Campos G, Tejedor Alonso MÁ, Marina Malanda N, Padilla Galo A, Urrutia Landa I, Michel de la Rosa FJ, García-Moguel I. Asthma Control in Patients with Severe Eosinophilic Asthma Treated with Reslizumab: Spanish Real-Life Data. J Asthma Allergy 2022; 15:79-88. [PMID: 35058696 PMCID: PMC8765543 DOI: 10.2147/jaa.s340562] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/23/2021] [Indexed: 12/17/2022] Open
Abstract
Background Reslizumab is an anti-interleukin 5 monoclonal antibody that has demonstrated to reduce the risk of severe exacerbations and to improve symptoms, lung function, and quality of life in randomized controlled trials that included patients with severe eosinophilic uncontrolled asthma (SEUA) and a history of severe exacerbations. Objective The aim of the present study was to evaluate the effectiveness of add-on reslizumab in a cohort of patients with SEUA under real-life conditions. Methods This was a multi-centre, retrospective, real-life study that included subjects with SEUA treated with reslizumab in 44 asthma units throughout Spain. Eligible patients were those who had received at least one dose of reslizumab as part of normal clinical practice. The primary endpoint was complete asthma control at 52 weeks, defined as absence of severe exacerbations, ACT ≥20 and no maintenance oral corticosteroids (OCS). Demographic, clinical, and functional data were collected at baseline (T0), after four to six months (T1); after 12 months (T2) and beyond 12 months of therapy (T3). Results Treatment with reslizumab achieved complete asthma control in 40% of the 208 included SEUA patients and led to a significant reduction in exacerbations (from 3.0; IQR: 2.0–4.0 at V0 to 0.0; IQR: 0.0–0.0 at V2), maintenance OCS use (from 54.8% (95% CI: 48.0–61.6 at T0 to 18.5% (95% CI: 12.5–24.5 at T2) and a meaningful improvement in symptoms in the entire treated population: ACT increased from 12.8 ± 4.5 at V0 to 20.0 ± 5.1 at V2 (p < 0.001). Most of the improvement achieved at 12 months was obtained at 4–6 months. The retention (continuation) rate of reslizumab was 75% through 2 years (95CI%: 1.9–2.1). Overall, reslizumab showed an adequate safety profile. Conclusion Reslizumab is an effective therapy for SEUA with adequate safety profile in real-life conditions.
Collapse
Affiliation(s)
| | - Borja G Cosío
- Pneumology Service, Hospital Universitario Son Espases-IdISBa-Ciberes, Palma de Mallorca, Spain
- Correspondence: Borja G Cosío Department of Respiratory Medicine, Hospital Universitario Son ESPASES-IDISBA, Palma de Mallorca, Spain Email
| | | | - Gregorio Soto Campos
- Pneumology and Allergy Unit, University Hospital of Jerez, Jerez de la Frontera, Cádiz, Spain
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Benson VS, Hartl S, Barnes N, Galwey N, Van Dyke MK, Kwon N. Blood eosinophil counts in the general population and airways disease: a comprehensive review and meta-analysis. Eur Respir J 2022; 59:2004590. [PMID: 34172466 PMCID: PMC8756293 DOI: 10.1183/13993003.04590-2020] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The clinical context for using blood eosinophil (EOS) counts as treatment-response biomarkers in asthma and COPD requires better understanding of EOS distributions and ranges. We describe EOS distributions and ranges published in asthma, COPD, control (non-asthma/COPD) and general populations. METHODS We conducted a comprehensive literature review and meta-analysis of observational studies (January 2008 to November 2018) that included EOS counts in asthma, severe asthma, COPD, control and general populations. Excluded studies had total sample sizes <200, EOS as inclusion criterion, hospitalised population only and exclusively paediatric participants. RESULTS Overall, 91 eligible studies were identified, most had total-population-level data available: asthma (39 studies), severe asthma (12 studies), COPD (23 studies), control (seven studies) and general populations (14 studies); some articles reported data for multiple populations. Reported EOS distributions were right-skewed (seven studies). Reported median EOS counts ranged from 157-280 cells·µL-1 (asthma, 22 studies); 200-400 cells·µL-1 (severe asthma, eight studies); 150-183 cells·µL-1 (COPD, six studies); and 100-160 cells·µL-1 (controls, three studies); and 100-200 cells·µL-1 (general populations, six studies). The meta-analysis showed that observed variability was mostly between studies rather than within studies. Factors reportedly associated with higher blood EOS counts included current smoking, positive skin-prick test, elevated total IgE, comorbid allergic rhinitis, age ≤18 years, male sex, spirometric asthma/COPD diagnosis, metabolic syndrome and adiposity. CONCLUSION EOS distribution and range varied by study population, and were affected by clinical factors including age, smoking history and comorbidities, which, regardless of severity, should be considered during treatment decision-making.
Collapse
Affiliation(s)
- Victoria S Benson
- Epidemiology, Value Evidence and Outcomes (VEO), Global Medical R&D, GlaxoSmithKline, Brentford, UK
| | - Sylvia Hartl
- Dept of Respiratory and Critical Care Medicine and Ludwig Boltzmann Institute for Lung Health, Clinic Penzing, WiGev and Sigmund Freud University, Medical School, Vienna, Austria
| | - Neil Barnes
- Respiratory Therapy Area, GlaxoSmithKline, Brentford, UK
- William Harvey Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | | | - Melissa K Van Dyke
- Epidemiology, Value Evidence and Outcomes (VEO), Global Medical R&D, GlaxoSmithKline, Upper Providence, PA, USA
| | - Namhee Kwon
- Respiratory Research and Development, GlaxoSmithKline, Brentford, UK
| |
Collapse
|
27
|
Maglio A, Vitale C, Pellegrino S, Calabrese C, D’Amato M, Molino A, Pelaia C, Triggiani M, Pelaia G, Stellato C, Vatrella A. Real-Life Effectiveness of Mepolizumab on Forced Expiratory Flow between 25% and 75% of Forced Vital Capacity in Patients with Severe Eosinophilic Asthma. Biomedicines 2021; 9:1550. [PMID: 34829778 PMCID: PMC8615088 DOI: 10.3390/biomedicines9111550] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 01/20/2023] Open
Abstract
Severe eosinophilic asthma (SEA) is associated with high peripheral blood and airway eosinophilia, recurrent disease exacerbations and severe airflow limitation. Eosinophilic inflammation is also responsible for small airway disease (SAD) development. SEA patients experience poor disease control and response to standard therapy and are prime candidates for anti-IL5 biologicals, such as mepolizumab, but the effect of treatment on SAD is unclear. We investigated the effect of mepolizumab on lung function in SEA patients, focusing on SAD parameters, and searched for an association between patients' phenotypic characteristics and changes in small airways function. In this real-life study, data from 105 patients with SEA were collected at baseline and after 6, 12 and 18 months of mepolizumab treatment. Along with expected improvements in clinical and lung function parameters brought by Mepolizumab treatment, FEF2525-75% values showed a highly significant, gradual and persistent increase (from 32.7 ± 18.2% at baseline to 48.6 ± 18.4% after 18 months) and correlated with ACT scores at 18 months (r = 0.566; p ≤ 0.0001). A patient subgroup analysis showed that changes in FEF25-75% values were higher in patients with a baseline peripheral blood eosinophil count ≥400 cells/μL and oral corticosteroid use. Mepolizumab significantly improves small airway function. This effect correlates with clinical benefits and may represent an accessible parameter through which to evaluate therapeutic response. This study provides novel insights into the phenotypic characteristics associated with the improved functional outcome provided by mepolizumab treatment.
Collapse
Affiliation(s)
- Angelantonio Maglio
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84100 Salerno, Italy; (A.M.); (C.V.); (S.P.); (M.T.); (C.S.)
| | - Carolina Vitale
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84100 Salerno, Italy; (A.M.); (C.V.); (S.P.); (M.T.); (C.S.)
| | - Simona Pellegrino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84100 Salerno, Italy; (A.M.); (C.V.); (S.P.); (M.T.); (C.S.)
| | - Cecilia Calabrese
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80100 Naples, Italy;
| | - Maria D’Amato
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80100 Naples, Italy; (M.D.); (A.M.)
| | - Antonio Molino
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80100 Naples, Italy; (M.D.); (A.M.)
| | - Corrado Pelaia
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (C.P.); (G.P.)
| | - Massimo Triggiani
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84100 Salerno, Italy; (A.M.); (C.V.); (S.P.); (M.T.); (C.S.)
| | - Girolamo Pelaia
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (C.P.); (G.P.)
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84100 Salerno, Italy; (A.M.); (C.V.); (S.P.); (M.T.); (C.S.)
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84100 Salerno, Italy; (A.M.); (C.V.); (S.P.); (M.T.); (C.S.)
| |
Collapse
|
28
|
Hassoun D, Malard O, Barbarot S, Magnan A, Colas L. Type 2 immunity-driven diseases: Towards a multidisciplinary approach. Clin Exp Allergy 2021; 51:1538-1552. [PMID: 34617355 PMCID: PMC9292742 DOI: 10.1111/cea.14029] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022]
Abstract
Asthma, atopic dermatitis and chronic rhinoconjunctivitis are highly heterogeneous. However, epidemiologic associations exist between phenotypic groups of patients. Atopic march is one such association but is not the only common point. Indeed, beyond such phenotypes, hallmarks of type 2 immunity have been found in these diseases involving immune dysregulation as well as environmental triggers and epithelial dysfunction. From the canonical Th2 cytokines (IL-4, IL-5, IL-13), new cellular and molecular actors arise, from the epithelium's alarmins to new innate immune cells. Their interactions are now better understood across the different environmental barriers, and slight differences appeared. In parallel, the development of type 2-targeting biotherapies not only raised hope to treat those diseases but also raised new questions regarding their true pathophysiological involvement. Here, we review the place of type 2 immunity in the different phenotypes of asthma, chronic rhinitis, chronic rhinosinusitis and atopic dermatitis, highlighting nuances between them. New hypotheses rising from the use of biotherapies will be discussed along with the uncertainties and unmet needs of this field.
Collapse
Affiliation(s)
- Dorian Hassoun
- CHU Nantes, CNRS, INSERM, l'institut du Thorax, Université de Nantes, Nantes, France
| | - Olivier Malard
- Department of Otorhinolaryngology and Head and Neck Surgery, Nantes University Hospital, Nantes, France
| | - Sébastien Barbarot
- Department of Dermatology, CHU Nantes, UMR 1280 PhAN, INRA, Nantes Université, Nantes, France
| | - Antoine Magnan
- INRAe UMR_S 0892, Hôpital Foch, Université de Versailles Saint-Quentin, Paris Saclay, France
| | - Luc Colas
- Plateforme Transversale d'Allergologie et d'Immunologie Clinique, Institut du Thorax, CHU de Nantes, Nantes, France.,INSERM, CHU Nantes, Centre de Recherche en Transplantation et Immunologie UMR1064, Nantes Université, ITUN, Nantes, France
| |
Collapse
|
29
|
Hosking L, Yeo A, Hoffman J, Chiano M, Fraser D, Ghosh S, Lipson DA, Martin N, Condreay LD, Cox C, St Jean P. Genetics plays a limited role in predicting chronic obstructive pulmonary disease treatment response and exacerbation. Respir Med 2021; 187:106573. [PMID: 34428673 DOI: 10.1016/j.rmed.2021.106573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 07/27/2021] [Accepted: 08/08/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Combination treatments, targeting multiple disease processes, benefit subjects with acute exacerbation of chronic obstructive pulmonary disease (AECOPD). However, predicting treatment response and exacerbation risk remain challenging. OBJECTIVE To identify genetic associations with AECOPD risk and response to combination therapy (fluticasone furoate, umeclidinium bromide and vilanterol). METHODS The genetic basis of AECOPD disease was investigated in 19,841 subjects from 23 clinical studies and 2 disease cohorts to identify exacerbation disease targets. AECOPD pharmacogenetic effects were examined in 8439 moderate to severe COPD patients with exacerbation rate, lung function and quality of life endpoints; results were followed up in an additional 2201 subjects. RESULTS We did not identify significant associations in the AECOPD disease analysis. In the AECOPD pharmacogenetics analysis, rs56195836 (MAPK8) was significantly associated with moderate to severe exacerbation rate in subjects on fluticasone furoate with baseline blood eosinophils ≥150 cells/μl (P = 1.8 × 10-8). Post-hoc, one variant was associated with on-treatment moderate to severe exacerbation rate stratifying by exacerbation history. AZU1 rs1962343 was significantly associated in subjects with frequent moderate exacerbation history when treated with fluticasone furoate/vilanterol (P = 1.1 × 10-8). Neither of these signals was supported in independent follow-up. CONCLUSION Common genetic variants do not play major roles in AECOPD disease nor predict response to triple therapy or its components in moderate to very severe COPD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - David A Lipson
- GSK, Collegeville, PA, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Neil Martin
- GSK, Brentford, Middlesex, UK; University of Leicester, Leicester, Leicestershire, UK.
| | | | | | | |
Collapse
|
30
|
van der Meer AN, de Jong K, Hoekstra-Kuik A, Bel EH, Ten Brinke A. Targeting dynamic hyperinflation in moderate-to-severe asthma: a randomised controlled trial. ERJ Open Res 2021; 7:00738-2020. [PMID: 34409093 PMCID: PMC8365142 DOI: 10.1183/23120541.00738-2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/23/2021] [Indexed: 12/02/2022] Open
Abstract
Background Dynamic hyperinflation is highly prevalent in moderate-to-severe asthma, which may significantly impede activities of daily life. We hypothesised that dynamic hyperinflation in asthma is due to inflammation of large and small airways and can be reduced by systemic anti-inflammatory treatment. Therefore, we investigated the effect of systemic glucocorticoids on dynamic hyperinflation in moderate-to-severe asthma patients and explored the relationships between inflammatory markers and changes in dynamic hyperinflation. Methods In this randomised placebo-controlled trial we included 32 asthma patients on inhaled glucocorticoid therapy showing dynamic hyperinflation, defined by a ≥10% reduction in inspiratory capacity measured by standardised metronome-paced tachypnea test. Patients received either triamcinolone (80 mg) or placebo intramuscularly. Before and 2 weeks after treatment, patients completed respiratory health questionnaires, had blood eosinophils and exhaled nitric oxide levels measured, and underwent lung function and dynamic hyperinflation testing. Results After adjustment for potential confounders, dynamic hyperinflation was significantly reduced by 28.1% in the triamcinolone group and increased by 9.4% in the placebo group (p=0.027). In the triamcinolone-treated patients, the reduction in dynamic hyperinflation was greater in patients with higher blood eosinophils at baseline (r=−0.592, p=0.020) and tended to be associated with a reduction in blood eosinophils (r=0.412, p=0.127) and exhaled nitric oxide (r=0.442, p=0.099). Conclusions This exploratory study suggests that dynamic hyperinflation in asthma can be reduced by systemic anti-inflammatory treatment, particularly in patients with elevated blood eosinophils. This supports the hypothesis that dynamic hyperinflation in asthma is due to airway inflammation and should be considered an important target for treatment. Dynamic hyperinflation: a target for treatment in asthma, which can be reduced by systemic anti-inflammatory treatmenthttps://bit.ly/3gfyxYv
Collapse
Affiliation(s)
| | - Kim de Jong
- Medical Centre Leeuwarden, Leeuwarden, The Netherlands
| | | | - Elisabeth H Bel
- Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | | |
Collapse
|
31
|
Principe S, Porsbjerg C, Bolm Ditlev S, Kjaersgaard Klein D, Golebski K, Dyhre-Petersen N, van Dijk YE, van Bragt JJMH, Dankelman LLH, Dahlen SE, Brightling CE, Vijverberg SJH, Maitland-van der Zee AH. Treating severe asthma: Targeting the IL-5 pathway. Clin Exp Allergy 2021; 51:992-1005. [PMID: 33887082 PMCID: PMC8453879 DOI: 10.1111/cea.13885] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/05/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023]
Abstract
Severe asthma is a heterogeneous disease with different phenotypes based on clinical, functional or inflammatory parameters. In particular, the eosinophilic phenotype is associated with type 2 inflammation and increased levels of interleukin (IL)-4, IL-5 and IL-13). Monoclonal antibodies that target the eosinophilic inflammatory pathways (IL-5R and IL-5), namely mepolizumab, reslizumab, and benralizumab, are effective and safe for severe eosinophilic asthma. Eosinophils threshold represents the most indicative biomarker for response to treatment with all three monoclonal antibodies. Improvement in asthma symptoms scores, lung function, the number of exacerbations, history of late-onset asthma, chronic rhinosinusitis with nasal polyposis, low oral corticosteroids use and low body mass index represent predictive clinical markers of response. Novel Omics studies are emerging with proteomics data and exhaled breath analyses. These may prove useful as biomarkers of response and non-response biologics. Moreover, future biomarker studies need to be undertaken in paediatric patients affected by severe asthma. The choice of appropriate biologic therapy for severe asthma remains challenging. The importance of finding biomarkers that can predict response continuous an open issue that needs to be further explored. This review describes the clinical effects of targeting the IL-5 pathway in severe asthma in adult and paediatric patients, focusing on predictors of response and non-response.
Collapse
Affiliation(s)
- Stefania Principe
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Dipartimento Universitario di Promozione della Salute, Materno Infantile, Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro"(PROMISE) c/o Pneumologia, University of Palermo, Palermo, Italy.,AOUP "Policlinico Paolo Giaccone", Palermo, Italy
| | - Celeste Porsbjerg
- Respiratory Research Unit, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Sisse Bolm Ditlev
- Respiratory Research Unit, Bispebjerg University Hospital, Copenhagen, Denmark
| | | | - Korneliusz Golebski
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Yoni E van Dijk
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Pulmonology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Job J M H van Bragt
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lente L H Dankelman
- Department of Pediatric Pulmonology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sven-Erik Dahlen
- The Institute of Environmental Medicine Department of Medicine, Solna Campus, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,The Center for Allergy Research, Department of Medicine, Solna Campus, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Susanne J H Vijverberg
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Pulmonology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anke H Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Pulmonology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Bai H, Sha B, Xu X, Yu L. Gender Difference in Chronic Cough: Are Women More Likely to Cough? Front Physiol 2021; 12:654797. [PMID: 34025449 PMCID: PMC8138462 DOI: 10.3389/fphys.2021.654797] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/14/2021] [Indexed: 11/15/2022] Open
Abstract
Chronic cough is a common complaint for patients to seek medical cares all over the world. Worldwide, about two thirds of chronic cough patients are females. However, in some regions of China the prevalence of chronic cough between sexes is roughly the same. Estrogen and progesterone can not only have an effect on transient receptor potential vanilloid 1 channel, eosinophils and mast cells, but also influence laryngeal dysfunction, gastroesophageal reflux disease and obstructive sleep apnea hypopnea syndrome, which may lead to increased cough sensitivity in women. On the other hand, the quality of life was adversely affected more in female patients with chronic cough. Both hormones possibly cause gender difference in chronic cough.
Collapse
Affiliation(s)
| | | | - Xianghuai Xu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Yu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Jiang Y, An R, Cheng L, Yue Q, Zhang H, Zhang Y, Kong X, Ma H, Chen F, Guo Y. Classification of non-acute bronchial asthma according to allergy and eosinophil characteristics: a retrospective study. Allergy Asthma Clin Immunol 2021; 17:45. [PMID: 33941235 PMCID: PMC8091754 DOI: 10.1186/s13223-021-00546-1] [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: 12/15/2020] [Accepted: 04/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Investigating the endotypes of the different asthma phenotypes would help disease monitoring, prognosis determination, and improving asthma management standardization. This study aimed to classify asthma into four endotypes according to the allergic and eosinophilic characteristics and explore the phenotypes (clinical characteristics, pulmonary functions, and fractional expired nitric oxide (FeNO)) of each endotype. METHODS This retrospective study included non-acute asthma patients treated at the First Hospital of Shanxi Medical University (05/2016-01/2018). The patients were classified into the eosinophilic allergic, eosinophilic non-allergic, non-eosinophilic allergic, and non-eosinophilic non-allergic asthma endotypes. Serum sIgE, lung function, FeNO, and induced sputum cytology were tested and compared among groups. RESULTS Of the 171 included patients, 22 had eosinophilic allergic asthma, 17 had eosinophilic non-allergic asthma, 66 had non-eosinophilic allergic asthma, and 66 had non-eosinophilic non-allergic asthma. Lung function measurements (FEV1%, FEF25%, FEF50%, FEF75%, and FEF25-75%) showed that airway dysfunction was worse in eosinophilic non-allergic asthma than in the other three endotypes (all P < 0.001). In allergic asthma patients, eosinophilic asthma had worse airway dysfunction than non-eosinophilic asthma (all P < 0.05). Similar results were found in non-allergic asthma (all P < 0.01). The FeNO levels in eosinophilic allergic asthma were higher than in eosinophilic non-allergic and non-eosinophilic non-allergic asthma (both P = 0.001). CONCLUSIONS FeNO can objectively reflect eosinophilic airway inflammation in asthma. Endotypic classification of asthma patients regarding the allergic and eosinophilic characteristics is conducive to the effective management of patients with asthma.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Ruoli An
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Cheng
- General Internal Medicine Department, People's Hospital of Wenshui County, Shanxi Province, Wenshui, Shanxi, China
| | - Qianru Yue
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hanwei Zhang
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yali Zhang
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaomei Kong
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongxia Ma
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fang Chen
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yufeng Guo
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
34
|
Laidlaw TM, Bachert C, Amin N, Desrosiers M, Hellings PW, Mullol J, Maspero JF, Gevaert P, Zhang M, Mao X, Khan AH, Kamat S, Patel N, Graham NMH, Ruddy M, Staudinger H, Mannent LP. Dupilumab improves upper and lower airway disease control in chronic rhinosinusitis with nasal polyps and asthma. Ann Allergy Asthma Immunol 2021; 126:584-592.e1. [PMID: 33465455 DOI: 10.1016/j.anai.2021.01.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps (CRSwNP) and type 2 asthma share the same inflammatory pathophysiology and are frequent comorbidities. Dupilumab, a fully human monoclonal antibody, blocks the shared receptor component for interleukin 4 and interleukin 13, which are key and central drivers of type 2 inflammation. OBJECTIVE We report the effect of dupilumab vs placebo on outcome measures of the upper and lower airways and health-related quality of life (HRQoL) in the pooled population of patients with CRSwNP and comorbid asthma from the phase 3 SINUS-24 (NCT02912468) and SINUS-52 (NCT02898454) studies. METHODS In these randomized, double-blind, placebo-controlled trials, patients received subcutaneous dupilumab 300 mg (n = 438) or placebo (n = 286) every 2 weeks on a background of mometasone furoate nasal spray. Changes from baseline at week 24 in the upper and lower airway outcome measures are reported. RESULTS Of the 724 patients randomized, 428 (59.1%) had comorbid asthma. In patients with asthma at week 24, dupilumab vs placebo improved the nasal polyp score (-2.04), patient-reported nasal congestion score (-1.04), Lund-Mackay computed tomography scan score (-6.43), peak nasal inspiratory flow (46.15 L/min), and 22-item sinonasal outcome test score (-21.42; all P < .001). The forced expiratory volume in 1 second and 6-item asthma control questionnaire scores were also markedly improved with dupilumab vs placebo. The most common adverse events (nasopharyngitis, headache, injection-site erythema, worsening of nasal polyposis, and asthma) were more frequent with placebo than dupilumab. CONCLUSION Dupilumab improved upper and lower airway outcome measures and HRQoL in patients with severe CRSwNP and comorbid asthma and was well tolerated. TRIAL REGISTRATION ClinicalTrials.gov Identifiers: NCT02912468 (SINUS-24) and NCT02898454 (SINUS-52).
Collapse
Affiliation(s)
- Tanya M Laidlaw
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Claus Bachert
- Department of Head & Skin, Upper Airways Research Laboratory, Faculty of Medicine, Ghent University, Ghent, Belgium; Division of Ear, Nose and Throat (ENT) Diseases, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Nikhil Amin
- Regeneron Pharmaceuticals Inc, Tarrytown, New York
| | - Martin Desrosiers
- Department of Otolaryngology, Centre de recherche du Centre hospitalier de l'Université de Montreal (CRCHUM), Montreal, Quebec, Canada
| | - Peter W Hellings
- Clinical Division of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Joaquim Mullol
- Rhinology Unit & Smell Clinic, Department of Otorhinolaryngology, Hospital Clínic, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Universitat de Barcelona, Ciber De Enfermedades Respiratorias (CIBERES), Barcelona, Catalonia, Spain
| | - Jorge F Maspero
- Allergy and Respiratory Research Unit, Fundación CIDEA, Buenos Aires, Argentina
| | - Philippe Gevaert
- Department of Head & Skin, Upper Airways Research Laboratory, Faculty of Medicine, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jarrell L. Eosinophilic asthma and the role of monoclonal antibodies. Nurse Pract 2021; 46:21-27. [PMID: 33739323 DOI: 10.1097/01.npr.0000737188.21326.c1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Eosinophilic asthma presents with continuous airway inflammation resistant to inhaled corticosteroids but responsive to oral glucocorticoids and monoclonal antibodies. Diagnostic criteria include significantly elevated blood or sputum eosinophils and/or fractional exhaled nitric oxide. Five monoclonal antibodies are used for treatment, with accurate diagnosis and early intervention essential to better outcomes.
Collapse
Affiliation(s)
- Lynda Jarrell
- Lynda Jarrell is a clinical assistant professor at the University of Texas at Arlington, Arlington, Tex
| |
Collapse
|
36
|
Denton E, Price DB, Tran TN, Canonica GW, Menzies-Gow A, FitzGerald JM, Sadatsafavi M, Perez de Llano L, Christoff G, Quinton A, Rhee CK, Brusselle G, Ulrik C, Lugogo N, Hore-Lacy F, Chaudhry I, Bulathsinhala L, Murray RB, Carter VA, Hew M. Cluster Analysis of Inflammatory Biomarker Expression in the International Severe Asthma Registry. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:2680-2688.e7. [PMID: 33744476 DOI: 10.1016/j.jaip.2021.02.059] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Allergy, eosinophilic inflammation, and epithelial dysregulation are implicated in severe asthma pathogenesis. OBJECTIVE We characterized biomarker expression in adults with severe asthma. METHODS Within the International Severe Asthma Registry (ISAR), we analyzed data from 10 countries in North America, Europe, and Asia, with prespecified thresholds for biomarker positivity (serum IgE ≥ 75 kU/L, blood eosinophils ≥ 300 cells/μL, and FeNO ≥ 25 ppb), and with hierarchical cluster analysis using biomarkers as continuous variables. RESULTS Of 1,175 patients; 64% were female, age (mean ± SD) 53 ± 15 years, body mass index (BMI) 30 ± 8, postbronchodilator forced expiratory volume in 1 second (FEV1) predicted 72% ± 20%. By prespecified thresholds, 59% were IgE positive, 57% eosinophil positive, and 58% FeNO positive. There was substantial inflammatory biomarker overlap; 59% were positive for either 2 or 3 biomarkers. Five distinct clusters were identified: cluster 1 (61%, low-to-medium biomarkers) comprised highly symptomatic, older females with elevated BMI and frequent exacerbations; cluster 2 (18%, elevated eosinophils and FeNO) older females with lower BMI and frequent exacerbations; cluster 3 (14%, extremely high FeNO) older, highly symptomatic, lower BMI, and preserved lung function; cluster 4 (6%, extremely high IgE) younger, long duration of asthma, elevated BMI, and poor lung function; cluster 5 (1.2%, extremely high eosinophils) younger males with low BMI, poor lung function, and high burden of sinonasal disease and polyposis. CONCLUSIONS There is significant overlap of biomarker positivity in severe asthma. Distinct clusters according to biomarker expression exhibit unique clinical characteristics, suggesting the occurrence of discrete patterns of underlying inflammatory pathway activation and providing pathogenic insights relevant to the era of monoclonal biologics.
Collapse
Affiliation(s)
- Eve Denton
- Allergy, Asthma, and Clinical Immunology, Alfred Health, Melbourne, Australia; Public Health and Preventive Medicine, Monash University, Melbourne, Australia.
| | - David B Price
- Optimum Patient Care, Cambridge, UK; Observational and Pragmatic Research Institute, Singapore, Singapore; Centre of Academic Primary Care, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | | | - G Walter Canonica
- Personalized Medicine, Asthma and Allergy, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Andrew Menzies-Gow
- UK Severe Asthma Network and National Registry, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - J Mark FitzGerald
- The Centre for Heart Lung Health, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Mohsen Sadatsafavi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Luis Perez de Llano
- Department of Respiratory Medicine, Hospital Universitario Lucus Augusti, Lugo, Spain
| | - George Christoff
- Faculty of Public Health, Medical University of Sofia, Sofia, Bulgaria
| | | | - Chin Kook Rhee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Guy Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium; Department of Epidemiology and Respiratory Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Charlotte Ulrik
- Department of Respiratory Medicine, Hvidovre Hospital, Hvidovre, Denmark
| | - Njira Lugogo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Mich
| | - Fiona Hore-Lacy
- Allergy, Asthma, and Clinical Immunology, Alfred Health, Melbourne, Australia; Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | | | | | | | | | - Mark Hew
- Allergy, Asthma, and Clinical Immunology, Alfred Health, Melbourne, Australia; Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
37
|
Bendien SA, van Loon-Kooij S, Kramer G, Huijgen W, Altenburg J, Ten Brinke A, Maitland-van der Zee AH. Bronchiectasis in Severe Asthma: Does It Make a Difference? Respiration 2020; 99:1-9. [PMID: 33321494 DOI: 10.1159/000511459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 08/31/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Asthma and bronchiectasis are 2 heterogeneous diseases that frequently coexist, particularly in severe asthma. Recognition of this co-diagnosis may importantly affect treatment decisions and outcome. Previous studies in asthma with bronchiectasis show inconsistent outcomes, probably due to the heterogeneity of the included asthma cohorts. OBJECTIVES We hypothesized that bronchiectasis contributes to asthma severity and that patients with severe asthma and bronchiectasis present with distinct characteristics resulting in different treatable traits. In addition, we explored whether bronchiectasis in severe asthma is more common in a specific phenotype. METHODS This is a single-center study consecutively including patients with severe asthma from a tertiary referral center. Severe asthma was diagnosed according to the ATS/ERS guidelines. Asthma and infectious exacerbations were defined by the attending specialist as respiratory symptoms requiring treatment with systemic steroids or antibiotics, respectively. Two independent blinded radiologists evaluated each CT. RESULTS 19% of patients with severe asthma showed bronchiectasis on CT. Patients with bronchiectasis had a lower FEV1% predicted (p = 0.02) and FEV1/FVC (p = 0.004) and more infectious exacerbations (p = 0.003) compared to patients without bronchiectasis. Bronchiectasis is more common in patients with a longer duration of asthma, sensitization to A. fumigatus or a positive sputum culture. Sputum cultures of patients with severe asthma and bronchiectasis revealed more P. aeruginosa, S. maltophilia, H. parainfluenzae, and A. fumigates compared to the non-bronchiectasis group. The adult-onset, eosinophilic asthma phenotype showed the highest prevalence of bronchiectasis (29.4%). CONCLUSIONS Patients with severe asthma and coexisting bronchiectasis were found to represent a distinct group, in terms of disease severity, microbiology, and asthma phenotype. Performing (HR)CT and sputum cultures can help to identify these patients. These results can possibly contribute to early recognition and targeted treatment of this patient group.
Collapse
Affiliation(s)
- Sarah Alice Bendien
- Department of Respiratory Medicine, Haga Teaching Hospital, The Hague, The Netherlands,
| | | | - Gerdien Kramer
- Department of Radiology, Martini Hospital, Groningen, The Netherlands
| | - Willemijn Huijgen
- Department of Radiology, Haga Teaching Hospital, The Hague, The Netherlands
| | - Josje Altenburg
- Department of Respiratory Medicine, Amsterdam University Medical Center (Amsterdam UMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Anneke Ten Brinke
- Department of Respiratory Medicine, Medical Centre Leeuwarden, Leeuwarden, The Netherlands
| | - Anke-Hilse Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam University Medical Center (Amsterdam UMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Richness of sputum microbiome in acute exacerbations of eosinophilic chronic obstructive pulmonary disease. Chin Med J (Engl) 2020; 133:542-551. [PMID: 32053571 PMCID: PMC7065869 DOI: 10.1097/cm9.0000000000000677] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The eosinophilic chronic obstructive pulmonary disease (COPD) is known to be more sensitive to corticosteroid. The sputum microbiome has been shown to affect COPD prognosis, but its role in acute exacerbations of eosinophilic COPD is unclear. This study aimed to investigate the dynamic changes of the airway microbiome in patients with acute exacerbations of eosinophilic COPD. METHODS Fifty-seven patients with acute exacerbations of COPD from the First Affiliated Hospital of Guangxi Medical University between June 2017 and June 2018 were divided into two groups. Patients with eosinophils ≥300 cells/μL in the peripheral venous blood were assigned to the eosinophilic group (Eos) and the rest to the non-eosinophilic group (Noneos). All patients received similar treatment including inhaled budesonide according to the guidelines. The induced sputum microbiome was analyzed on the 1st and 7th day of treatment using the 16S ribosomal RNA (rRNA) method. The levels of interleukin (IL)-6 and IL-8 were measured in the plasma and the sensitivity to corticosteroids was determined in isolated peripheral blood mononuclear cells. Quantitative data were compared between the two groups using the independent samples t test or Mann-Whitney U test. Categorical data were evaluated using Chi-squared test or Fisher's exact test. RESULTS Twenty-six patients were classified into Eos group and 31 patients were classified into Noneos group. Prior to treatment, the alpha diversity (Shannon index) (2.65 ± 0.63 vs. 2.56 ± 0.54, t = 0.328, P = 0.747) and the structure of the sputum microbiome were similar in the Eos group and the Noneos group. After 7 days of treatment, alpha diversity increased in both groups, while the microbiome richness (Ace index) was significantly lower in the Eos group (561.87 ± 109.13 vs. 767.88 ± 148.48, t = -3.535, P = 0.002). At the same time, IL-6 (12.09 ± 2.85 pg/mL vs. 15.54 ± 2.45 pg/mL, t = -4.913, P < 0.001) and IL-8 (63.64 ± 21.69 pg/mL vs. 78.97 ± 17.13 pg/mL, t = -2.981, P = 0.004) decreased more significantly in the Eos group, and the percentages of inhibition of IL-8 at dexamethasone concentrations 10 to 10 mol/L were significantly higher in the Eos group than those in the Noneos group (all P < 0.05). CONCLUSIONS The induced sputum microbiome richness decreased more significantly following treatment in the Eos patients compared to the Noneos patients. The lower plasma inflammatory factor levels and the higher percentage of inhibition of IL-8 might be due to higher corticosteroid sensitivity in Eos patients.
Collapse
|
39
|
Porsbjerg CM, Sverrild A, Lloyd CM, Menzies-Gow AN, Bel EH. Anti-alarmins in asthma: targeting the airway epithelium with next-generation biologics. Eur Respir J 2020; 56:2000260. [PMID: 32586879 PMCID: PMC7676874 DOI: 10.1183/13993003.00260-2020] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022]
Abstract
Monoclonal antibody therapies have significantly improved treatment outcomes for patients with severe asthma; however, a significant disease burden remains. Available biologic treatments, including anti-immunoglobulin (Ig)E, anti-interleukin (IL)-5, anti-IL-5Rα and anti-IL-4Rα, reduce exacerbation rates in study populations by approximately 50% only. Furthermore, there are currently no effective treatments for patients with severe, type 2-low asthma. Existing biologics target immunological pathways that are downstream in the type 2 inflammatory cascade, which may explain why exacerbations are only partly abrogated. For example, type 2 airway inflammation results from several inflammatory signals in addition to IL-5. Clinically, this can be observed in how fractional exhaled nitric oxide (F eNO), which is driven by IL-13, may remain unchanged during anti-IL-5 treatment despite reduction in eosinophils, and how eosinophils may remain unchanged during anti-IL-4Rα treatment despite reduction in F eNO The broad inflammatory response involving cytokines including IL-4, IL-5 and IL-13 that ultimately results in the classic features of exacerbations (eosinophilic inflammation, mucus production and bronchospasm) is initiated by release of "alarmins" thymic stromal lymphopoietin (TSLP), IL-33 and IL-25 from the airway epithelium in response to triggers. The central, upstream role of these epithelial cytokines has identified them as strong potential therapeutic targets to prevent exacerbations and improve lung function in patients with type 2-high and type 2-low asthma. This article describes the effects of alarmins and discusses the potential role of anti-alarmins in the context of existing biologics. Clinical phenotypes of patients who may benefit from these treatments are also discussed, including how biomarkers may help identify potential responders.
Collapse
Affiliation(s)
| | - Asger Sverrild
- Dept of Respiratory Medicine, Bispebjerg Hospital, Copenhagen, Denmark
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Elisabeth H Bel
- Dept of Respiratory Medicine, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Choi Y, Sim S, Park HS. Distinct functions of eosinophils in severe asthma with type 2 phenotype: clinical implications. Korean J Intern Med 2020; 35:823-833. [PMID: 32460456 PMCID: PMC7373972 DOI: 10.3904/kjim.2020.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/06/2020] [Indexed: 12/16/2022] Open
Abstract
Asthma is commonly recognized as a heterogeneous condition with a complex pathophysiology. With advances in the development of multiple medications for patients with asthma, most asthma symptoms are well managed. Nevertheless, 5% to 10% of adult asthmatic patients (called severe asthma) are in uncontrolled or partially controlled status despite intensive treatment. Especially, severe eosinophilic asthma is one of the severe asthma phenotypes characterized by eosinophilia in sputum/blood driven by type 2 immune responses. Eosinophils have been widely accepted as a central effector cell in the lungs. Some evidence has demonstrated that persistent eosinophilia in upper and lower airway mucosa contributes to asthma severity by producing various mediators including cytokines, chemokines and granule proteins. Moreover, extracellular traps released from eosinophils have been revealed to enhance type 2 inflammation in patients with severe asthma. These novel molecules have the ability to induce airway inf lammation and hyperresponsiveness through enhancing innate and type 2 immune responses. In this review, we highlight recent insight into the function of eosinophil extracellular traps in patients with severe asthma. In addition, the role of eosinophil extracellular vesicles in severe asthma is also proposed. Finally, current biologics are suggested as a potential strategy for effective management of severe eosinophilic asthma.
Collapse
Affiliation(s)
- Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Soyoon Sim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
- Correspondence to Hae-Sim Park, M.D. Department of Allergy and Clinical Immunology, Ajou University School of Medicine, 164 World cup-ro, Yeongtonggu, Suwon 16499, Korea Tel: +82-31-219-5196, Fax: +82-31-219-5154, E-mail:
| |
Collapse
|
41
|
Harvey ES, Langton D, Katelaris C, Stevens S, Farah CS, Gillman A, Harrington J, Hew M, Kritikos V, Radhakrishna N, Bardin P, Peters M, Reynolds PN, Upham JW, Baraket M, Bowler S, Bowden J, Chien J, Chung LP, Grainge C, Jenkins C, Katsoulotos GP, Lee J, McDonald VM, Reddel HK, Rimmer J, Wark PAB, Gibson PG. Mepolizumab effectiveness and identification of super-responders in severe asthma. Eur Respir J 2020; 55:13993003.02420-2019. [PMID: 32139455 DOI: 10.1183/13993003.02420-2019] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/07/2020] [Indexed: 01/22/2023]
Abstract
Severe asthma is a high-burden disease. Real-world data on mepolizumab in patients with severe eosinophilic asthma is needed to assess whether the data from randomised controlled trials are applicable in a broader population.The Australian Mepolizumab Registry (AMR) was established with an aim to assess the use, effectiveness and safety of mepolizumab for severe eosinophilic asthma in Australia.Patients (n=309) with severe eosinophilic asthma (median age 60 years, 58% female) commenced mepolizumab. They had poor symptom control (median Asthma Control Questionnaire (ACQ)-5 score of 3.4), frequent exacerbations (median three courses of oral corticosteroids (OCS) in the previous 12 months), and 47% required daily OCS. Median baseline peripheral blood eosinophil level was 590 cells·µL-1 Comorbidities were common: allergic rhinitis 63%, gastro-oesophageal reflux disease 52%, obesity 46%, nasal polyps 34%.Mepolizumab treatment reduced exacerbations requiring OCS compared with the previous year (annualised rate ratio 0.34 (95% CI 0.29-0.41); p<0.001) and hospitalisations (rate ratio 0.46 (95% CI 0.33-0.63); p<0.001). Treatment improved symptom control (median ACQ-5 reduced by 2.0 at 6 months), quality of life and lung function. Higher blood eosinophil levels (p=0.003) and later age of asthma onset (p=0.028) predicted a better ACQ-5 response to mepolizumab, whilst being male (p=0.031) or having body mass index ≥30 (p=0.043) predicted a lesser response. Super-responders (upper 25% of ACQ-5 responders, n=61, 24%) had a higher T2 disease burden and fewer comorbidities at baseline.Mepolizumab therapy effectively reduces the significant and long-standing disease burden faced by patients with severe eosinophilic asthma in a real-world setting.
Collapse
Affiliation(s)
- Erin S Harvey
- Centre of Excellence in Severe Asthma and Priority Research Centre for Healthy Lungs, Faculty of Health, University of Newcastle, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - David Langton
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia.,Dept of Thoracic Medicine, Frankston Hospital, Frankston, Australia
| | - Constance Katelaris
- School of Medicine, Western Sydney University, Campbelltown, Australia.,Immunology and Allergy Unit, Campbelltown Hospital, Campbelltown, Australia
| | - Sean Stevens
- Centre of Excellence in Severe Asthma and Priority Research Centre for Healthy Lungs, Faculty of Health, University of Newcastle, Newcastle, Australia
| | - Claude S Farah
- Dept of Thoracic Medicine, Concord Hospital, Concord, Australia
| | - Andrew Gillman
- Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia
| | - John Harrington
- Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Mark Hew
- Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, Australia
| | - Vicky Kritikos
- Dept of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, Australia
| | | | - Philip Bardin
- Lung and Sleep Medicine, Monash University and Medical Centre, Clayton, Australia
| | - Matthew Peters
- Dept of Thoracic Medicine, Concord Hospital, Concord, Australia
| | - Paul N Reynolds
- Lung Research, Hanson Institute and Dept of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
| | - John W Upham
- Dept of Respiratory Medicine, Princess Alexandra Hospital, Woolloongabba, Australia.,The University of Queensland Diamantina Institute, Woolloongabba, Australia
| | - Melissa Baraket
- South Western Sydney Clinical School, University of New South Wales, Sydney, Australia.,Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Simon Bowler
- Dept of Respiratory Medicine, Mater Hospital Brisbane, South Brisbane, Australia
| | - Jeffrey Bowden
- Respiratory and Sleep Services, Flinders Medical Centre and Flinders University, Bedford Park, Australia
| | - Jimmy Chien
- Dept of Sleep and Respiratory Medicine, Westmead Hospital, Westmead, Australia.,School of Medicine, The University of Sydney, Sydney, Australia
| | - Li Ping Chung
- Dept of Respiratory Medicine, Fiona Stanley Hospital, Murdoch, Australia
| | - Christopher Grainge
- Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Christine Jenkins
- Dept of Thoracic Medicine, Concord Hospital, Concord, Australia.,Concord Clinical School University of Sydney, Concord, Australia
| | - Gregory P Katsoulotos
- St George Specialist Centre, Kogarah, Australia.,St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia.,Woolcock Institute of Medical Research, Glebe, Australia
| | - Joy Lee
- Austin Health and Monash University, Melbourne, Australia
| | - Vanessa M McDonald
- Centre of Excellence in Severe Asthma and Priority Research Centre for Healthy Lungs, Faculty of Health, University of Newcastle, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Helen K Reddel
- Dept of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Janet Rimmer
- Woolcock Institute of Medical Research, Glebe, Australia.,St Vincent's Clinic, Darlinghurst, Australia
| | - Peter A B Wark
- Centre of Excellence in Severe Asthma and Priority Research Centre for Healthy Lungs, Faculty of Health, University of Newcastle, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Peter G Gibson
- Centre of Excellence in Severe Asthma and Priority Research Centre for Healthy Lungs, Faculty of Health, University of Newcastle, Newcastle, Australia .,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| |
Collapse
|
42
|
Phenotypes and endotypes of adult asthma: Moving toward precision medicine. J Allergy Clin Immunol 2020; 144:1-12. [PMID: 31277742 DOI: 10.1016/j.jaci.2019.05.031] [Citation(s) in RCA: 282] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
Asthma is a chronic inflammatory disease of the airways that is challenging to dissect into subgroups because of the heterogeneity present across the spectrum of the disease. Efforts to subclassify asthma using advanced computational methods have identified a number of different phenotypes that suggest that multiple pathobiologically driven clusters of disease exist. The main phenotypes that have been identified include (1) early-onset allergic asthma, (2) early-onset allergic moderate-to-severe remodeled asthma, (3) late-onset nonallergic eosinophilic asthma, and (4) late-onset nonallergic noneosinophilic asthma. Subgroups of these phenotypes also exist but have not been as consistently identified. Advances in our understanding of the diverse immunologic perturbations that drive airway inflammation are consistent with clinical traits associated with these phenotypes and their response to biologic therapies. This has improved the clinician's approach to characterizing asthmatic patients in the clinic. Being able to define asthma endotypes using clinical characteristics and biomarkers will move physicians toward even more personalized management of asthma and precision-based care in the future. Here we will review the most prominent phenotypes and immunologic advances that suggest these disease subtypes represent asthma endotypes.
Collapse
|
43
|
Prediction of response to biological treatment with monoclonal antibodies in severe asthma. Biochem Pharmacol 2020; 179:113978. [PMID: 32305434 DOI: 10.1016/j.bcp.2020.113978] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
In recent years, major developments have occurred in severe asthma management. Different asthma phenotypes and subgroups have been identified and new treatment options have become available. A total of five monoclonal antibodies are currently approved in severe asthma treatment: omalizumab, mepolizumab, reslizumab, benralizumab and dupilumab. These drugs have been shown to reduce exacerbations and to have an oral corticosteroid-sparing effect in many severe asthma patients. However, biological treatment is not successful in all patients and should be discontinued in non-responsive patients. Treating the right patient with the right biologic, and therefore biologic response prediction, has become a major point of interest in severe asthma management. A variety of response outcomes is utilized in the different clinical trials, as well as a huge range of potential predicting factors. Also, regarding the timing of the response evaluation, there are considerable differences between studies. This review summarizes the results from studies on predicting responses and responders to biological treatment in severe asthma, taking into account clinical, functional and inflammatory parameters assessed prior to the start of treatment as well as following a few months of therapy. In addition, future perspectives are discussed, highlighting the need for more research to improve patient identification and treatment responses in the field of biological treatment in severe asthma.
Collapse
|
44
|
Matsumoto H. Role of serum periostin in the management of asthma and its comorbidities. Respir Investig 2020; 58:144-154. [PMID: 32205146 DOI: 10.1016/j.resinv.2020.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/17/2020] [Accepted: 02/07/2020] [Indexed: 12/15/2022]
Abstract
Type-2 airway inflammation is a major characteristic of asthma. Assessing its degree of severity is, therefore, essential in asthma management. Periostin, a matricellular protein belonging to the fasciclin family, is a key molecule linking type-2 airway inflammation and airway remodeling. Fortunately, periostin can be detected in the blood and used to provide sustaining airway information on type-2 inflammation and remodeling. Serum periostin is elevated in the eosinophilic/type 2 subtype of severe asthma, and its levels remain relatively stable and reflect genetic backgrounds. This suggests that serum periostin may serve as a marker of geno-endophenotype with type-2 airway inflammation and thus could be a predictive marker of the long-term prognosis of asthma under treatment. As expected, serum periostin is particularly elevated in comorbidities associated with the eosinophilic/type 2 subtype of severe asthma, including eosinophilic chronic rhinosinusitis, aspirin-exacerbated respiratory diseases, allergic bronchopulmonary aspergillosis, and eosinophilic granulomatosis with polyangiitis. Conversely, serum periostin levels are relatively lower in the overweight/obese. Serum periostin measurements may help to significantly improve the management of patients with severe asthma.
Collapse
Affiliation(s)
- Hisako Matsumoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
45
|
Holgate ST, Walker S, West B, Boycott K. The Future of Asthma Care: Personalized Asthma Treatment. Clin Chest Med 2020; 40:227-241. [PMID: 30691714 DOI: 10.1016/j.ccm.2018.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although once considered a single disease entity, asthma is now known to be a complex inflammatory disease engaging a range of causal pathways. The most frequent forms of asthma are identified by sputum/blood eosinophilia and activation of type 2 inflammatory pathways involving interleukins-3, -4, -5, and granulocyte-macrophage colony-stimulating factor. The use of diagnostics that identify T2 engagement linked to the selective use of highly targeted biologics has opened up a new way of managing severe disease. Novel technologies, such as wearables and intelligent inhalers, enable real-time remote monitoring of asthma, creating a unique opportunity for personalized health care.
Collapse
Affiliation(s)
- Stephen T Holgate
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, The Sir Henry Wellcome Research Laboratories, Southampton General Hospital, Mail Point 810, Level, Southampton SO166YD, UK.
| | | | | | - Kay Boycott
- Asthma UK, 18 Mansell Street, London E1 8AA, UK
| |
Collapse
|
46
|
Nelson RK, Bush A, Stokes J, Nair P, Akuthota P. Eosinophilic Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2019; 8:465-473. [PMID: 31786254 DOI: 10.1016/j.jaip.2019.11.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/19/2019] [Accepted: 11/24/2019] [Indexed: 02/06/2023]
Abstract
Asthma endotypes are constantly evolving. Currently, there are no universally accepted criteria to define endotypes. The TH2-high endotype can have either allergic or nonallergic underpinnings and is typically characterized by some degree of eosinophilic airway inflammation. Unbiased clustering analyses have led to the identification of pediatric and adult phenotypes characterized by TH2 inflammation and associated endotypes with eosinophilic inflammation. Aspirin-exacerbated respiratory disease has also long been recognized as a unique asthma phenotype. An approach to identify these groups with biomarkers and subsequently choose a targeted therapeutic modality, particularly in severe disease requiring biologic agents, is outlined.
Collapse
Affiliation(s)
- Ryan K Nelson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, Calif
| | - Andrew Bush
- Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, and National Heart and Lung Institute, Imperial School of Medicine, London, United Kingdom
| | - Jeffrey Stokes
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine in St Louis, St Louis, Mo
| | - Parameswaran Nair
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada; Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Praveen Akuthota
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, Calif.
| |
Collapse
|
47
|
Chung KF, Adcock IM. Precision medicine for the discovery of treatable mechanisms in severe asthma. Allergy 2019; 74:1649-1659. [PMID: 30865306 DOI: 10.1111/all.13771] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022]
Abstract
Although the complex disease of asthma has been defined as being heterogeneous, the extent of its endophenotypes remains unclear. The pharmacological approach to initiating treatment has, until recently, been based on disease control and severity. The introduction of antibody therapies targeting the Type 2 inflammation pathway for patients with severe asthma has resulted in the recognition of an allergic and an eosinophilic phenotype, which are not mutually exclusive. Concomitantly, molecular phenotyping based on a transcriptomic analysis of bronchial epithelial and sputum cells has identified a Type 2 high inflammation cluster characterized by eosinophilia and recurrent exacerbations, as well as Type 2 low clusters linked with IL-6 trans-signalling, interferon pathways, inflammasome activation and mitochondrial oxidative phosphorylation pathways. Systems biology approaches are establishing the links between these pathways or mechanisms, and clinical and physiologic features. Validation of these pathways contributes to defining endotypes and treatable mechanisms. Precision medicine approaches are necessary to link treatable mechanisms with treatable traits and biomarkers derived from clinical, physiologic and inflammatory features of clinical phenotypes. The deep molecular phenotyping of airway samples along with noninvasive biomarkers linked to bioinformatic and machine learning techniques will enable the rapid detection of molecular mechanisms that transgresses beyond the concept of treatable traits.
Collapse
Affiliation(s)
- Kian Fan Chung
- National Heart & Lung Institute Imperial College London and Royal Brompton & Harefield NHS Trust London UK
| | - Ian M. Adcock
- National Heart & Lung Institute Imperial College London and Royal Brompton & Harefield NHS Trust London UK
| |
Collapse
|
48
|
Murray DD, Itenov TS, Sivapalan P, Eklöf JV, Holm FS, Schuetz P, Jensen JU. Biomarkers of Acute Lung Injury The Individualized Approach: for Phenotyping, Risk Stratification and Treatment Surveillance. J Clin Med 2019; 8:jcm8081163. [PMID: 31382587 PMCID: PMC6722821 DOI: 10.3390/jcm8081163] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
Do we need biomarkers of lung damage and infection: For what purpose and how should they be used properly? Biomarkers of lung damage can be used for diagnosis, risk stratification/prediction, treatment surveillance and adjustment of targeted therapy. Additionally, novel "omics" methods may offer a completely different and effective way of improving the understanding of pathogenesis of lung damage and a way to develop new candidate lung damage biomarkers. In the current review, we give an overview within the field of acute lung damage of (i) disease mechanism biomarkers, (ii) of "ready to use" evidence-based biomarker-guided lung infection management, (iii) of novel strategies of inflammatory phenotyping and how this can be used to tailor corticosteroid treatment, (iv) a future perspective of where "omics" technologies and mindsets may become increasingly important in developing new strategies for treatment and for understanding the development of acute lung damage.
Collapse
Affiliation(s)
- Daniel D Murray
- PERSIMUNE, Department of Infectious Diseases, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | | | - Pradeesh Sivapalan
- Respiratory Medicine Section, Department of Internal Medicine, Herlev-Gentofte Hospital, DK-2900 Hellerup, Denmark
| | - Josefin Viktoria Eklöf
- Respiratory Medicine Section, Department of Internal Medicine, Herlev-Gentofte Hospital, DK-2900 Hellerup, Denmark
| | - Freja Stæhr Holm
- Respiratory Medicine Section, Department of Internal Medicine, Herlev-Gentofte Hospital, DK-2900 Hellerup, Denmark
| | - Philipp Schuetz
- Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
- Faculty of Medicine, University of Basel, 4001 Basel, Switzerland
| | - Jens Ulrik Jensen
- PERSIMUNE, Department of Infectious Diseases, Rigshospitalet, DK-2100 Copenhagen, Denmark.
- Respiratory Medicine Section, Department of Internal Medicine, Herlev-Gentofte Hospital, DK-2900 Hellerup, Denmark.
| |
Collapse
|
49
|
Abstract
Objective: Asthma is a common heterogeneous disease characterized by airway inflammation and bronchoconstriction. Current treatment guidelines provide recommendations for categorizing disease severity, asthma control and management. This paper reviews asthma assessment in primary care and describes the pathophysiology, clinical characteristics and new targeted treatments available for patients with severe eosinophilic asthma. Methods: A non-systematic PubMed literature search was conducted and articles, primarily from the last 5 years, were selected based on relevance to primary care practice, asthma pathophysiology and biologic therapies. Results: Despite optimal therapy including high-dose inhaled corticosteroids (ICS), long-acting β2-agonists and tiotropium, ∼4-10% of all patients with severe asthma continue to have poor asthma control. These patients have impaired quality of life, frequent exacerbations and are exposed to the side effects of repeated courses of oral steroids. Approximately 50% of patients with severe uncontrolled asthma have eosinophilic asthma, with increased airway expression of type 2 cytokines IL-4, IL-5 and IL-13. Eosinophilic asthma is identified in primary care by having eosinophils ≥150-300 cells/μL on a complete blood count with differential. Conclusions: A new class of agents is available for patients with moderate to severe eosinophilic asthma. Four biologic therapies - mepolizumab, reslizumab, benralizumab and dupilumab - that interfere with the regulation and activity of eosinophils have been approved by the FDA for patients with moderate to severe asthma with an eosinophilic phenotype. Primary care physicians should be familiar with these medications to explain part of the rationale for referral to specialist care and manage patient expectations for treatment.
Collapse
|
50
|
Eger KA, Bel EH. The emergence of new biologics for severe asthma. Curr Opin Pharmacol 2019; 46:108-115. [PMID: 31229937 DOI: 10.1016/j.coph.2019.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/29/2019] [Accepted: 05/15/2019] [Indexed: 02/08/2023]
Abstract
Patients with severe asthma experience severe symptoms and frequent exacerbations despite intensive treatment with inhaled and oral glucocorticoids. Biologics for severe asthma aim to reduce asthma-related and glucocorticoid-induced morbidity. Recently, new biologics targeting interleukin (IL)-5, IL-5 receptor and IL-4/IL-13, which are all cytokines involved in so-called type 2 airway inflammation, were approved for severe asthma. They show a reduction in exacerbation rate and an oral glucocorticoid-sparing effect. Studies with upstream biologics targeting alarmin cytokines such as thymic stromal lymphopoietin (TSLP) and IL-33 are underway, and newly designed bispecific antibodies targeting more than one pathway are in early phases of development. Such pathway-targeted add-on treatments will soon become standard of care for all patients with severe asthma.
Collapse
Affiliation(s)
- Katrien Ab Eger
- Amsterdam UMC, Academic Medical Centre, University of Amsterdam, Dept. of Respiratory Medicine, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Elisabeth H Bel
- Amsterdam UMC, Academic Medical Center, University of Amsterdam, Dept. of Respiratory Medicine, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|