1
|
Boboltz A, Kumar S, Duncan GA. Inhaled drug delivery for the targeted treatment of asthma. Adv Drug Deliv Rev 2023; 198:114858. [PMID: 37178928 PMCID: PMC10330872 DOI: 10.1016/j.addr.2023.114858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/14/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
Asthma is a chronic lung disease affecting millions worldwide. While classically acknowledged to result from allergen-driven type 2 inflammatory responses leading to IgE and cytokine production and the influx of immune cells such as mast cells and eosinophils, the wide range in asthmatic pathobiological subtypes lead to highly variable responses to anti-inflammatory therapies. Thus, there is a need to develop patient-specific therapies capable of addressing the full spectrum of asthmatic lung disease. Moreover, delivery of targeted treatments for asthma directly to the lung may help to maximize therapeutic benefit, but challenges remain in design of effective formulations for the inhaled route. In this review, we discuss the current understanding of asthmatic disease progression as well as genetic and epigenetic disease modifiers associated with asthma severity and exacerbation of disease. We also overview the limitations of clinically available treatments for asthma and discuss pre-clinical models of asthma used to evaluate new therapies. Based on the shortcomings of existing treatments, we highlight recent advances and new approaches to treat asthma via inhalation for monoclonal antibody delivery, mucolytic therapy to target airway mucus hypersecretion and gene therapies to address underlying drivers of disease. Finally, we conclude with discussion on the prospects for an inhaled vaccine to prevent asthma.
Collapse
Affiliation(s)
- Allison Boboltz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States
| | - Sahana Kumar
- Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, United States
| | - Gregg A Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States; Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
2
|
Ignatova GL, Antonov VN, Makarova EA, Kochetkova SA. [Experience of using dupilumab in the treatment of severe asthma]. TERAPEVT ARKH 2020; 92:95-99. [PMID: 33346468 DOI: 10.26442/00403660.2020.08.000718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 11/22/2022]
Abstract
The article provides data on modern approaches to the treatment of patients with severe uncontrolled bronchial asthma with an emphasis on the use of dupilumab, a human recombinant monoclonal antibody to the alpha subunit of the interleukin (IL)-4 receptor, which inhibits signal transmission from both IL-4 and IL-13. The results of dupilumab pivotal randomized clinical trials DRI12544, QUEST and VENTURE are summarized. Indications for use of this medicinal product are listed in Federal Clinical Guidelines on the management of asthma (year of revision 2019). Clinical cases with various clinical course of bronchial asthma are described, including cases with frequent exacerbations. In conclusion, dupilumab could be a treatment of choice for the patients with severe bronchial asthma and it is reasonable from an economic, clinical and pathogenetic point of view.
Collapse
|
3
|
YALCIN ARZUDIDEM, ONBASI KEVSER, UZUN RUSEN, HERTH FELIX, SCHNABEL PHILIPPALBERT. Human(ized) monoclonal antibodies in atopic patients - state of the art. Cent Eur J Immunol 2020; 45:195-201. [PMID: 33456331 PMCID: PMC7792442 DOI: 10.5114/ceji.2020.97909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/13/2017] [Indexed: 02/05/2023] Open
Abstract
Asthma is an important chronic disease affecting a lot of people worldwide. Treatment options for asthma like biological agents are being developed more frequently nowadays. Despite a lot of treatment options, some patients still remain symptomatic. As more and more practitioners choose treatment with biologic agents as a convenient way of therapy, biologic agents and other valuable methods must be discovered in order to cope with a growing number of treatment agents. This manuscript emphasizes on new generation monoclonal human(ized) antibodies in asthmatics and off-label use . The first developed biologic agent is the anti-immunoglobulin E monoclonal antibody called omalizumab. Currently it is an approved treatment option for asthma.
Collapse
Affiliation(s)
- ARZU DIDEM YALCIN
- Department of Internal Medicine, Allergy and Clinical Immunology, Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Department of Allergy and Clinical Immunology, Antalya Training and Research Hospital, University of Medical Science, Antalya, Turkey
| | - KEVSER ONBASI
- Department of Internal Medicine, Allergy and Clinical Immunology, Dumlupinar University, Kütahya, Turkey
| | - RUSEN UZUN
- Department of Pulmonology, Antalya Training and Research Hospital, University of Medical Science, Antalya, Turkey
| | - FELIX HERTH
- Department of Pulmonary and Critical Care Medicine, Thoraxklinik Translational Lung Research Center, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
4
|
Tibble H, Flook M, Sheikh A, Tsanas A, Horne R, Vrijens B, De Geest S, Stagg HR. Measuring and reporting treatment adherence: What can we learn by comparing two respiratory conditions? Br J Clin Pharmacol 2020; 87:825-836. [PMID: 32639589 DOI: 10.1111/bcp.14458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/02/2020] [Accepted: 06/24/2020] [Indexed: 01/03/2023] Open
Abstract
Medication non-adherence, defined as any deviation from the regimen recommended by their healthcare provider, can increase morbidity, mortality and side effects, while reducing effectiveness. Through studying two respiratory conditions, asthma and tuberculosis (TB), we thoroughly review the current understanding of the measurement and reporting of medication adherence. In this paper, we identify major methodological issues in the standard ways that adherence has been conceptualised, defined and studied in asthma and TB. Between and within the two diseases there are substantial variations in adherence reporting, linked to differences in dosing intervals and treatment duration. Critically, the communicable nature of TB has resulted in dose-by-dose monitoring becoming a recommended treatment standard. Through the lens of these similarities and contrasts, we highlight contemporary shortcomings in the generalised conceptualisation of medication adherence. Furthermore, we outline elements in which knowledge could be directly transferred from one condition to the other, such as the application of large-scale cost-effective monitoring methods in TB to resource-poor settings in asthma. To develop a more robust evidence-based approach, we recommend the use of standard taxonomies detailed in the ABC taxonomy when measuring and discussing adherence. Regimen and intervention development and use should be based on sufficient evidence of the commonality and type of adherence behaviours displayed by patients with the relevant condition. A systematic approach to the measurement and reporting of adherence could improve the value and generalisability of research across all health conditions.
Collapse
Affiliation(s)
- Holly Tibble
- Usher Institute, Edinburgh Medical School, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.,Asthma UK Centre for Applied Research, Usher Institute, Centre for Medical Informatics, University of Edinburgh, Edinburgh, UK
| | - Mary Flook
- Usher Institute, Edinburgh Medical School, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Aziz Sheikh
- Usher Institute, Edinburgh Medical School, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.,Asthma UK Centre for Applied Research, Usher Institute, Centre for Medical Informatics, University of Edinburgh, Edinburgh, UK.,Health Data Research UK, London, UK
| | - Athanasios Tsanas
- Usher Institute, Edinburgh Medical School, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.,Asthma UK Centre for Applied Research, Usher Institute, Centre for Medical Informatics, University of Edinburgh, Edinburgh, UK
| | - Rob Horne
- Asthma UK Centre for Applied Research, Usher Institute, Centre for Medical Informatics, University of Edinburgh, Edinburgh, UK.,Centre for Behavioural Medicine, Department for Practice and Policy, UCL School of Pharmacy, University College London, London, UK
| | - Bernard Vrijens
- AARDEX Group, Seraing, Belgium.,Liège University, Liège, Belgium
| | - Sabina De Geest
- Institute of Nursing Science, University of Basel, Basel, Switzerland.,Academic Centre for Nursing and Midwifery, KU Leuven, Leuven, Belgium
| | - Helen R Stagg
- Usher Institute, Edinburgh Medical School, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Toki S, Goleniewska K, Zhang J, Zhou W, Newcomb DC, Zhou B, Kita H, Boyd KL, Peebles RS. TSLP and IL-33 reciprocally promote each other's lung protein expression and ILC2 receptor expression to enhance innate type-2 airway inflammation. Allergy 2020; 75:1606-1617. [PMID: 31975538 PMCID: PMC7354889 DOI: 10.1111/all.14196] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 11/16/2019] [Accepted: 11/28/2019] [Indexed: 01/10/2023]
Abstract
Background The epithelial cell‐derived danger signal mediators thymic stromal lymphopoietin (TSLP) and IL‐33 are consistently associated with adaptive Th2 immune responses in asthma. In addition, TSLP and IL‐33 synergistically promoted group 2 innate lymphoid cell (ILC2) activation to induce innate allergic inflammation. However, the mechanism of this synergistic ILC2 activation is unknown. Methods BALB/c WT and TSLP receptor‐deficient (TSLPR−/−) mice were challenged intranasally with Alternaria extract (Alt‐Ext) or PBS for 4 consecutive days to evaluate innate airway allergic inflammation. WT mice pre‐administered with rTSLP or vehicle, TSLPR−/− mice, and IL‐33 receptor‐deficient (ST2−/−) mice were challenged intranasally with Alt‐Ext or vehicle once or twice to evaluate IL‐33 release and TSLP expression in the lung. TSLPR and ST2 expression on lung ILC2 were measured by flow cytometry after treatment of rTSLP, rIL‐33, rTSLP + rIL‐33, or vehicle. Results Thymic stromal lymphopoietin receptor deficient mice had significantly decreased the number of lung ILC2 expressing IL‐5 and IL‐13 following Alt‐Ext‐challenge compared to WT mice. Further, eosinophilia, protein level of lung IL‐4, IL‐5, and IL‐13, and airway mucus score were also significantly decreased in TSLPR−/− mice compared to WT mice. Endogenous and exogenous TSLP increased Alt‐Ext‐induced IL‐33 release into BALF, and ST2 deficiency decreased Alt‐Ext‐induced TSLP expression in the lung. Further, rTSLP and rIL‐33 treatment reciprocally increased each other's receptor expression on lung ILC2 in vivo and in vitro. Conclusion Thymic stromal lymphopoietin and IL‐33 signaling reciprocally enhanced each other's protein release and expression in the lung following Alt‐Ext‐challenge and each other's receptor expression on lung ILC2 to enhance ILC2 activation.
Collapse
Affiliation(s)
- Shinji Toki
- Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University School of Medicine Nashville TN USA
| | - Kasia Goleniewska
- Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University School of Medicine Nashville TN USA
| | - Jian Zhang
- Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University School of Medicine Nashville TN USA
| | - Weisong Zhou
- Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University School of Medicine Nashville TN USA
| | - Dawn C. Newcomb
- Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University School of Medicine Nashville TN USA
- Department of Pathology, Microbiology, and Immunology Vanderbilt University School of Medicine Nashville TN USA
| | - Baohua Zhou
- Wells Center for Pediatric Research Department of Pediatrics Indiana University School of Medicine Indianapolis IN USA
| | - Hirohito Kita
- Division of Allergic Diseases Department of Internal Medicine Mayo Clinic Rochester MN USA
| | - Kelli L. Boyd
- Department of Pathology, Microbiology, and Immunology Vanderbilt University School of Medicine Nashville TN USA
| | - Ray S. Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University School of Medicine Nashville TN USA
- Department of Pathology, Microbiology, and Immunology Vanderbilt University School of Medicine Nashville TN USA
| |
Collapse
|
6
|
Magat JM, Thomas JL, Dumouchel JP, Murray F, Li WX, Li J. Endogenous IL-33 and Its Autoamplification of IL-33/ST2 Pathway Play an Important Role in Asthma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1592-1597. [PMID: 31988179 PMCID: PMC7065953 DOI: 10.4049/jimmunol.1900690] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/31/2019] [Indexed: 12/29/2022]
Abstract
IL-33 and its receptor ST2 are contributing factors to airway inflammation and asthma exacerbation. The IL-33/ST2 signaling pathway is involved in both the onset and the acute exacerbations of asthma. In this study, we address the role of endogenous IL-33 and its autoamplification of the IL-33/ST2 pathway in Ag-dependent and Ag-independent asthma-like models. Wild-type, IL-33 knockout, ST2 knockout mice were either intratracheally administrated with 500 ng of rIL-33 per day for four consecutive days or were sensitized and challenged with OVA over 21 d. In wild-type mice, IL-33 or OVA induced similar airway hyperresponsiveness and eosinophilic airway inflammation. IL-33 induced its own mRNA and ST2L mRNA expression in the lung. IL-33 autoamplified itself and ST2 protein expression in airway epithelial cells. OVA also induced IL-33 and ST2 protein expression. In IL-33 knockout mice, the IL-33- and OVA-induced airway hyperresponsiveness and eosinophilic airway inflammation were both significantly attenuated, whereas IL-33-induced ST2L mRNA expression was preserved, although no autoamplification of IL-33/ST2 pathway was observed. In ST2 knockout mice, IL-33 and OVA induced airway hyperresponsiveness and eosinophilic airway inflammation were both completely diminished, and no IL-33/ST2 autoamplification was observed. These results suggest that endogenous IL-33 and its autoamplification of IL-33/ST2 pathway play an important role in the induction of asthma-like phenotype. Thus an intact IL-33/ST2 pathway is necessary for both Ag-dependent and Ag-independent asthma-like mouse models.
Collapse
Affiliation(s)
- Jenna M Magat
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Joanna L Thomas
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92093
| | - Justin P Dumouchel
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Fiona Murray
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Willis X Li
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Jinghong Li
- Department of Medicine, University of California San Diego, La Jolla, CA 92093; and
| |
Collapse
|
7
|
Ando K, Tanaka A, Sagara H. Comparative Efficacy and Safety of Dupilumab and Benralizumab in Patients with Inadequately Controlled Asthma: A Systematic Review. Int J Mol Sci 2020; 21:ijms21030889. [PMID: 32019141 PMCID: PMC7037967 DOI: 10.3390/ijms21030889] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/30/2022] Open
Abstract
No head-to-head trials have compared the efficacy and safety between the licensed dosage and administration dosage of dupilumab and benralizumab for inadequately controlled asthma. We conducted an indirect treatment comparison to estimate differences in the efficacy and safety between dupilumab and benralizumab for inadequately controlled asthma using the Bayesian approach. The primary efficacy endpoint was annual exacerbation rate (AER). A subgroup analysis by blood eosinophil count was also performed. The primary safety endpoint was the incidence of any adverse events (AAEs). The results demonstrate that there was no significant difference in the AER between dupilumab and benralizumab in overall patients and the subgroup with the blood eosinophil count of <150. However, the AER was significantly lower in the dupilumab group than in the benralizumab group in the subgroup with a blood eosinophil count of ≥150 but <300, and ≥300 with the rate ratio and 95% credible interval of 0.51 (0.29–0.92) and 0.58 (0.39–0.84), respectively. There was no significant difference in the AAEs between the dupilumab and benralizumab groups. This indirect treatment comparison indicates that dupilumab is superior to benralizumab in patients with inadequately controlled asthma having higher blood eosinophil counts. A direct comparison is required to provide definitive evidence. Systematic Review Registration: UMIN-CTR no. UMIN000036256.
Collapse
|
8
|
Affinity improvement of the fully human anti‑TSLP recombinant antibody. Mol Med Rep 2019; 21:759-767. [PMID: 31974622 PMCID: PMC6947841 DOI: 10.3892/mmr.2019.10880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/25/2019] [Indexed: 12/30/2022] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is a potentially important target for the treatment of asthma and malignancies. However, a fully human antibody reactive with TSLP is currently unavailable for clinical use. In a previous study, a human anti-TSLP-single-chain antibody variable fragment (anti-TSLP-scFv) 84 was selected by phage display from a constructed human scFv library. In the present study, a computer simulation method was developed using Discovery Studio 4.5 software, to increase the affinity of anti-TSLP-scFv-84. Specific primers were designed and mutated DNA sequences of anti-TSLP-scFvs were obtained by overlap extension PCR. The mutant scFvs were expressed in pLZ16 and affinity-enhanced anti-TSLP-scFv-M4 was screened using ELISA. However, in general the scFvs have low stability and short half-lives in vivo. Therefore, scFv-84 and scFv-M4 were inserted into eukaryotic expression vectors (pcDNA3.1-sp-Fc and PMH3EN-sp-Fc) and then transfected into 293F cells to express anti-TSLP-scFv-Fc. ELISA and western blotting results indicated the size of the anti-TSLP-scFv-Fc to be ~50 kDa. Binding of anti-TSLP-scFv-Fc-M4 to TSLP was enhanced compared with the pre-mutated scFv-Fc-84. The affinity of the mutated recombinant antibody was determined using the BIAcore technique and found to be ~10-fold greater than the pre-mutated antibody.
Collapse
|
9
|
Hawiger J, Zienkiewicz J. Decoding inflammation, its causes, genomic responses, and emerging countermeasures. Scand J Immunol 2019; 90:e12812. [PMID: 31378956 PMCID: PMC6883124 DOI: 10.1111/sji.12812] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/03/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022]
Abstract
Inflammation is the mechanism of diseases caused by microbial, autoimmune, allergic, metabolic and physical insults that produce distinct types of inflammatory responses. This aetiologic view of inflammation informs its classification based on a cause‐dependent mechanism as well as a cause‐directed therapy and prevention. The genomic era ushered in a new understanding of inflammation by highlighting the cell's nucleus as the centre of the inflammatory response. Exogenous or endogenous inflammatory insults evoke genomic responses in immune and non‐immune cells. These genomic responses depend on transcription factors, which switch on and off a myriad of inflammatory genes through their regulatory networks. We discuss the transcriptional paradigm of inflammation based on denying transcription factors’ access to the nucleus. We present two approaches that control proinflammatory signalling to the nucleus. The first approach constitutes a novel intracellular protein therapy with bioengineered physiologic suppressors of cytokine signalling. The second approach entails control of proinflammatory transcriptional cascades by targeting nuclear transport with a cell‐penetrating peptide that inhibits the expression of 23 out of the 26 mediators of inflammation along with the nine genes required for metabolic responses. We compare these emerging anti‐inflammatory countermeasures to current therapies. The transcriptional paradigm of inflammation offers nucleocentric strategies for microbial, autoimmune, metabolic, physical and other types of inflammation afflicting millions of people worldwide.
Collapse
Affiliation(s)
- Jacek Hawiger
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jozef Zienkiewicz
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA
| |
Collapse
|
10
|
Wang L, Feng M, Li Q, Qiu C, Chen R. Advances in nanotechnology and asthma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:180. [PMID: 31168461 DOI: 10.21037/atm.2019.04.62] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
According to the World Health Organization, Asthma is the fastest-growing disease in the world alongside HIV/AIDS, and its socioeconomic burden exceeds the sum of HIV/AIDS and tuberculosis. Its high disability and mortality rates have become serious social and public health concerns. Asthma is a heterogeneous disease in which genetic polymorphisms interact with the environmental factors. While no specific treatment has been available for asthma due to its complex pathogenesis, the advances in nanotechnology have brought new hope for the early diagnosis, treatment, and prevention of asthma. Nanotechnology can achieve targeted delivery of drugs or genes, reduce toxic effects, and improve drug bioavailability. The nano-modifications of drugs and the development of new nano-drugs have become new research directions. Studies have demonstrated the safety and effectiveness of nanocarriers. However, many challenges still need to be overcome before nanotherapy can be applied in clinical practice. In this article we review the new research highlights in this area, with an attempt to explore the great potential and feasibility of nanotechnology in treating asthma.
Collapse
Affiliation(s)
- Lingwei Wang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen Institute of Respiratory Diseases, Shenzhen 518020, China
| | - Mengjie Feng
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen Institute of Respiratory Diseases, Shenzhen 518020, China
| | - Qiuwen Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen Institute of Respiratory Diseases, Shenzhen 518020, China
| | - Chen Qiu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen Institute of Respiratory Diseases, Shenzhen 518020, China
| | - Rongchang Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen Institute of Respiratory Diseases, Shenzhen 518020, China
| |
Collapse
|
11
|
Busse PJ, Farkas H, Banerji A, Lumry WR, Longhurst HJ, Sexton DJ, Riedl MA. Lanadelumab for the Prophylactic Treatment of Hereditary Angioedema with C1 Inhibitor Deficiency: A Review of Preclinical and Phase I Studies. BioDrugs 2019; 33:33-43. [PMID: 30539362 PMCID: PMC6373397 DOI: 10.1007/s40259-018-0325-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hereditary angioedema (HAE) with C1 esterase inhibitor (C1-INH) deficiency (C1-INH-HAE) is a rare disease characterized by diminished levels or dysfunctional activity of C1-INH, leading to dysregulated plasma kallikrein activity within the kallikrein-kinin pathway. Symptoms manifest as painful, potentially life-threatening swelling of subcutaneous tissues throughout the body and/or submucosal edema in the upper airway or gastrointestinal tract. Attacks recur with unpredictable frequency, intensity, and duration, placing a heavy burden on patients' daily lives. Despite improved availability of medications for on-demand treatment during attacks and prophylaxis of future attacks, unmet needs remain. Lanadelumab, a fully human monoclonal antibody, may help address some of the limitations of existing prophylactic options (e.g., the need for intravenous administration or frequent dosing). Preclinical studies demonstrate that it is highly potent and specifically inhibits plasma kallikrein, and findings from phase Ia and Ib studies suggest this agent is well tolerated and provides sustained inhibition of plasma kallikrein, allowing for less frequent dosing. The phase III HELP Study (NCT02586805) evaluating the efficacy and safety of lanadelumab in preventing HAE attacks has been completed, and its open-label extension (NCT02741596) is ongoing. Lanadelumab is now approved in the USA and Canada for prophylaxis to prevent attacks of HAE in patients aged ≥ 12 years. This review provides an overview of the discovery and clinical development of lanadelumab, from preclinical through phase Ib studies, characterizing its safety/tolerability, efficacy, and pharmacokinetic and pharmacodynamic profiles. It also highlights how this agent may positively impact clinical care of patients with C1-INH-HAE.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Angioedemas, Hereditary/drug therapy
- Angioedemas, Hereditary/pathology
- Angioedemas, Hereditary/physiopathology
- Angioedemas, Hereditary/prevention & control
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Clinical Trials, Phase I as Topic
- Drug Evaluation, Preclinical
- Humans
- Middle Aged
- Plasma Kallikrein/antagonists & inhibitors
- Plasma Kallikrein/drug effects
- Young Adult
Collapse
Affiliation(s)
- Paula J Busse
- Division of Clinical Immunology and Allergy, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Room 11-20, New York, NY, 10029, USA.
| | - Henriette Farkas
- Hungarian Angioedema Reference Center, 3rd Department of Internal Medicine, Semmelweis University, Kutvolgyi ut 4, Budapest, 1125, Hungary
| | - Aleena Banerji
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Cox 201, Boston, MA, 02114, USA
| | - William R Lumry
- Allergy and Asthma Research Associates, 10100 N. Central Expressway, Suite 100, Dallas, TX, 75231, USA
| | - Hilary J Longhurst
- Department of Immunology, Addenbrookes Hospital Cambridge University NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | | | - Marc A Riedl
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, 8899 University Center Lane, Suite 230, San Diego, CA, 92122, USA
| |
Collapse
|
12
|
Translating Asthma: Dissecting the Role of Metabolomics, Genomics and Personalized Medicine. Indian J Pediatr 2018; 85:643-650. [PMID: 29185231 DOI: 10.1007/s12098-017-2520-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/27/2017] [Indexed: 01/26/2023]
Abstract
The management of asthma has largely stagnated over the last 25 years, but we are at the dawning of a new age wherein -omics technology can help us manage the disease objectively and rationally. Even in this new scientific age, getting the basics of asthma management right remains essential. The new technologies which can be applied to multiple biological samples include genomics (study of the genome), transcriptomics (gene transcription), lipidomics, proteomics and metabolomics (lipids, proteins and metabolites, respectively) and breathomics, using exhaled breath as a source of biomarkers, which is of particular interest in view of its non-invasive nature in pediatrics. Important applications will include the diagnosis of airways disease, including its components; the pathways driving airway pathology; monitoring the response to treatment; and measuring future risk (asthma attacks, poor lung growth trajectory). With the advent of a wide range of novel biologicals to treat asthma, -omics technology to personalize therapy will be especially important. The U-BIOPRED (Europe) and SARP (USA) groups have been most active in this field, especially using bronchoscopically obtained samples to perform cluster analyses to define new asthma endotypes. However, stability over time and consistency between investigators is imperfect. This is perhaps unsurprising; results of biomarker studies in asthma will be a composite of the underlying disease, the (variable) effects of adverse drivers such as allergen exposure and pollution, the effects of treatment, and the effects of adherence or otherwise to treatment. Ultimately, the aim should be an exhaled breath based tool with a rapid result that can be used as a routine in the clinic. However, at the moment, there are as yet no clinical applications in children of -omics technology.
Collapse
|
13
|
The effect of omalizumab treatment on severe allergic asthma and allergic comorbidities: real-life experience from the Czech Anti-IgE Registry. Postepy Dermatol Alergol 2018; 35:510-515. [PMID: 30429711 PMCID: PMC6232552 DOI: 10.5114/ada.2018.77243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/11/2018] [Indexed: 01/23/2023] Open
Abstract
Introduction Omalizumab is indicated for the treatment of severe allergic asthma (SAA) and chronic spontaneous urticaria, although a number of studies have confirmed the effectiveness of this therapy also for other IgE-mediated diseases. Aim To assess the impact of anti-IgE therapy on SAA and comorbid IgE-mediated allergic diseases in patients treated with omalizumab for SAA enrolled in the CAR (Czech Anti-IgE Registry). Material and methods Three hundred and ten patients with SAA treated with omalizumab were enrolled in the CAR. Two hundred and twenty-nine individuals were evaluated after 12 months of omalizumab treatment for asthma control test (ACT), examination of fractional exhaled nitric oxide (FENO), forced expiratory volume in 1 s (FEV1), the use of systemic corticosteroids, side effects of treatment and clinical effect of omalizumab on allergic comorbidities (allergic rhinitis, chronic urticaria, atopic dermatitis and food allergy). Results After 12 months of treatment with omalizumab, patients experienced a significant improvement of ACT and FEV1, reduction of FENO, use of systemic corticosteroids for asthma exacerbations and dose of maintenance oral corticosteroid therapy. The positive effect of treatment with omalizumab was observed in 82.2% of patients with allergic rhinitis, in 85.7% of patients with chronic urticaria, in 82.1% of patients with atopic dermatitis, and in 67.3% of patients with food allergy. Conclusions In the CAR registry, patients with SAA treated with omalizumab showed a significant positive effect of anti-IgE therapy not only on the asthma control, but also on allergic comorbidities.
Collapse
|
14
|
Tian BP, Li F, Li R, Hu X, Lai TW, Lu J, Zhao Y, Du Y, Liang Z, Zhu C, Shao W, Li W, Chen ZH, Sun X, Chen X, Ying S, Ling D, Shen H. Nanoformulated ABT-199 to effectively target Bcl-2 at mitochondrial membrane alleviates airway inflammation by inducing apoptosis. Biomaterials 2018; 192:429-439. [PMID: 30500724 DOI: 10.1016/j.biomaterials.2018.06.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023]
Abstract
Elimination of airway inflammatory cells is essential for asthma control. As Bcl-2 protein is highly expressed on the mitochondrial outer membrane in inflammatory cells, we chose a Bcl-2 inhibitor, ABT-199, which can inhibit airway inflammation and airway hyperresponsiveness by inducing inflammatory cell apoptosis. Herein, we synthesized a pH-sensitive nanoformulated Bcl-2 inhibitor (Nf-ABT-199) that could specifically deliver ABT-199 to the mitochondria of bronchial inflammatory cells. The proof-of-concept study of an inflammatory cell mitochondria-targeted therapy using Nf-ABT-199 was validated in a mouse model of allergic asthma. Nf-ABT-199 was proven to significantly alleviate airway inflammation by effectively inducing eosinophil apoptosis and inhibiting both inflammatory cell infiltration and mucus hypersecretion. In addition, the nanocarrier or Nf-ABT-199 showed no obvious influence on cell viability, airway epithelial barrier and liver function, implying excellent biocompatibility and with non-toxic effect. The nanoformulated Bcl-2 inhibitor Nf-ABT-199 accumulates in the mitochondria of inflammatory cells and efficiently alleviates allergic asthma.
Collapse
Affiliation(s)
- Bao-Ping Tian
- Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Site of National Clinical Research Center for Respiratory Disease, Hangzhou, Zhejiang 310009, China; Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Fangyuan Li
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Ruiqing Li
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xi Hu
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Tian-Wen Lai
- Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Site of National Clinical Research Center for Respiratory Disease, Hangzhou, Zhejiang 310009, China
| | - Jingxiong Lu
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yun Zhao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Site of National Clinical Research Center for Respiratory Disease, Hangzhou, Zhejiang 310009, China
| | - Yang Du
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zeyu Liang
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Chen Zhu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Site of National Clinical Research Center for Respiratory Disease, Hangzhou, Zhejiang 310009, China
| | - Wei Shao
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Site of National Clinical Research Center for Respiratory Disease, Hangzhou, Zhejiang 310009, China
| | - Zhi-Hua Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Site of National Clinical Research Center for Respiratory Disease, Hangzhou, Zhejiang 310009, China
| | - Xiaolian Sun
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Songmin Ying
- Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Site of National Clinical Research Center for Respiratory Disease, Hangzhou, Zhejiang 310009, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| | - Daishun Ling
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Huahao Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Key Site of National Clinical Research Center for Respiratory Disease, Hangzhou, Zhejiang 310009, China; State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong 510120, China.
| |
Collapse
|
15
|
Li J, Saruta K, Dumouchel JP, Magat JM, Thomas JL, Ajami D, Rebek M, Rebek J, Bigby TD. Small Molecule Mimetics of α-Helical Domain of IRAK2 Attenuate the Proinflammatory Effects of IL-33 in Asthma-like Mouse Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:4036-4043. [PMID: 29728508 PMCID: PMC5988972 DOI: 10.4049/jimmunol.1700693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 04/10/2018] [Indexed: 01/04/2023]
Abstract
IL-33 and its receptor ST2 play important roles in airway inflammation and contribute to asthma onset and exacerbation. The IL-33/ST2 signaling pathway recruits adapter protein myeloid differentiation primary response 88 (MyD88) to transduce intracellular signaling. MyD88 forms a complex with IL-R-associated kinases (IRAKs), IRAK4 and IRAK2, called the Myddosome (MyD88-IRAK4-IRAK2). The myddosome subsequently activates downstream NF-κB and MAPKs p38 and JNK. We established an asthma-like mouse model by intratracheal administration of IL-33. The IL-33 model has a very similar phenotype compared with the OVA-induced mouse asthma model. The importance of MyD88 in the IL-33/ST2 signaling transduction was demonstrated by the MyD88 knockout mice, which were protected from the IL-33-induced asthma. We synthesized small molecule mimetics of the α-helical domain of IRAK2 with drug-like characteristics based on the recent advances in the designing of α-helix compounds. The mimetics can competitively interfere in the protein-protein interaction between IRAK2 and IRAK4, leading to disruption of Myddosome formation. A series of small molecules were screened using an NF-κB promoter assay in vitro. The lead compound, 7004, was further studied in the IL-33-induced and OVA-induced asthma mouse models in vivo. Compound 7004 can inhibit the IL-33-induced NF-κB activity, disrupt Myddosome formation, and attenuate the proinflammatory effects in asthma-like models. Our data indicate that the Myddosome may represent a novel intracellular therapeutic target for diseases in which IL-33/ST2 plays important roles, such as asthma and other inflammatory diseases.
Collapse
Affiliation(s)
- Jinghong Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093;
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Kunio Saruta
- The Scripps Research Institute, La Jolla, CA 92037
| | - Justin P Dumouchel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Jenna M Magat
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Joanna L Thomas
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | | | - Mitra Rebek
- The Scripps Research Institute, La Jolla, CA 92037
| | - Julius Rebek
- The Scripps Research Institute, La Jolla, CA 92037
| | - Timothy D Bigby
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| |
Collapse
|
16
|
Peters SP, Busse WW. New and Anticipated Therapies for Severe Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 5:S15-S24. [PMID: 28888244 DOI: 10.1016/j.jaip.2017.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/10/2017] [Accepted: 07/13/2017] [Indexed: 10/18/2022]
Abstract
Asthma is frequently undertreated, resulting in a relatively high prevalence of patients with uncontrolled disease, characterized by the presence of symptoms and risk of adverse outcomes. Patients with uncontrolled asthma have a higher risk of morbidity and mortality, underscoring the importance of identifying uncontrolled disease and modifying management plans to improve control. Several assessment tools exist to evaluate control with various cutoff points and measures, but these tools do not reliably correlate with physiological measures and should be considered a supplement to physiological tests. When attempting to improve control in patients, nonpharmacological interventions should always be attempted before changing or adding pharmacotherapies. Among patients with severe, uncontrolled asthma, individualized treatment based on asthma phenotype and eosinophil presence should be considered. The efficacy of the anti-IgE antibody omalizumab has been well established for patients with allergic asthma, and novel biologic agents targeting IL-5, IL-13, IL-4, and other allergic pathways have been investigated for patients with allergic or eosinophilic asthma. Fevipiprant (a CRTH2 [chemokine receptor homologous molecule expressed on Th2 cells] antagonist) and imatinib (a tyrosine kinase inhibition) are examples of nonbiologic therapies that may be useful for patients with severe, uncontrolled asthma. Incorporation of new and emerging treatment into therapeutic strategies for patients with severe asthma may improve outcomes for this patient population.
Collapse
Affiliation(s)
- Stephen P Peters
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC.
| | - William W Busse
- UW Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
17
|
Sánchez-Borges M, Martin BL, Muraro AM, Wood RA, Agache IO, Ansotegui IJ, Casale TB, Fleisher TA, Hellings PW, Papadopoulos NG, Peden DB, Sublett JL, Tilles SA, Rosenwasser L. The importance of allergic disease in public health: an iCAALL statement. World Allergy Organ J 2018; 11:8. [PMID: 29743965 PMCID: PMC5921992 DOI: 10.1186/s40413-018-0187-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 11/17/2022] Open
Affiliation(s)
- Mario Sánchez-Borges
- 1Allergy and Clinical Immunology Department, Centro Medico Docente La Trinidad, Allergy Service, Clinica El Avila, 6a. Transversal Urb. Altamira, Piso 8, Consultorio 803, Caracas, 1060 Venezuela
| | | | | | | | | | | | - Thomas B Casale
- 7University of South Florida Morsani College of Medicine, Tampa, FL USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Han H, Roan F, Ziegler SF. The atopic march: current insights into skin barrier dysfunction and epithelial cell-derived cytokines. Immunol Rev 2018; 278:116-130. [PMID: 28658558 DOI: 10.1111/imr.12546] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atopic dermatitis often precedes the development of other atopic diseases. The atopic march describes this temporal relationship in the natural history of atopic diseases. Although the pathophysiological mechanisms that underlie this relationship are poorly understood, epidemiological and genetic data have suggested that the skin might be an important route of sensitization to allergens. Animal models have begun to elucidate how skin barrier defects can lead to systemic allergen sensitization. Emerging data now suggest that epithelial cell-derived cytokines such as thymic stromal lymphopoietin (TSLP), IL-33, and IL-25 may drive the progression from atopic dermatitis to asthma and food allergy. This review focuses on current concepts of the role of skin barrier defects and epithelial cell-derived cytokines in the initiation and maintenance of allergic inflammation and the atopic march.
Collapse
Affiliation(s)
- Hongwei Han
- Immunology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Florence Roan
- Immunology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute, Seattle, WA, USA.,Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
19
|
Pascoe SJ, Wu W, Collison KA, Nelsen LM, Wurst KE, Lee LA. Use of clinical characteristics to predict spirometric classification of obstructive lung disease. Int J Chron Obstruct Pulmon Dis 2018; 13:889-902. [PMID: 29559773 PMCID: PMC5856300 DOI: 10.2147/copd.s153426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background There is no consensus on how to define patients with symptoms of asthma and chronic obstructive pulmonary disease (COPD). A diagnosis of asthma–COPD overlap (ACO) syndrome has been proposed, but its value is debated. This study (GSK Study 201703 [NCT02302417]) investigated the ability of statistical modeling approaches to define distinct disease groups in patients with obstructive lung disease (OLD) using medical history and spirometric data. Methods Patients aged ≥18 years with diagnoses of asthma and/or COPD were categorized into three groups: 1) asthma (nonobstructive; reversible), 2) ACO (obstructive; reversible), and 3) COPD (obstructive; nonreversible). Obstruction was defined as a post-bronchodilator forced expiratory volume in 1 second (FEV1)/forced vital capacity <0.7, and reversibility as a post-albuterol increase in FEV1 ≥200 mL and ≥12%. A primary model (PM), based on patients’ responses to a health care practitioner-administered questionnaire, was developed using multinomial logistic regression modeling. Other multivariate statistical analysis models for identifying asthma and COPD as distinct entities were developed and assessed using receiver operating characteristic (ROC) analysis. Partial least squares discriminant analysis (PLS-DA) assessed the degree of overlap between groups. Results The PM predicted spirometric classifications with modest sensitivity. Other analysis models performed with high discrimination (area under the ROC curve: asthma model, 0.94; COPD model, 0.87). PLS-DA identified distinct phenotypic groups corresponding to asthma and COPD. Conclusion Within the OLD spectrum, patients with asthma or COPD can be identified as two distinct groups with a high degree of precision. Patients outside these classifications do not constitute a homogeneous group.
Collapse
Affiliation(s)
- Steven J Pascoe
- Respiratory Medicines Development Center, GSK, Research Triangle Park, NC, USA
| | - Wei Wu
- Biostatistics, PAREXEL International, Research Triangle Park, NC, USA.,Clinical Statistics, GSK, Research Triangle Park, NC, USA
| | - Kathryn A Collison
- Respiratory Medicines Development Center, GSK, Research Triangle Park, NC, USA
| | | | | | | |
Collapse
|
20
|
Salter BMA, Smith SG, Mukherjee M, Plante S, Krisna S, Nusca G, Oliveria JP, Irshad A, Gauvreau GM, Chakir J, Nair P, Sehmi R. Human Bronchial Epithelial Cell-derived Factors from Severe Asthmatic Subjects Stimulate Eosinophil Differentiation. Am J Respir Cell Mol Biol 2018; 58:99-106. [PMID: 28853918 DOI: 10.1165/rcmb.2016-0262oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Activated bronchial epithelial cells (BEC) release various alarmins, including thymic stromal lymphopoietin (TSLP), that drive type 2 inflammation. We hypothesize that BEC-derived factors promote in situ eosinophil differentiation and maturation, a process that is driven by an IL-5-rich microenvironment in asthmatic airways. To assess the eosinophilopoietic potential of epithelial-derived factors, eosinophil/basophil colony forming units (Eo/B-CFU) were enumerated in 14-day methylcellulose cultures of blood-derived nonadherent mononuclear cells incubated with BEC supernatants (BECSN) from healthy nonatopic controls (n = 8), mild atopic asthmatics (n = 9), and severe asthmatics (n = 5). Receptor-blocking antibodies were used to evaluate the contribution of alarmins. Modulation of the mRNA expression of transcription factors that are crucial for eosinophil differentiation was evaluated. BECSN stimulated the clonogenic expansion of eosinophil progenitors in vitro. In the presence of IL-5, Eo/B-CFU numbers were significantly greater in cocultures of BESCN from severe asthmatics compared with other groups. This was attenuated in the presence of a TSLP (but not an IL-33) receptor-blocking antibody. Recombinant human TSLP (optimal at 100 pg/ml) stimulated Eo/B-CFU growth, which was significantly enhanced in the presence of IL-5 (1 ng/ml). Overnight culture of CD34+ cells with IL-5 and TSLP synergistically increased GATA-binding factor 2 and CCAAT/enhancer-binding protein α mRNA expression. The eosinophilopoietic potential of factors derived from BEC is increased in severe asthma. Our data suggest that TSLP is a key alarmin that is produced by BECs and promotes in situ eosinophilopoiesis in a type 2-rich microenvironment.
Collapse
Affiliation(s)
- Brittany M A Salter
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Steven G Smith
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Manali Mukherjee
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Sophie Plante
- 2 Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Sakktee Krisna
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Graeme Nusca
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - John Paul Oliveria
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Anam Irshad
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Gail M Gauvreau
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Jamila Chakir
- 2 Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Parameswaran Nair
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Roma Sehmi
- 1 Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| |
Collapse
|
21
|
Ntontsi P, Papathanassiou E, Loukides S, Bakakos P, Hillas G. Targeted anti-IL-13 therapies in asthma: current data and future perspectives. Expert Opin Investig Drugs 2018; 27:179-186. [PMID: 29334288 DOI: 10.1080/13543784.2018.1427729] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION The identification of patients with severe asthma who will benefit from a personalized management approach remains an unmet need. Interleukin-13 (IL-13) is a cytokine possessing a significant role in asthma pathogenesis and progression of disease. Humanised monoclonal antibodies against IL-13 and IL-13 and IL-4 receptors are mainly proposed as add-on therapy in patients with TH2-high inflammation with uncontrolled asthma despite maximum therapy. AREAS COVERED The role of IL-13 in airway inflammation in severe asthma, the targeted anti-IL-13 therapies and biomarkers that predict response to anti-IL-13 treatment are discussed. EXPERT OPINION New effective individualized therapies in severe asthma are urgently needed to block specific inflammatory pathways using monoclonal antibodies. Studies on anti-IL-13 therapies showed that asthmatic patients could benefit from this novel targeted therapy as far as lung function and exacerbation rate are concerned. TH2-high and especially periostin-high groups of asthmatics with moderate-to-severe uncontrolled asthma seem to compose the group that could benefit from anti-IL-13 therapy. Targeting IL-13 alone may not be sufficient to achieve asthma control. Inhibition of IL-13 and IL-4 with mabs may be more encouraging and patients will probably have additional benefits from these therapeutic interventions because of IL-13/IL-4 overlapping actions in asthma pathophysiology.
Collapse
Affiliation(s)
- Polyxeni Ntontsi
- a 2nd Respiratory Medicine Department , National and Kapodistrian University of Athens, Medical School, Attikon Hospital , Athens , Greece
| | - Evgenia Papathanassiou
- a 2nd Respiratory Medicine Department , National and Kapodistrian University of Athens, Medical School, Attikon Hospital , Athens , Greece
| | - Stelios Loukides
- a 2nd Respiratory Medicine Department , National and Kapodistrian University of Athens, Medical School, Attikon Hospital , Athens , Greece
| | - Petros Bakakos
- b 1st Respiratory Medicine Department , National and Kapodistrian University of Athens, Medical School, Sotiria Chest Hospital , Athens , Greece
| | - Georgios Hillas
- c Department of Critical Care and Pulmonary Services , National and Kapodistrian University of Athens, Medical School, Evangelismos Hospital , Athens , Greece
| |
Collapse
|
22
|
ANDO K, TANAKA A, MIKUNI H, KAWAHARA T, KUWAHARA N, MANABE R, JINNO M, MIYATA Y, HIRAI K, OHNISHI T, INOUE S, SAGARA H. Indirect Comparison of Dupilumab and Mepolizumab Treatments for Uncontrolled Eosinophilic Asthma. ACTA ACUST UNITED AC 2018. [DOI: 10.15369/sujms.30.189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Koichi ANDO
- Department of Medicine, Division of Respiratory Medicine and Allergology
- Showa University Dental Hospital Medical Clinic
| | - Akihiko TANAKA
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Hatsuko MIKUNI
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Tomoko KAWAHARA
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Naota KUWAHARA
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Ryo MANABE
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Megumi JINNO
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Yoshito MIYATA
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Kuniaki HIRAI
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Tsukasa OHNISHI
- Department of Medicine, Division of Respiratory Medicine and Allergology
| | - Shin INOUE
- Showa University Dental Hospital Medical Clinic
| | - Hironori SAGARA
- Department of Medicine, Division of Respiratory Medicine and Allergology
| |
Collapse
|
23
|
Tian BP, Zhang GS, Lou J, Zhou HB, Cui W. Efficacy and safety of benralizumab for eosinophilic asthma: A systematic review and meta-analysis of randomized controlled trials. J Asthma 2017; 55:956-965. [PMID: 29211545 DOI: 10.1080/02770903.2017.1379534] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
CONTEXT Benralizumab is a humanized monoclonal antibody that targets the α chain of the IL-5 receptor (IL-5Rα) and is currently being assessed in clinical trials for asthma control. OBJECTIVE Our systematic review and meta-analysis intends to evaluate the therapeutic efficacy and safety of benralizumab in patients with eosinophilic asthma. DATA SOURCES AND EXTRACTION Literature searches of PubMed, Embase, and the Cochrane Library were performed to identify randomized controlled trials of benralizumab and clinic outcomes in asthmatics. RESULTS In total, 7 articles with 2,321 subjects met our inclusion criteria. From this pooled analysis, we found that benralizumab significantly reduces exacerbations (RR: 0.63, 95% CI: 0.52-0.76, p < 0.00001; I2 = 52%, p = 0.06) compared to placebo in eosinophilic asthma. There was no statistical trend for improvement in forced expiratory volume in 1 second or asthma control indices such as Quality of Life Assessment (AQLQ) and Asthma Control Questionnaire score in benralizumab-treated patients. In addition, safety data indicated that benralizumab administration resulted no increasing incidence of adverse events and was well tolerated (RR: 1.00, 95% CI: 0.95-1.05, p = 0.96; I2 = 40%, p = 0.13). CONCLUSION These results demonstrate the efficacy and safety of benralizumab for asthma patients with severe or uncontrolled symptoms and elevated eosinophils and provide support for benralizumab as an ideal option to treat asthma in this patient population.
Collapse
Affiliation(s)
- Bao-Ping Tian
- a Department of Critical Care Medicine , Second Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , China
| | - Gen-Sheng Zhang
- a Department of Critical Care Medicine , Second Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , China
| | - Jian Lou
- a Department of Critical Care Medicine , Second Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , China
| | - Hong-Bin Zhou
- b Department of Respiratory Medicine , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , China.,c People's Hospital of Hangzhou Medical College , Hangzhou , Zhejiang , China
| | - Wei Cui
- a Department of Critical Care Medicine , Second Affiliated Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , China
| |
Collapse
|
24
|
Gonzalez-Barcala FJ, Calvo-Alvarez U, Salgado-Castro FJ, Facal D, Garcia-Sanz MT, Muñoz X, Garcia-Couceiro N, Paz-Neira O, San-Jose E, Valdes-Cuadrado L, Carreira JM. Asthma exacerbations: factors related to longer hospital stay. Acta Clin Belg 2017; 72:379-384. [PMID: 28245723 DOI: 10.1080/17843286.2017.1295524] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Hospital admissions account for a significant part of asthma cost, but with significant differences between geographic areas. AIMS The aim of our study is to analyse hospital admissions due to asthma, as well as the factors associated with longer hospital stays. METHODS A review was retrospectively carried out on all admissions of patients over 18 years old due to exacerbation of asthma occurring in our hospital between the years 2000 and 2010. The personal characteristics of each patient, the asthma personal history, characteristics of every exacerbation, as well as the treatment before admission and after hospital discharge were recorded. RESULTS During the study period, there were 2163 hospital admissions in 1316 patients (mean age 62.6 years; mean hospital stay 11.6 days). The admissions mainly occur in winter, in the 56-75-year age group, and in patients with severe asthma. Female sex, higher comorbidity, a greater number of emergencies due to asthma in the previous year, and baseline treatment with theophylline were independently associated to longer hospital stay. CONCLUSIONS The management of asthma in our population seems improvable. There appears to be a need to optimise both the diagnosis and treatment of the disease, and to identify risk factors as important as tobacco habits. As regards exacerbations, the hospital stay and mortality must be significantly reduced.
Collapse
Affiliation(s)
- Francisco-Javier Gonzalez-Barcala
- Department of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
- Spanish Biomedical Research Networking Centre, CIBERES, Santiago de Compostela, Spain
- Department of Respiratory Medicine, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Uxio Calvo-Alvarez
- Department of Respiratory Medicine, Hospital of El Ferrol, El Ferrol, Spain
| | - Francisco-Javier Salgado-Castro
- Faculty of Biology/CIBUS, Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - David Facal
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Department of Developmental and Educational Psychology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Xavier Muñoz
- Spanish Biomedical Research Networking Centre, CIBERES, Santiago de Compostela, Spain
- Department of Medicine, University Autònoma of Barcelona, Barcelona, Spain
- Department of Respiratory Medicine, Hospital Vall d’Hebron, Barcelona, Spain
| | - Nuria Garcia-Couceiro
- Department of Respiratory Medicine, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Olalla Paz-Neira
- Department of Respiratory Medicine, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Esther San-Jose
- Central Laboratory, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Luis Valdes-Cuadrado
- Department of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Respiratory Medicine, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Jose-Martin Carreira
- Department of Radiology, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
25
|
Efficacy and safety of benralizumab in patients with eosinophilic asthma: a meta-analysis of randomized placebo-controlled trials. Front Med 2017; 12:340-349. [PMID: 29086236 DOI: 10.1007/s11684-017-0565-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 05/12/2017] [Indexed: 02/05/2023]
Abstract
Benralizumab is a monoclonal antibody that targets interleukin-5 receptor α to deplete blood eosinophils and improve the clinical outcomes of allergic asthma. We conducted a meta-analysis to evaluate the safety and efficacy of different doses of benralizumab in patients with eosinophilic asthma. All randomized controlled trials involving benralizumab treatment for patients with eosinophilic asthma, which were searched in PubMed, Embase, and the Cochrane Library published until January 2017, as well as the rate of asthmatic exacerbation, pulmonary functionality, asthma control, quality of life scores, and adverse events were included. Randomized-effect models were used in the meta-analysis to calculate the pooled mean difference, relative risks, and 95% confidence intervals. Five studies involving 1951 patients were identified. Compared with the placebo, benralizumab treatment demonstrated significant improvements in the forced expiratory volume in 1 s (FEV1), Asthma Quality of Life Questionnaire scores, decreased asthmatic exacerbation and Asthma Control Questionnaire-6 (ACQ-6) scores. Benralizumab treatment was also not associated with increased adverse events. These findings indicated that benralizumab can be safely used to improve FEV1, enhance patient symptom control and quality of life, and reduce the risk of exacerbations and ACQ-6 scores in patients with eosinophilic asthma. Furthermore, our meta-analysis showed that benralizumab with 30 mg (every eight weeks) dosage can improve the health-related quality of life and appear to be more effective than 30 mg (every four weeks) dosage. Overall, data indicated that the optimal dosing regimen for benralizumab was possibly 30 mg (every eight weeks).
Collapse
|
26
|
Belvisi MG, Birrell MA. The emerging role of transient receptor potential channels in chronic lung disease. Eur Respir J 2017; 50:50/2/1601357. [PMID: 28775042 DOI: 10.1183/13993003.01357-2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/14/2017] [Indexed: 12/12/2022]
Abstract
Chronic lung diseases such as asthma, chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis are a major and increasing global health burden with a high unmet need. Drug discovery efforts in this area have been largely disappointing and so new therapeutic targets are needed. Transient receptor potential ion channels are emerging as possible therapeutic targets, given their widespread expression in the lung, their role in the modulation of inflammatory and structural changes and in the production of respiratory symptoms, such as bronchospasm and cough, seen in chronic lung disease.
Collapse
Affiliation(s)
- Maria G Belvisi
- Respiratory Pharmacology Group, Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| | - Mark A Birrell
- Respiratory Pharmacology Group, Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
27
|
Tworek D, Heroux D, O'Byrne SN, Mitchell P, O'Byrne PM, Denburg JA. Toll-like receptor-induced expression of epithelial cytokine receptors on haemopoietic progenitors is altered in allergic asthma. Clin Exp Allergy 2017; 47:900-908. [PMID: 28252235 DOI: 10.1111/cea.12913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 01/13/2017] [Accepted: 02/17/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Haemopoietic progenitor cells (HPC) migrate to sites of allergic inflammation where, upon stimulation with epithelial cytokines, they produce Th2 cytokines and differentiate into mature eosinophils and basophils. They also express Toll-like receptors (TLR) involved in antimicrobial responses. OBJECTIVE The objective of this study was to compare TLR expression on peripheral blood HPC and TLR-induced responses, in particular changes in epithelial cytokine receptors, in healthy and asthmatic subjects at baseline and following allergen challenge. METHODS Ten healthy and 11 allergic asthmatic subjects were studied. HPC-enriched cell populations were stimulated with TLR-2, TLR-4 or TLR-9 ligands. TLR expression by circulating HPC and interleukin (IL)-25 (IL-17RB), IL-33 (ST2) and thymic stromal lymphopoietin receptor (TSLPR) expression after TLR ligation were examined by flow cytometry at baseline and, in asthmatics, following allergen challenge. The effects of dexamethasone (Dex) on TLR-induced responses were also assessed. RESULTS Asthmatics had significantly lower circulating HPC expressing TLR-2 and TLR-9 with a similar trend for TLR-4. TLR-4 stimulation of HPC yielded higher numbers of TSLPR+ cells in asthmatics compared with healthy subjects. A similar trend was seen for TLR-9 ligation, an effect further augmented by allergen inhalation. Allergen challenge also enhanced TLR-induced ST2 expression on HPC. Treatment with Dex in vitro increased TLR-4-induced TSLPR expression but had no effect on other epithelial cytokine receptors. CONCLUSIONS AND CLINICAL RELEVANCE These data demonstrate an interaction between allergen and TLR ligand exposure in asthmatics. Allergen inhalation augments the TLR-induced inflammatory response by HPC, possibly leading to increased "in situ haemopoiesis" through up-regulation of TSLPR. These findings show that HPC may be a part of the pro-inflammatory cascade in pathogen-induced asthma exacerbation through their increased responsiveness to TLR stimulation.
Collapse
Affiliation(s)
- D Tworek
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,Department of Internal Diseases, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| | - D Heroux
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - S N O'Byrne
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - P Mitchell
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - P M O'Byrne
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - J A Denburg
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
28
|
Katial RK, Bensch GW, Busse WW, Chipps BE, Denson JL, Gerber AN, Jacobs JS, Kraft M, Martin RJ, Nair P, Wechsler ME. Changing Paradigms in the Treatment of Severe Asthma: The Role of Biologic Therapies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2017; 5:S1-S14. [PMID: 28143691 DOI: 10.1016/j.jaip.2016.11.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 11/30/2022]
Abstract
Cytokine antagonists are monoclonal antibodies that offer new treatment options for refractory asthma but will also increase complexity because they are effective only for patients with certain asthma subtypes that remain to be more clearly defined. The clinical and inflammatory heterogeneity within refractory asthma makes it difficult to manage the disease and to determine which, if any, biologic therapy is suitable for a specific patient. The purpose of this article is to provide a data-driven discussion to clarify the use of biologic therapies in patients with refractory asthma. We first discuss the epidemiology and pathophysiology of refractory asthma. We then interpret current evidence for biomarkers of eosinophilic or type 2-high asthma so that clinicians can determine potential treatments for patients based on knowledge of their effectiveness in specific asthma phenotypes. We then assess clinical data on the efficacy, safety, and mechanisms of action of approved and pipeline biologic therapies. We conclude by discussing the potential of phenotyping or endotyping refractory asthma and how biologic therapies can play a role in treating patients with refractory asthma.
Collapse
Affiliation(s)
- Rohit K Katial
- Department of Medicine, Division of Allergy and Clinical Immunology, National Jewish Health, Denver, Colo.
| | - Greg W Bensch
- Allergy, Immunology and Asthma Medical Group, Stockton, Calif
| | - William W Busse
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Bradley E Chipps
- Capital Allergy and Respiratory Disease Center, Sacramento, Calif
| | - Joshua L Denson
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colo; Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado at Denver, Anschutz Medical Campus, Aurora, Colo
| | - Anthony N Gerber
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colo; Department of Biomedical Research, National Jewish Health, Denver, Colo
| | - Joshua S Jacobs
- Allergy and Asthma Clinical Research, Inc., Walnut Creek, Calif
| | - Monica Kraft
- Department of Medicine, Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Ariz
| | | | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael E Wechsler
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colo
| |
Collapse
|
29
|
|
30
|
Efficacy and Safety of Anti-Interleukin-5 Therapy in Patients with Asthma: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0166833. [PMID: 27875559 PMCID: PMC5119789 DOI: 10.1371/journal.pone.0166833] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/05/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Recent trials have assessed the efficacy and safety of novel monoclonal antibodies such as reslizumab and benralizumab. However, the overall efficacy and safety anti-interleukin (IL) 5 treatment in asthma have not been thoroughly assessed. METHODS Randomized controlled trials (RCTs) of anti-IL-5 treatment on patients with asthma published up to October 2016 in PubMed, Embase, and Cochrane Central Register of Controlled Trials (CENTRAL) that reported pulmonary function, quality of life scores, asthmatic exacerbation rate, blood and sputum eosinophil counts, short-acting β-agonist (SABA) rescue use, and adverse events were included. The pooled mean difference, and relative risks (RR), and 95% confidence intervals (CIs) were calculated using random-effects models. RESULTS Twenty studies involving 7100 patients were identified. Pooled analysis revealed significant improvements in FEV1 (first second forced expiratory volume) (MD = 0.09, 95% CI: 0.06-0.12, I2 = 10%), FEV1% (MD = 3.75, 95% CI: 1.66-5.83, I2 = 19%), Asthma Quality of Life Questionnaire (AQLQ) score (MD = 0.22, 95% CI: 0.15-0.30, I2 = 0%), decreased blood, sputum eosinophils and asthmatic exacerbation (RR = 0.66, 95% CI: 0.59-0.73, I2 = 51%); peak expiratory flow (PEF) (MD = 5.45, 95% CI: -2.83-13.72, I2 = 0%), histamine PC20 (MD = -0.62, 95% CI: -1.92-0.68, I2 = 0%) or SABA rescue use (MD = -0.11, 95% CI: -0.3-0.07, I2 = 30%) were unaffected; adverse events were not increased (RR = 0.93, 95% CI: 0.89-0.98, I2 = 46%). No publication bias was observed (Egger's P = 0.78). CONCLUSIONS Anti-interleukin 5 monoclonal therapies for asthma could be safe for slightly improving FEV1 (or FEV1% of predicted value), quality of life, and reducing exacerbations risk and blood and sputum eosinophils, but have no significant effect on PEF, histamine PC20, and SABA rescue use. Further trials required to establish to clarify the optimal antibody for different patients.
Collapse
|
31
|
Matera MG, Page C, Rogliani P, Calzetta L, Cazzola M. Therapeutic Monoclonal Antibodies for the Treatment of Chronic Obstructive Pulmonary Disease. Drugs 2016; 76:1257-1270. [DOI: 10.1007/s40265-016-0625-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
32
|
Pera T, Penn RB. Bronchoprotection and bronchorelaxation in asthma: New targets, and new ways to target the old ones. Pharmacol Ther 2016; 164:82-96. [PMID: 27113408 PMCID: PMC4942340 DOI: 10.1016/j.pharmthera.2016.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/07/2016] [Indexed: 01/01/2023]
Abstract
Despite over 50years of inhaled beta-agonists and corticosteroids as the default management or rescue drugs for asthma, recent research suggests that new therapeutic options are likely to emerge. This belief stems from both an improved understanding of what causes and regulates airway smooth muscle (ASM) contraction, and the identification of new targets whose inhibition or activation can relax ASM. In this review we discuss the recent findings that provide new insight into ASM contractile regulation, a revolution in pharmacology that identifies new ways to "tune" G protein-coupled receptors to improve therapeutic efficacy, and the discovery of several novel targets/approaches capable of effecting bronchoprotection or bronchodilation.
Collapse
Affiliation(s)
- Tonio Pera
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| | - Raymond B Penn
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
33
|
Pelaia G, Vatrella A, Busceti MT, Gallelli L, Preianò M, Lombardo N, Terracciano R, Maselli R. Role of biologics in severe eosinophilic asthma - focus on reslizumab. Ther Clin Risk Manag 2016; 12:1075-82. [PMID: 27445482 PMCID: PMC4936812 DOI: 10.2147/tcrm.s111862] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Within the context of the heterogeneous phenotypic stratification of asthmatic population, many patients are characterized by moderate-to-severe eosinophilic asthma, not adequately controlled by relatively high dosages of inhaled and even oral corticosteroids. Therefore, these subjects can obtain significant therapeutic benefits by additional biologic treatments targeting interleukin-5 (IL-5), given the key pathogenic role played by this cytokine in maturation, activation, proliferation, and survival of eosinophils. In particular, reslizumab is a humanized anti-IL-5 monoclonal antibody that has been found to be an effective and safe add-on therapy, capable of decreasing asthma exacerbations and significantly improving disease control and lung function in patients experiencing persistent allergic or nonallergic eosinophilic asthma, despite the regular use of moderate-to-high doses of inhaled corticosteroids. These important therapeutic effects of reslizumab, demonstrated by several controlled clinical trials, have led to the recent approval by US Food and Drug Administration of its use, together with other antiasthma medications, for the maintenance treatment of patients suffering from severe uncontrolled asthma.
Collapse
Affiliation(s)
- Girolamo Pelaia
- Department of Medical and Surgical Sciences, Section of Respiratory Diseases, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Alessandro Vatrella
- Department of Medicine and Surgery, Section of Respiratory Diseases, University of Salerno, Salerno, Italy
| | - Maria Teresa Busceti
- Department of Medical and Surgical Sciences, Section of Respiratory Diseases, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Luca Gallelli
- Department of Health Science, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | | | - Nicola Lombardo
- Department of Medical and Surgical Sciences, Section of Respiratory Diseases, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Rosa Terracciano
- Department of Health Science, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Rosario Maselli
- Department of Medical and Surgical Sciences, Section of Respiratory Diseases, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
34
|
Waldman SA, Terzic A. Bioinnovation Enterprise: An engine driving breakthrough therapies. Clin Pharmacol Ther 2016; 99:8-13. [PMID: 26785918 DOI: 10.1002/cpt.272] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/29/2015] [Indexed: 12/14/2022]
Abstract
Biological advances have radically expanded our insights into the underpinnings of health and disease. New knowledge has formed the substrate for translation-expedited in turn by the biotechnology and pharmaceutical industry into novel therapeutic solutions impacting the management of patients and populations. Indeed, this Bioinnovation Enterprise has become the dominant growth sector in drug development and the engine driving the translation of breakthrough therapies worldwide. This annual Therapeutic Innovations issue highlights recent exceptional advances by the Bioinnovation Enterprise in translating molecular insights in pathobiology into transformative therapies.
Collapse
Affiliation(s)
- S A Waldman
- Department of Pharmacology and Experimental Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - A Terzic
- Mayo Clinic Center for Regenerative Medicine, Divisions of Cardiovascular Diseases and Clinical Pharmacology, Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics and Medical Genetics, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
35
|
Development of therapeutic antibodies to G protein-coupled receptors and ion channels: Opportunities, challenges and their therapeutic potential in respiratory diseases. Pharmacol Ther 2016; 169:113-123. [PMID: 27153991 DOI: 10.1016/j.pharmthera.2016.04.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of recombinant antibody therapeutics continues to be a significant area of growth in the pharmaceutical industry with almost 50 approved monoclonal antibodies on the market in the US and Europe. Therapeutic drug targets such as soluble cytokines, growth factors and single transmembrane spanning receptors have been successfully targeted by recombinant monoclonal antibodies and the development of new product candidates continues. Despite this growth, however, certain classes of important disease targets have remained intractable to therapeutic antibodies due to the complexity of the target molecules. These complex target molecules include G protein-coupled receptors and ion channels which represent a large target class for therapeutic intervention with monoclonal antibodies. Although these targets have typically been addressed by small molecule approaches, the exquisite specificity of antibodies provides a significant opportunity to provide selective modulation of these important regulators of cell function. Given this opportunity, a significant effort has been applied to address the challenges of targeting these complex molecules and a number of targets are linked to the pathophysiology of respiratory diseases. In this review, we provide a summary of the importance of GPCRs and ion channels involved in respiratory disease and discuss advantages offered by antibodies as therapeutics at these targets. We highlight some recent GPCRs and ion channels linked to respiratory disease mechanisms and describe in detail recent progress made in the strategies for discovery of functional antibodies against challenging membrane protein targets such as GPCRs and ion channels.
Collapse
|