1
|
Pasha MA, Alnabulsi R, Wan A, Hopp RJ, Yang Q. Dual role of mucosal-associated invariant T cells (MAIT) in asthma. J Asthma 2025:1-6. [PMID: 39969254 DOI: 10.1080/02770903.2025.2469319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/15/2025] [Indexed: 02/20/2025]
Abstract
OBJECTIVE Mucosal-associated invariant T cells (MAIT) are the predominant type of innate-like T cells in humans, and they represent a unique subset of microbiota-dependent invariant T cells. This Commentary reviews recent clinical studies and animal model research elucidating the multifaceted roles of MAIT cells in asthma. METHOD A literature search was performed using PubMed and Google Scholar, and covered the period from 1960 to 2024. The search yielded more than 50 articles, and only essential original research articles and selected review articles were evaluated. RESULTS Recent studies indicate that MAIT cell-derived effector molecules may play dual roles in asthma and allergic airway inflammation. While MAIT cells can produce the anti-inflammatory enzyme IL4I1 and the Th1 cytokine IFN-γ to repress allergic airway inflammation and airway hyperresponsiveness (AHR), they may also secrete IL-17. Which induces neutrophil infiltration and exacerbates AHR. In addition, some clinical studies from the literature search revealed a negative association between MAIT cell abundance and asthma. Regarding allergic airway inflammation, mouse model studies suggested that MAIT cells may play a protective role. CONCLUSION These findings raise critical questions as to whether MAIT cells are friend or foe in asthma, and whether distinct subsets of MAIT cells play different roles in allergic airway inflammation. Further studies are needed to better understand the implication of MAIT cells in asthma pathogenesis, particularly in patients with severe asthma.
Collapse
Affiliation(s)
- M Asghar Pasha
- Division of Allergy and Immunology, Department of Medicine, Albany Medical College, Albany, NY, USA
| | - Rawaa Alnabulsi
- Division of Allergy and Immunology, Department of Medicine, Albany Medical College, Albany, NY, USA
| | - Aiden Wan
- Department of Pediatrics, Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Russell J Hopp
- Department of Pediatrics, University of NE Medical Center and Children's Hospital and Medical Center, Omaha, NE, USA
| | - Qi Yang
- Department of Pediatrics, Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
2
|
Datsyuk JK, De Rubis G, Paudel KR, Kokkinis S, Oliver BGG, Dua K. Cellular probing using phytoceuticals encapsulated advanced delivery systems in ameliorating lung diseases: Current trends and future prospects. Int Immunopharmacol 2024; 141:112913. [PMID: 39137633 DOI: 10.1016/j.intimp.2024.112913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
Chronic respiratory diseases such as Chronic Obstructive Pulmonary Disease (COPD) and asthma have posed a significant healthcare and economic cost over a prolonged duration worldwide. At present, available treatments are limited to a range of preventive medicines, such as mono- or multiple-drug therapy, which necessitates daily use and are not considered as viable treatments to reverse the inflammatory processes of airway remodelling which is inclusive of the alteration of intra and extracellular matrix of the airway tract, death of epithelial cells, the increase in smooth muscle cell and the activation of fibroblasts. Hence, with the problem in mind a considerable body of study has been dedicated to comprehending the underlying factors that contribute to inflammation within the framework of these disorders. Hence, adequate literature that has unveiled the necessary cellular probing to reduce inflammation in the respiratory tract by improving the selectivity and precision of a novel treatment. However, through cellular probing cellular mechanisms such as the downregulation of various markers, interleukin 8, (IL-8), Interleukin 6 (IL-6), interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α) have been uncovered. Hence, to target such cellular probes implementation of phytoceuticals encapsulated in an advanced drug delivery system has shown potential to be a solution with in vitro and in vivo studies highlighting their anti-inflammatory and antioxidant effects. However, the high costs associated with advanced drug delivery systems and the limited literature focused exclusively on nanoparticles pose significant challenges. Additionally, the biochemical characteristics of phytoceuticals due to poor solubility, limited bioavailability, and difficulties in mass production makes it difficult to implement this product as a treatment for COPD and asthma. This study aims to examine the integration of many critical features in the context of their application for the treatment of chronic inflammation in respiratory disorders.
Collapse
Affiliation(s)
- Jessica Katrine Datsyuk
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sofia Kokkinis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Brian Gregory George Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, New South Wales, Australia; School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| |
Collapse
|
3
|
Liao JM, Hong S, Wang Y, Cheng Y, Ho K, Toh S, Shih O, Jeng U, Lyu P, Hu I, Huang M, Chang C, Cheng T. Integrating molecular dynamics simulation with small- and wide-angle X-ray scattering to unravel the flexibility, antigen-blocking, and protease-restoring functions in a hindrance-based pro-antibody. Protein Sci 2024; 33:e5124. [PMID: 39145427 PMCID: PMC11325194 DOI: 10.1002/pro.5124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/11/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Spatial hindrance-based pro-antibodies (pro-Abs) are engineered antibodies to reduce monoclonal antibodies' (mAbs) on-target toxicity using universal designed blocking segments that mask mAb antigen-binding sites through spatial hindrance. By linking through protease substrates and linkers, these blocking segments can be removed site-specifically. Although many types of blocking segments have been developed, such as coiled-coil and hinge-based Ab locks, the molecular structure of the pro-Ab, particularly the region showing how the blocking fragment blocks the mAb, has not been elucidated by X-ray crystallography or cryo-EM. To achieve maximal effect, a pro-Ab must have high antigen-blocking and protease-restoring efficiencies, but the unclear structure limits its further optimization. Here, we utilized molecular dynamics (MD) simulations to study the dynamic structures of a hinge-based Ab lock pro-Ab, pro-Nivolumab, and validated the simulated structures with small- and wide-angle X-ray scattering (SWAXS). The MD results were closely consistent with SWAXS data (χ2 best-fit = 1.845, χ2 allMD = 3.080). The further analysis shows a pronounced flexibility of the Ab lock (root-mean-square deviation = 10.90 Å), yet it still masks the important antigen-binding residues by 57.3%-88.4%, explaining its 250-folded antigen-blocking efficiency. The introduced protease accessible surface area method affirmed better protease efficiency for light chain (33.03 Å2) over heavy chain (5.06 Å2), which aligns with the experiments. Overall, we developed MD-SWAXS validation method to study the dynamics of flexible blocking segments and introduced methodologies to estimate their antigen-blocking and protease-restoring efficiencies, which would potentially be advancing the clinical applications of any spatial hindrance-based pro-Ab.
Collapse
Affiliation(s)
- Jun Min Liao
- Drug Development and Value Creation Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
| | - Shih‐Ting Hong
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yeng‐Tseng Wang
- Department of BiochemistryKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yi‐An Cheng
- Drug Development and Value Creation Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Biomedical Science and Environmental BiologyKaohsiung Medical UniversityKaohsiungTaiwan
- Precisemab Biotech Co. LtdTaipeiTaiwan
| | - Kai‐Wen Ho
- Drug Development and Value Creation Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
| | - Shu‐Ing Toh
- Department of Biological Science and TechnologyNational Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Orion Shih
- National Synchrotron Radiation Research CenterHsinchu Science ParkHsinchuTaiwan
| | - U‐Ser Jeng
- National Synchrotron Radiation Research CenterHsinchu Science ParkHsinchuTaiwan
- Department of Chemical Engineering &College of Semiconductor ResearchNational Tsing Hua UniversityHsinchuTaiwan
| | - Ping‐Chiang Lyu
- Institute of Bioinformatics and Structural BiologyNational Tsing Hua UniversityHsinchuTaiwan
| | - I‐Chen Hu
- Institute of Bioinformatics and Structural BiologyNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Yii Huang
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Radiation OncologyKaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiungTaiwan
| | - Chin‐Yuan Chang
- Department of Biomedical Science and Environmental BiologyKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Biological Science and TechnologyNational Yang Ming Chiao Tung UniversityHsinchuTaiwan
- Center for Intelligent Drug Systems and Smart Bio‐devicesNational Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Tian‐Lu Cheng
- Drug Development and Value Creation Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Biomedical Science and Environmental BiologyKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Medical ResearchKaohsiung Medical University HospitalKaohsiungTaiwan
| |
Collapse
|
4
|
Rodríguez Parejo G, Carmona González M, Montero-Peña C, Murillo García D. [Assessment of blood eosinophil count in respiratory disease from primary care]. Semergen 2024; 50:102193. [PMID: 38484418 DOI: 10.1016/j.semerg.2024.102193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 09/06/2024]
Abstract
Most physicians in general, and family physicians in particular, are familiar with certain parameters when ordering a hematological study, such as hemoglobin (including hematocrit and its features), leukocytes (including lymphocytes) and platelets. Nevertheless, there are two values that we use to overlook which are eosinophils and basophils. Specifically, eosinophils have a tendency to increase with allergic pathology. This article focuses on this type of cells, helping to interpret the values obtained and highlighting their importance in two of the most frequent respiratory pathologies in primary care: asthma and COPD. In addition to observing how the increase or normality of these parameters condition the diagnosis, phenotype and even the treatment.
Collapse
Affiliation(s)
- G Rodríguez Parejo
- Medicina Familiar y Comunitaria, Centro de Salud Don Benito Oeste, Badajoz, España
| | - M Carmona González
- Medicina Familiar y Comunitaria, Centro de Salud Don Benito Oeste, Badajoz, España
| | - C Montero-Peña
- Medicina Familiar y Comunitaria, Centro de Salud Don Benito Oeste, Badajoz, España.
| | - D Murillo García
- Medicina Familiar y Comunitaria, Centro de Salud Pueblonuevo del Guadiana, Badajoz, España
| |
Collapse
|
5
|
Yilmaz D, Sahin E. All-cause mortality and related factors in patients with varying degrees of peripheral blood eosinophilia. Medicine (Baltimore) 2024; 103:e38359. [PMID: 39259127 PMCID: PMC11142823 DOI: 10.1097/md.0000000000038359] [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: 11/02/2023] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 09/12/2024] Open
Abstract
The aim of this study was to investigate all-cause mortality rates and related factors in patients with different levels of eosinophilia. This retrospective cohort study was conducted between January 2020 and December 2022 in the Internal Medicine Department of Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkiye. A total of 161 patients with eosinophilia (at least 3 times) were included and divided into groups with absolute eosinophil counts of 500-999/µL (mild), 1000-1500/µL (moderate), and >1500/µL (severe). The mean age of patients was 65.67 ± 16.64 years at the time of admission, and 45 patients (57.8%) were male. The rates of mortality, oncological disease, and organ involvement were significantly higher in the severe group (P < .05). Increased serum total immunoglobulin E and vitamin B12, hematocrit value, eosinophil-to-lymphocyte ratio, and leukocyte were observed in eosinophilic patients. Decreased lymphocyte count, hemoglobin and hematocrit values were higher in deceased patients than in survivors (P < .05). Increased eosinophil-to-lymphocyte ratio, C-reactive protein, vitamin B12, and lactate dehydrogenase (LDH) activity were observed in participants who died compared to those who survived (P < .05). Multivariable logistic regression revealed that advanced age and higher LDH activity were independently associated with greater mortality risk while receiving non-steroid anti-inflammatory drugs or proton-pump inhibitors were associated with reduced mortality risk (P < .05). Advanced age and increased LDH activity were independently associated with greater risk for mortality, whereas absolute eosinophil counts was not. Considering the literature on this topic, our results show the need for further clinical and fundamental research to understand the role of eosinophils in human disease.
Collapse
Affiliation(s)
- Deniz Yilmaz
- Department of Internal Diseases, University of Health Sciences Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Ezgi Sahin
- Department of Rheumatology, Istanbul University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
6
|
Pan Q, Sun J, Gao S, Liu Z, Huang Y, Lian Y. Incremental long-term benefit of drug therapies for chronic obstructive pulmonary disease in quality of life but not mortality: a network meta-analysis. Arch Med Sci 2024; 20:1586-1596. [PMID: 39649253 PMCID: PMC11623174 DOI: 10.5114/aoms/183025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/23/2024] [Indexed: 12/10/2024] Open
Abstract
Introduction The latest evidence revealed that dupilumab, an interleukin-4 (IL-4) and interleukin-13 (IL-13) blocker, significantly reduces the exacerbation risk in patients with chronic obstructive pulmonary disease (COPD). The efficacy of dupilumab compared with conventional inhaled drugs remains incompletely determined. This study aimed to investigate the comparative efficacy of dupilumab and conventional inhaled drugs in patients with stable COPD. Material and methods This study retrieved randomised clinical trials (RCTs) with follow-up ≥ 48 weeks on long-acting β-agonists (LABAs), long-acting muscarinic receptor antagonists (LAMAs), inhaled corticosteroids (ICSs), and dupilumab in the PubMed, EMBASE, and Cochrane Central Register of Controlled Trials databases. The information on eligible studies was extracted after the screening. The comparative efficacy of 4 drugs and their combinations in acute exacerbation and mortality was assessed using Bayesian network meta-analysis models. Results This network meta-analysis identified 69 eligible RCTs on 7 classes of drug therapies after stepwise screening and included 125,331 COPD patients. Compared with placebo, the 7 drug interventions significantly reduced the risk of acute exacerbation, and the reduction degree increased with the incremental use of drug classes. ICS/LABA/LAMA/dupilumab was the most effective in decreasing exacerbation risk (OR = 0.561 [95% CI: 0.387-0.810]), followed by ICS/LABA/LAMA (OR = 0.717 [95% CI: 0.626-0.817]). The 7 drug therapies were not significantly associated with a lower risk of death compared to placebo. Nevertheless, ICS/LABA/LAMA/dupilumab is the most likely to be effective in decreasing mortality. Conclusions The incremental use of combinations of conventional and novel drugs contributed to the long-term benefits in acute exacerbation but not death in COPD. The optimal drug combination in terms of acute COPD exacerbation was ICS/LABA/LAMA/dupilumab.
Collapse
Affiliation(s)
- Qiong Pan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jiongzhou Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Shiyuan Gao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zian Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yiwen Huang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yixin Lian
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
7
|
Calzetta L, Page C, Matera MG, Cazzola M, Rogliani P. Use of human airway smooth muscle in vitro and ex vivo to investigate drugs for the treatment of chronic obstructive respiratory disorders. Br J Pharmacol 2024; 181:610-639. [PMID: 37859567 DOI: 10.1111/bph.16272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/21/2023] Open
Abstract
Isolated airway smooth muscle has been extensively investigated since 1840 to understand the pharmacology of airway diseases. There has often been poor predictability from murine experiments to drugs evaluated in patients with asthma or chronic obstructive pulmonary disease (COPD). However, the use of isolated human airways represents a sensible strategy to optimise the development of innovative molecules for the treatment of respiratory diseases. This review aims to provide updated evidence on the current uses of isolated human airways in validated in vitro methods to investigate drugs in development for the treatment of chronic obstructive respiratory disorders. This review also provides historical notes on the pioneering pharmacological research on isolated human airway tissues, the key differences between human and animal airways, as well as the pivotal differences between human medium bronchi and small airways. Experiments carried out with isolated human bronchial tissues in vitro and ex vivo replicate many of the main anatomical, pathophysiological, mechanical and immunological characteristics of patients with asthma or COPD. In vitro models of asthma and COPD using isolated human airways can provide information that is directly translatable into humans with obstructive lung diseases. Regardless of the technique used to investigate drugs for the treatment of chronic obstructive respiratory disorders (i.e., isolated organ bath systems, videomicroscopy and wire myography), the most limiting factors to produce high-quality and repeatable data remain closely tied to the manual skills of the researcher conducting experiments and the availability of suitable tissue.
Collapse
Affiliation(s)
- Luigino Calzetta
- Department of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Clive Page
- Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, London, UK
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
8
|
Palacionyte J, Januskevicius A, Vasyle E, Rimkunas A, Bajoriuniene I, Vitkauskiene A, Miliauskas S, Malakauskas K. Novel Serum Biomarkers for Patients with Allergic Asthma Phenotype. Biomedicines 2024; 12:232. [PMID: 38275403 PMCID: PMC10813071 DOI: 10.3390/biomedicines12010232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
In distinguishing the allergic asthma (AA) phenotype, it has been identified that specific biomarkers could assist; however, none of them are considered ideal. This study aimed to analyze three groups of biologically active substances in the serum. Twenty steroid-free AA patients, sensitized to Dermatophagoides pteronyssinus, and sixteen healthy subjects (HSs) were enrolled in this study. Blood samples were collected from all patients. Additionally, all AA patients underwent a bronchial allergen challenge (BAC) with Dermatophagoides pteronyssinus, all of which were positive, and blood samples were collected again 24 h later. The concentrations of ten biologically active substances were measured in the serum samples, using enzyme-linked immunosorbent assay (ELISA) and the Luminex® 100/200™ System technology for bead-based multiplex and singleplex immunoassays. Descriptive and analytical statistical methods were used. A p-value of 0.05 or lower was considered statistically significant. The soluble interleukin 5 receptor subunit alpha (sIL-5Rα) and thioredoxin 1 (TRX1) concentrations were significantly increased, whereas those of tyrosine-protein kinase Met (MET), pentraxin 3 (PTX3), and I C-telopeptide of type I collagen (ICTP) were decreased in the AA group compared with the HS group. A significant positive correlation was noted for sIL-5Rα with fractional exhaled nitric oxide (FeNO), blood eosinophil (EOS) count, and total immunoglobulin E (IgE) levels, and a negative correlation was noted with forced expiratory volume in 1 s (FEV1). Moreover, PTX3 showed negative correlations with blood EOS count and total IgE levels, whereas ICTP exhibited a negative correlation with the blood EOS count. In conclusion, this study demonstrated that the serum concentrations of MET, PTX3, TRX1, ICTP, and particularly sIL-5Rα could potentially serve as biomarkers of the AA phenotype.
Collapse
Affiliation(s)
- Jolita Palacionyte
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (S.M.); (K.M.)
| | - Andrius Januskevicius
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (E.V.); (A.R.)
| | - Egle Vasyle
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (E.V.); (A.R.)
| | - Airidas Rimkunas
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (E.V.); (A.R.)
| | - Ieva Bajoriuniene
- Department of Immunology and Allergology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania;
| | - Astra Vitkauskiene
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania;
| | - Skaidrius Miliauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (S.M.); (K.M.)
| | - Kestutis Malakauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (S.M.); (K.M.)
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania; (A.J.); (E.V.); (A.R.)
| |
Collapse
|
9
|
Jeon T, Luther DC, Goswami R, Bell C, Nagaraj H, Anil Cicek Y, Huang R, Mas-Rosario JA, Elia JL, Im J, Lee YW, Liu Y, Scaletti F, Farkas ME, Mager J, Rotello VM. Engineered Polymer-siRNA Polyplexes Provide Effective Treatment of Lung Inflammation. ACS NANO 2023; 17:4315-4326. [PMID: 36802503 PMCID: PMC10627429 DOI: 10.1021/acsnano.2c08690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Uncontrolled inflammation is responsible for acute and chronic diseases in the lung. Regulating expression of pro-inflammatory genes in pulmonary tissue using small interfering RNA (siRNA) is a promising approach to combatting respiratory diseases. However, siRNA therapeutics are generally hindered at the cellular level by endosomal entrapment of delivered cargo and at the organismal level by inefficient localization in pulmonary tissue. Here we report efficient anti-inflammatory activity in vitro and in vivo using polyplexes of siRNA and an engineered cationic polymer (PONI-Guan). PONI-Guan/siRNA polyplexes efficiently deliver siRNA cargo to the cytosol for highly efficient gene knockdown. Significantly, these polyplexes exhibit inherent targeting to inflamed lung tissue following intravenous administration in vivo. This strategy achieved effective (>70%) knockdown of gene expression in vitro and efficient (>80%) silencing of TNF-α expression in lipopolysaccharide (LPS)-challenged mice using a low (0.28 mg/kg) siRNA dosage.
Collapse
Affiliation(s)
- Taewon Jeon
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - David C. Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Charlotte Bell
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Javier A. Mas-Rosario
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
| | - James L. Elia
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Jungkyun Im
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
- Department of Chemical Engineering, and Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, 22 Soonchunhyangro, Asan, 31538, Republic of Korea
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Yuanchang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Federica Scaletti
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Michelle E. Farkas
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 661 N Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent M. Rotello
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, Massachusetts, 01003, USA
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
10
|
Sim S, Choi Y, Park HS. Immunologic Basis of Type 2 Biologics for Severe Asthma. Immune Netw 2022; 22:e45. [PMID: 36627938 PMCID: PMC9807964 DOI: 10.4110/in.2022.22.e45] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 12/30/2022] Open
Abstract
Asthma is a chronic airway inflammatory disease characterized by reversible airway obstruction and airway hyperreactivity to various environmental stimuli, leading to recurrent cough, dyspnea, and wheezing episodes. Regarding inflammatory mechanisms, type 2/eosinophilic inflammation along with activated mast cells is the major one; however, diverse mechanisms, including structural cells-derived and non-type 2/neutrophilic inflammations are involved, presenting heterogenous phenotypes. Although most asthmatic patients could be properly controlled by the guided treatment, patients with severe asthma (SA; classified as a treatment-refractory group) suffer from uncontrolled symptoms with frequent asthma exacerbations even on regular anti-inflammatory medications, raising needs for additional controllers, including biologics that target specific molecules found in asthmatic airway, and achieving the precision medicine for asthma. This review summarizes the immunologic basis of airway inflammatory mechanisms and current biologics for SA in order to address unmet needs for future targets.
Collapse
Affiliation(s)
- Soyoon Sim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Korea
| | - Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Korea
| |
Collapse
|
11
|
Kawabata H, Ohishi T, Suzuki H, Asano T, Kawada M, Suzuki H, Kaneko MK, Kato Y. A Defucosylated Mouse Anti-CD10 Monoclonal Antibody (31-mG 2a-f) Exerts Antitumor Activity in a Mouse Xenograft Model of Renal Cell Cancers. Monoclon Antib Immunodiagn Immunother 2022; 41:320-327. [PMID: 35483055 DOI: 10.1089/mab.2021.0049] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CD10 is a cell surface metalloendopeptidase that cleaves and degrades many secreted physiologically active peptides by its enzymatic activity. Although CD10 expression has been found in various types of cells, its expression is increased in several cancers, including renal cancer. In this study, the antitumor activity of a novel anti-human CD10 monoclonal antibody (mAb) was investigated. A defucosylated mouse IgG2a version of C10Mab-31 (31-mG2a-f) was created from an anti-CD10 mAb, C10Mab-31 (IgG1, kappa). Both C10Mab-31 and 31-mG2a-f specifically reacted with endogenous CD10 in renal cancer cells, VMRC-RCW, with the dissociation constant (KD) values of 6.3 × 10-9 M and 1.1 × 10-9 M, respectively, indicating high binding affinity. To further examine the anti-CD10 mAb-mediated effector functions, the antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) were examined. The 31-mG2a-f significantly exhibited ADCC and CDC against VMRC-RCW cells in vitro. Furthermore, 31-mG2a-f exhibited antitumor activities in mouse xenografts of VMRC-RCW cells. These results suggest that 31-mG2a-f exerts antitumor activities against CD10-expressing renal cancers and could be a valuable therapeutic candidate for treating them.
Collapse
Affiliation(s)
- Hiroki Kawabata
- Research and Development Center, Fuso Pharmaceutical Industries, Ltd., Osaka, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu-shi, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu-shi, Japan
| | - Hiroyoshi Suzuki
- Department of Pathology and Laboratory Medicine, Sendai Medical Center, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
12
|
Bieber T, Paller AS, Kabashima K, Feely M, Rueda MJ, Ross Terres JA, Wollenberg A. Atopic dermatitis: pathomechanisms and lessons learned from novel systemic therapeutic options. J Eur Acad Dermatol Venereol 2022; 36:1432-1449. [PMID: 35575442 DOI: 10.1111/jdv.18225] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/13/2022] [Indexed: 12/01/2022]
Abstract
Atopic dermatitis (AD) is a chronic, heterogenous, inflammatory skin disorder associated with a high skin-related health burden, typically starting in childhood and often persisting into adulthood. AD is characterized by a wide range of clinical phenotypes, reflecting multiple underlying pathophysiological mechanisms and interactions between genetics, immune system dysregulation, and environmental factors. In this review, we describe the diverse cellular and molecular mechanisms involved in AD, including the critical role of T cell-driven inflammation, primarily via T helper (Th) 2- and Th17-derived cytokines, many of which are mediated by the Janus kinase (JAK) signaling pathway. These local inflammatory processes interact with sensory neuronal pathways, contributing to the clinical manifestations of AD, including itch, pain, and sleep disturbance. The recent elucidation of the molecular pathways involved in AD has allowed treatment strategies to evolve from broad-acting systemic immunosuppressive therapies to more targeted agents, including JAK inhibitors and cytokine-specific biologic agents. Evidence from the clinical development of these targeted therapies has reinforced and expanded our understanding of the pathophysiological mechanisms underlying AD and holds promise for individualized treatment strategies tailored to specific AD subtypes.
Collapse
Affiliation(s)
- T Bieber
- Department of Dermatology and Allergy, University Medical Center, Bonn, Germany.,Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - A S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - K Kabashima
- Department Dermatology, Kyoto University School of Medicine, Kyoto, Japan
| | - M Feely
- Eli Lilly and Company, Indianapolis, IN, USA.,Department of Dermatology, Mount Sinai Hospital, New York, NY, USA
| | - M J Rueda
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - A Wollenberg
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximillian University, Munich, Germany.,Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Department of Dermatology, Brussels, Belgium
| |
Collapse
|
13
|
Lan SH, Lai CC, Chang SP, Hsu CC, Chen CH, Wang YH, Huang YL, Wang CY, Lin YS. Efficacy and safety of anti-interleukin-5 therapy in patients with chronic obstructive pulmonary disease: A meta-analysis of randomized, controlled trials. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:26-35. [PMID: 33349601 DOI: 10.1016/j.jmii.2020.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/18/2020] [Accepted: 11/06/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Anti-interleukin-5 (IL-5) therapy has been proposed as a novel treatment option for patients with chronic obstructive pulmonary disease (COPD). However, its efficacy for preventing COPD exacerbation remains unclear. METHODS A literature review was conducted to August 26th 2019. Only randomized controlled trials (RCTs) that investigated the clinical efficacy and adverse effects of anti-IL-5 therapy were included in the meta-analysis. The primary outcome was the risk of COPD exacerbation. RESULTS A total of 3 articles containing 5 RCTs were included in the study. Overall, 2837 and 1442 patients received anti-IL-5 therapy (mepolizumab, n = 865; benralizumab, n = 1972) and placebo, respectively. In the pooled analysis, anti-IL-5 therapy was associated with a lower risk of COPD exacerbation compared with the placebo (rate ratio, 0.92; 95% CI, 0.86-0.97, I2 = 0%). In addition, no significant differences in the changes in SGRQ scores and FEV1 from baseline were found between the anti-IL-5 therapy and placebo (SGRQ, mean difference, -0.86, 95% CI, -1.92 - 0.19, I2 = 0%; FEV1, mean difference, 0.01, 95% CI, -0.01 - 0.03, I2 = 0%). Anti-IL-5 therapy had a similar risk of any adverse event (risk ratio, 1.02; 95% CI, 0.99-1.05), an event leading to treatment discontinuation (risk ratio, 1.04; 95% CI, 0.72-1.48) and any serious adverse events (risk ratio, 0.93; 95% CI, 0.85-1.01) when compared with the placebo. CONCLUSION Anti-IL-5 therapy was associated with a lower rate of COPD exacerbation compared with placebo. In addition, anti-IL-5 therapy was well tolerated for COPD patients.
Collapse
Affiliation(s)
| | - Chih-Cheng Lai
- Department of Internal Medicine, Kaohsiung Veterans General Hospital, Tainan Branch, Tainan, Taiwan
| | | | - Chun-Chun Hsu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Hsin Chen
- Department of Internal Medicine, Cardinal Tien Hospital and School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Ya-Hui Wang
- Medical Research Center, Cardinal Tien Hospital and School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yueh Lan Huang
- Department of Internal Medicine, Cardinal Tien Hospital and School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - Cheng-Yi Wang
- Department of Internal Medicine, Cardinal Tien Hospital and School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - You-Shuei Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
14
|
Shi WW, Shi C, Wang TY, Li YL, Zhou YK, Zhang XH, Bierer D, Zheng JS, Liu L. Total Chemical Synthesis of Correctly Folded Disulfide-Rich Proteins Using a Removable O-Linked β- N-Acetylglucosamine Strategy. J Am Chem Soc 2022; 144:349-357. [PMID: 34978456 DOI: 10.1021/jacs.1c10091] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Disulfide-rich proteins are useful as drugs or tool molecules in biomedical studies, but their synthesis is complicated by the difficulties associated with their folding. Here, we describe a removable glycosylation modification (RGM) strategy that expedites the chemical synthesis of correctly folded proteins with multiple or even interchain disulfide bonds. Our strategy comprises the introduction of simple O-linked β-N-acetylglucosamine (O-GlcNAc) groups at the Ser/Thr sites that effectively improve the folding of disulfide-rich proteins by stabilization of their folding intermediates. After folding, the O-GlcNAc groups can be efficiently removed using O-GlcNAcase (OGA) to afford the correctly folded proteins. Using this strategy, we completed the synthesis of correctly folded hepcidin, an iron-regulating hormone bearing four pairs of disulfide-bonds, and the first total synthesis of correctly folded interleukin-5 (IL-5), a 26 kDa homodimer cytokine responsible for eosinophil growth and differentiation.
Collapse
Affiliation(s)
- Wei-Wei Shi
- Tsinghua-Peking Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, State Key Laboratory of Chemical Oncogenomics (Shenzhen), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | | | - Tong-Yue Wang
- Tsinghua-Peking Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, State Key Laboratory of Chemical Oncogenomics (Shenzhen), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yu-Lei Li
- Tsinghua-Peking Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, State Key Laboratory of Chemical Oncogenomics (Shenzhen), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | | | | | - Donald Bierer
- Bayer AG, Department of Medicinal Chemistry, Aprather Weg 18A, 42096 Wuppertal, Germany
| | | | - Lei Liu
- Tsinghua-Peking Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, State Key Laboratory of Chemical Oncogenomics (Shenzhen), Department of Chemistry, Tsinghua University, Beijing 100084, China
| |
Collapse
|
15
|
Abstract
Recent therapeutic advances in the management of asthma have underscored the importance of eosinophilia and the role of pro-eosinophilic mediators such as IL-5 in asthma. Given that a subset of patients with COPD may display peripheral eosinophilia similar to what is observed in asthma, a number of recent studies have implied that eosinophilic COPD is a distinct entity. This review will seek to contrast the mechanisms of eosinophilia in asthma and COPD, the implications of eosinophilia for disease outcome, and review current data regarding the utility of peripheral blood eosinophilia in the management of COPD patients.
Collapse
|
16
|
Xie Z, Xiang C, Li X, Fan C, Chen T, Liu M, Ma Y, Bai F, Tang W, Hu Y. Discovery of Potent Antiallergic Agents Based on an o-Aminopyridinyl Alkynyl Scaffold. J Med Chem 2021; 64:13588-13603. [PMID: 34476950 DOI: 10.1021/acs.jmedchem.1c00976] [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: 11/29/2022]
Abstract
Effective therapeutic agents are highly desired for immune-mediated allergic diseases. Herein, we report the design, synthesis, and structure-activity relationship of an o-aminopyridinyl alkyne series as novel orally bioavailable antiallergic agents, which was identified through phenotypic screening. Compound optimization yielded a highly potent compound 36, which effectively suppressed mast cell degranulation in a dose-dependent manner (IC50, 2.54 nM for RBL-2H3 cells; 48.28 nM for peritoneal mast cells (PMCs)) with a good therapeutic index. It also regulated the activation of FcεRI-mediated downstream signaling proteins in IgE/Ag-stimulated RBL-2H3 cells. In addition, 36 exhibited excellent in vivo pharmacokinetic properties and antiallergic efficacy in both passive systemic anaphylaxis (PSA) and house dust mite (HDM)-induced murine models of pulmonary allergic inflammation. Furthermore, preliminary analysis of the kinases profile identified Src-family kinases as potential targets for 36. Compound 36 may serve as a new valuable lead compound for future antiallergic drug discovery.
Collapse
Affiliation(s)
- Zhicheng Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Caigui Xiang
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Xin Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Chen Fan
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Taiwen Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Moting Liu
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yanjie Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Fang Bai
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wei Tang
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Youhong Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
17
|
Abstract
RATIONALE Blood eosinophil count predicts response to inhaled corticosteroids and specific biologic therapies in selected asthma patients. Despite this important role, fundamental aspects of eosinophil behavior in asthma have not been studied. Objectives To investigate the behavior of blood eosinophils in a population comparing their distribution with the general population and studying their intra-individual variability over time in relation to hospital episodes (emergency department visits and hospitalizations) in clinical practice. METHODS The distribution and variability of 35,703 eosinophil determinations in 10,059 stable asthma patients were investigated in the Majorca Real-Life Investigation in COPD and Asthma cohort (MAJORICA). Eosinophil distribution in the asthma population was compared with a control sample from the general population of 8,557 individuals. Eosinophil variability and hospital episodes were analyzed using correlations, ROC curves and multiple regression analysis. We defined the Eosinophil Variability Index (EVI) as (Eosmax-Eosmin/Eosmax) x 100%. The findings of the asthma population were re-tested in an external well-characterized asthma cohort. RESULTS The eosinophil count values and variability were higher in the asthma population than in the general population (p-value<0.001). Variability data showed a better association with hospital episodes than the counting values. An EVI≥50% was more strongly associated with hospital episodes than any of the absolute counting values. These results were validated in the external cohort. CONCLUSION The eosinophil variability in asthma patients better identifies the risk of any hospital episode than the absolute counting values currently used to target specific treatments.
Collapse
|
18
|
Tateyama N, Nanamiya R, Ohishi T, Takei J, Nakamura T, Yanaka M, Hosono H, Saito M, Asano T, Tanaka T, Sano M, Kawada M, Kaneko MK, Kato Y. Defucosylated Anti-Epidermal Growth Factor Receptor Monoclonal Antibody 134-mG 2a-f Exerts Antitumor Activities in Mouse Xenograft Models of Dog Epidermal Growth Factor Receptor-Overexpressed Cells. Monoclon Antib Immunodiagn Immunother 2021; 40:177-183. [PMID: 34424762 DOI: 10.1089/mab.2021.0022] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a type I transmembrane protein, which is a member of the human epidermal growth factor receptor (HER) family of receptor tyrosine kinases. EGFR is a crucial mediator of cell growth and differentiation and forms homodimers or heterodimers with other HER family members to activate downstream signaling cascades. We previously established an anti-human EGFR (hEGFR) monoclonal antibody (mAb), clone EMab-134 (mouse IgG1), by immunizing mice with the ectodomain of hEGFR. In this study, the subclass of EMab-134 was converted from IgG1 to IgG2a (134-mG2a) and further defucosylated (134-mG2a-f) to facilitate antibody-dependent cellular cytotoxicity (ADCC). Although 134-mG2a-f was developed against hEGFR, it was shown to cross-react with dog EGFR (dEGFR) using flow cytometry. The dissociation constant (KD) of 134-mG2a-f against dEGFR-overexpressed CHO-K1 (CHO/dEGFR) cells was determined by flow cytometry to be 3.3 × 10-9 M, indicating that 134-mG2a-f possesses a high binding affinity to dEGFR. Analysis in vitro revealed that 134-mG2a-f contributed to high levels of ADCC and complement-dependent cytotoxicity (CDC) in experiments targeting CHO/dEGFR cells. Furthermore, the in vivo administration of 134-mG2a-f significantly inhibited the development of CHO/dEGFR in comparison with the results observed in response to control mouse IgG. Taken together, the findings of this study demonstrate that 134-mG2a-f could be useful as part of a therapeutic regimen for dEGFR-expressing canine cancers.
Collapse
Affiliation(s)
- Nami Tateyama
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ren Nanamiya
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu-shi, Japan
| | - Junko Takei
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideki Hosono
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masaki Saito
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masato Sano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu-shi, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
19
|
Huang BC, Lu YC, Liao JM, Liu HJ, Hong ST, Hsieh YC, Chuang CH, Chen HJ, Liao TY, Ho KW, Wang YT, Cheng TL. Development of a structure-based computational simulation to optimize the blocking efficacy of pro-antibodies. Chem Sci 2021; 12:9759-9769. [PMID: 34349949 PMCID: PMC8293997 DOI: 10.1039/d1sc01748a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/20/2021] [Accepted: 06/13/2021] [Indexed: 11/21/2022] Open
Abstract
The on-target toxicity of monoclonal antibodies (Abs) is mainly due to the fact that Abs cannot distinguish target antigens (Ags) expressed in disease regions from those in normal tissues during systemic administration. In order to overcome this issue, we “copied” an autologous Ab hinge as an “Ab lock” and “pasted” it on the binding site of the Ab by connecting a protease substrate and linker in between to generate a pro-Ab, which can be specifically activated in the disease region to enhance Ab selectivity and reduce side effects. Previously, we reported that 70% of pro-Abs can achieve more than 100-fold blocking ability compared to the parental Abs. However, 30% of pro-Abs do not have such efficient blocking ability. This is because the same Ab lock linker cannot be applied to every Ab due to the differences in the complementarity-determining region (CDR) loops. Here we designed a method which uses structure-based computational simulation (MSCS) to optimize the blocking ability of the Ab lock for all Ab drugs. MSCS can precisely adjust the amino acid composition of the linker between the Ab lock and Ab drug with the assistance of molecular simulation. We selected αPD-1, αIL-1β, αCTLA-4 and αTNFα Ab as models and attached the Ab lock with various linkers (L1 to L7) to form pro-Abs by MSCS, respectively. The resulting cover rates of the Ab lock with various linkers compared to the Ab drug were in the range 28.33–42.33%. The recombinant pro-Abs were generated by MSCS prediction in order to verify the application of molecular simulation for pro-Ab development. The binding kinetics effective concentrations (EC-50) for αPD-1 (200-250-fold), αIL-1β (152-186-fold), αCTLA-4 (68-150-fold) and αTNFα Ab (20-123-fold) were presented as the blocking ability of pro-Ab compared to the Ab drug. Further, there was a positive correlation between cover rate and blocking ability of all pro-Ab candidates. The results suggested that MSCS was able to predict the Ab lock linker most suitable for application to αPD-1, αIL-1β, αCTLA-4 and αTNFα Ab to form pro-Abs efficiently. The success of MSCS in optimizing the pro-Ab can aid the development of next-generation pro-Ab drugs to significantly improve Ab-based therapies and thus patients' quality of life. The pro-Ab blocks the Ag binding site using an Ab lock. We designed a method which uses structure-based computational simulation (MSCS) to predict the cover rate of Ab locks with various linkers and select the suitable linker for each Ab.![]()
Collapse
Affiliation(s)
- Bo-Cheng Huang
- Institute of Biomedical Sciences, National Sun Yat-Sen University Kaohsiung Taiwan
| | - Yun-Chi Lu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University Kaohsiung Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University Kaohsiung Taiwan
| | - Jun-Min Liao
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University Kaohsiung Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University Kaohsiung Taiwan
| | - Hui-Ju Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University Kaohsiung Taiwan
| | - Shih-Ting Hong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University Kaohsiung Taiwan
| | - Yuan-Chin Hsieh
- School of Medicine for International Students, I-Shou University Kaohsiung Taiwan
| | - Chih-Hung Chuang
- Drug Development and Value Creation Research Center, Kaohsiung Medical University Kaohsiung Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University Kaohsiung Taiwan
| | - Huei-Jen Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University Kaohsiung Taiwan
| | - Tzu-Yi Liao
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University Kaohsiung Taiwan
| | - Kai-Wen Ho
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University Kaohsiung Taiwan
| | - Yeng-Tseng Wang
- Department of Biochemistry, Kaohsiung Medical University Kaohsiung Taiwan
| | - Tian-Lu Cheng
- Institute of Biomedical Sciences, National Sun Yat-Sen University Kaohsiung Taiwan .,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University Kaohsiung Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University Kaohsiung Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital Kaohsiung Taiwan
| |
Collapse
|
20
|
Fieldes M, Bourguignon C, Assou S, Nasri A, Fort A, Vachier I, De Vos J, Ahmed E, Bourdin A. Targeted therapy in eosinophilic chronic obstructive pulmonary disease. ERJ Open Res 2021; 7:00437-2020. [PMID: 33855061 PMCID: PMC8039900 DOI: 10.1183/23120541.00437-2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common and preventable airway disease causing significant worldwide mortality and morbidity. Lifetime exposure to tobacco smoking and environmental particles are the two major risk factors. Over recent decades, COPD has become a growing public health problem with an increase in incidence. COPD is defined by airflow limitation due to airway inflammation and small airway remodelling coupled to parenchymal lung destruction. Most patients exhibit neutrophil-predominant airway inflammation combined with an increase in macrophages and CD8+ T-cells. Asthma is a heterogeneous chronic inflammatory airway disease. The most studied subtype is type 2 (T2) high eosinophilic asthma, for which there are an increasing number of biologic agents developed. However, both asthma and COPD are complex and share common pathophysiological mechanisms. They are known as overlapping syndromes as approximately 40% of patients with COPD present an eosinophilic airway inflammation. Several studies suggest a putative role of eosinophilia in lung function decline and COPD exacerbation. Recently, pharmacological agents targeting eosinophilic traits in uncontrolled eosinophilic asthma, especially monoclonal antibodies directed against interleukins (IL-5, IL-4, IL-13) or their receptors, have shown promising results. This review examines data on the rationale for such biological agents and assesses efficacy in T2-endotype COPD patients. Patients with severe COPD and eosinophilic inflammation experience uncontrolled symptoms despite optimal pharmaceutical treatment. The development of new biomarkers is needed for better phenotyping of patients to propose innovative targeted therapy.https://bit.ly/2KzWuNO
Collapse
Affiliation(s)
- Mathieu Fieldes
- IRMB, INSERM, Montpellier University Hospital, Montpellier, France
| | | | - Said Assou
- IRMB, INSERM, Montpellier University Hospital, Montpellier, France
| | - Amel Nasri
- IRMB, INSERM, Montpellier University Hospital, Montpellier, France
| | - Aurélie Fort
- Dept of Respiratory Diseases, Montpellier University Hospital, INSERM, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, Montpellier, France
| | - Isabelle Vachier
- Dept of Respiratory Diseases, Montpellier University Hospital, INSERM, Montpellier, France
| | - John De Vos
- IRMB, INSERM, Montpellier University Hospital, Montpellier, France.,Dept of Cell and Tissue Engineering, Montpellier University Hospital, Montpellier, France
| | - Engi Ahmed
- Dept of Respiratory Diseases, Montpellier University Hospital, INSERM, Montpellier, France
| | - Arnaud Bourdin
- Dept of Respiratory Diseases, Montpellier University Hospital, INSERM, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, Montpellier, France
| |
Collapse
|
21
|
Lucendo AJ, López-Sánchez P. Targeted Therapies for Eosinophilic Gastrointestinal Disorders. BioDrugs 2021; 34:477-493. [PMID: 32472465 DOI: 10.1007/s40259-020-00427-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The growing recognition of eosinophilic gastrointestinal disorders has revealed the limitations of current treatment (mainly based on dietary modification and corticosteroids), and include refractoriness, high recurrence rates, and the need for long-term therapy. Research efforts, mainly in eosinophilic esophagitis (EoE), have unveiled essential pathophysiological mechanisms leading to these disorders, which bear some similarities to those of atopic manifestations and are shared by eosinophilic gastroenteritis (EGE) and eosinophilic colitis (EC). Novel targeted therapies, some imported from bronchial asthma and atopic dermatitis, are currently being assessed in EoE. The most promising are monoclonal antibodies, including those targeting interleukin (IL)-13 (cendakimab) and IL-4 (dupilumab), with phase 3 trials currently ongoing. The potential of anti-integrin therapy (vedolizumab) and Siglec-8 blockers (antolimab) in EGE are also promising. Non-biological therapies for eosinophilic gut disorders, which include preventing the activation of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and chemoattractant receptor expressed on T helper 2 cells (CRTH2) signaling pathways, and other potential targets that deserve investigation in eosinophilic gut disorders, are reviewed.
Collapse
Affiliation(s)
- Alfredo J Lucendo
- Department of Gastroenterology, Hospital General de Tomelloso, Vereda de Socuéllamos, s/n.,, 13700, Tomelloso, Ciudad Real, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain. .,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain.
| | | |
Collapse
|
22
|
Abstract
The global COVID-19 pandemic has brought respiratory disease to the forefront of public health, but asthma prevalence has been rising globally for decades. Asthma is mediated by errant immune activation and airway remodeling, but the influences of environment, nutrition, and comorbidities (e.g., asthma-chronic obstructive pulmonary disorder-overlap [ACO]) are still poorly understood. Even as a new generation of biologic-based treatments offer better airway control and reductions in mortality, a lack of prophylactic treatments and mechanistic understanding complicates efforts to prevent pathogenesis. This review will explicate and synthesize current knowledge on the effect of ACO and biologics (omalizumab, mepolizumab, reslizumab, benralizumab, and dupilumab) on pathogenesis, treatment, and prognosis.
Collapse
|
23
|
Komalla V, Mehta M, Achi F, Dua K, Haghi M. The Potential for Phospholipids in the Treatment of Airway Inflammation: An Unexplored Solution. Curr Mol Pharmacol 2021; 14:333-349. [PMID: 33557743 DOI: 10.2174/1874467214666210208114439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 11/22/2022]
Abstract
Asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF) are major inflammatory respiratory diseases. Current mainstay therapy for asthma, and chronic obstructive pulmonary disease are corticosteroids, which have well-established side effect profiles. Phospholipids (PLs) are ubiquitous, diverse compounds with varying functions such as their structural role incell membrane, energy storage, and cell signaling.Recent advances in understanding PLs role as inflammatory mediators in the body as well as their widespread long-standing use as carrier molecules in drug delivery demonstrate the potential application of phospholipids in modulating inflammatory conditions. This review briefly explains the main mechanisms of inflammation in chronic respiratory diseases, currentanti-inflammatory treatments and areas of unmet need. The structural features, roles of endogenous and exogenous phospholipids, including their use as pharmaceutical excipients are reviewed. Current research on the immunomodulatory properties of PLs and their potentialapplication in inflammatory diseasesis the major section of this review. Considering the roles of PLs as inflammatory mediators and their safety profile established in pharmaceutical formulations, these small molecules demonstrate great potential as candidates in respiratory inflammation. Future studies need to focus on the immunomodulatory properties and the underlying mechanisms of phospholipids in respiratory inflammatory diseases.
Collapse
Affiliation(s)
- Varsha Komalla
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| | - Meenu Mehta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| | - Fatima Achi
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| | - Mehra Haghi
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| |
Collapse
|
24
|
Huang XP, Qin CY, Gao YM. miR-135a inhibits airway inflammatory response in asthmatic mice via regulating JAK/STAT signaling pathway. ACTA ACUST UNITED AC 2021; 54:e10023. [PMID: 33470387 PMCID: PMC7812909 DOI: 10.1590/1414-431x202010023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/10/2020] [Indexed: 11/24/2022]
Abstract
The objective of this study was to investigate the inhibitory effect of miR-135a in regulating JAK/STAT signaling pathway on airway inflammation in asthmatic mice. An asthma model was established by sensitization and stimulation with ovalbumin (OVA), and the corresponding drug intervention was given from the day of stimulation by means of nasal drops. Airway hyperresponsiveness was tested. The content of miR-135a in the lung tissue of mice was detected by RT-PCR. The pathological changes of lung tissue were evaluated by HE staining. Tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-5, and eotaxin in bronchoalveolar lavage fluid (BALF) and lung tissue were detected by ELISA and immunohistochemistry, respectively. The expression of JAK/STAT signaling pathway-related protein in lung tissue was detected by western blot. To further validate the effect of miR-135a overexpression on the JAK/STAT signaling pathway, pathway activators and inhibitors were added. Compared with the OVA group, the airway hyperresponsiveness of the mice was significantly decreased after treatment with the miR-135a agonist. The expression of miR-135a was significantly increased in the lung tissue and the pathological changes of the lung tissue were alleviated. The contents of TNF-α, IL-6, IL-5, and eotaxin in BALF and lung tissues were decreased. The expression of JAK/STAT signaling pathway-related proteins p-JAK3/JAK3, p-STAT1/STAT1, and p-STAT3/STAT3 were significantly reduced in lung tissue (P<0.05). Addition of JAK inhibitor AG490 reduced airway inflammation in asthmatic mice. miR-135a agonists inhibit airway inflammation in asthmatic mice by regulating the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Xue-Peng Huang
- Respiratory Department, People's Hospital of Rizhao Lanshan, Rizhao, Shandong, China
| | - Cheng-Yu Qin
- Respiratory Department, People's Hospital of Rizhao Lanshan, Rizhao, Shandong, China
| | - Yue-Ming Gao
- Respiratory Department, People's Hospital of Rizhao Lanshan, Rizhao, Shandong, China
| |
Collapse
|
25
|
Donovan T, Milan SJ, Wang R, Banchoff E, Bradley P, Crossingham I. Anti-IL-5 therapies for chronic obstructive pulmonary disease. Cochrane Database Syst Rev 2020; 12:CD013432. [PMID: 33295032 PMCID: PMC8106745 DOI: 10.1002/14651858.cd013432.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Exacerbations of chronic obstructive pulmonary disease (COPD) are a major cause of hospital admissions, disease-related morbidity and mortality. COPD is a heterogeneous disease with distinct inflammatory phenotypes, including eosinophilia, which may drive acute exacerbations in a subgroup of patients. Monoclonal antibodies targeting interleukin 5 (IL-5) or its receptor (IL-5R) have a role in the care of people with severe eosinophilic asthma, and may similarly provide therapeutic benefit for people with COPD of eosinophilic phenotype. OBJECTIVES To assess the efficacy and safety of monoclonal antibody therapies targeting IL-5 signalling (anti-IL-5 or anti-IL-5Rα) compared with placebo in the treatment of adults with COPD. SEARCH METHODS We searched the Cochrane Airways Trials Register, CENTRAL, MEDLINE, Embase, clinical trials registries, manufacturers' websites, and reference lists of included studies. Our most recent search was 23 September 2020. SELECTION CRITERIA We included randomised controlled trials comparing anti-IL-5 therapy with placebo in adults with COPD. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and analysed outcomes using a random-effects model.The primary outcomes were exacerbations requiring antibiotics or oral steroids, hospitalisations due to exacerbation of COPD, serious adverse events, and quality of life. We used standard methods expected by Cochrane. We used the GRADE approach to assess the certainty of the evidence. MAIN RESULTS Six studies involving a total of 5542 participants met our inclusion criteria. Three studies used mepolizumab (1530 participants), and three used benralizumab (4012 participants). The studies were on people with COPD, which was similarly defined with a documented history of COPD for at least one year. We deemed the risk of bias to be generally low, with all studies contributing data of robust methodology. Mepolizumab 100 mg reduces the rate of moderate or severe exacerbations by 19% in those with an eosinophil count of at least 150/μL (rate ratio (RR) 0.81, 95% confidence interval (CI) 0.71 to 0.93; participants = 911; studies = 2, high-certainty evidence). When participants with lower eosinophils are included, mepolizumab 100 mg probably reduces the exacerbation rate by 8% (RR 0.92, 95% CI 0.82 to 1.03; participants = 1285; studies = 2, moderate-certainty evidence). Mepolizumab 300 mg probably reduces the rate of exacerbations by 14% in participants all of whom had raised eosinophils (RR 0.86, 95% CI 0.70 to 1.06; participants = 451; studies = 1, moderate-certainty evidence); the evidence was uncertain for a single small study of mepolizumab 750 mg. In participants with high eosinophils, mepolizumab probably reduces the rate of hospitalisation by 10% (100 mg, RR 0.90, 95% CI 0.65 to 1.24; participants = 911; studies = 2, moderate-certainty evidence) and 17% (300 mg, RR 0.83, 95% CI 0.51 to 1.35; participants = 451; studies = 1, moderate-certainty evidence). Mepolizumab 100 mg increases the time to first moderate or severe exacerbation compared to the placebo group, in people with the eosinophilic phenotype (hazard ratio (HR) 0.78, 95% CI 0.66 to 0.92; participants = 981; studies 2, high-certainty evidence). When participants with lower eosinophils were included this difference was smaller and less certain (HR 0.87, 95% CI 0.75 to 1.0; participants = 1285; studies 2, moderate-certainty evidence). Mepolizumab 300 mg probably increases the time to first moderate or severe exacerbation in participants who all had eosinophilic phenotype (HR 0.77, 95% CI 0.60 to 0.99; participants = 451; studies = 1, moderate-certainty evidence). Benralizumab 100 mg reduces the rate of severe exacerbations requiring hospitalisation in those with an eosinophil count of at least 220/μL (RR 0.63, 95% CI 0.49 to 0.81; participants = 1512; studies = 2, high-certainty evidence). Benralizumab 10 mg probably reduces the rate of severe exacerbations requiring hospitalisation in those with an eosinophil count of at least 220/μL (RR 0.68, 95% CI 0.49 to 0.94; participants = 765; studies = 1, moderate-certainty evidence). There was probably little or no difference between the intervention and placebo for quality of life measures. Where there were differences the mean difference fell below the pre-specified minimum clinically significant difference. Treatment with mepolizumab and benralizumab appeared to be safe. All pooled analyses showed that there was probably little or no difference in serious adverse events, adverse events, or side effects between the use of a monoclonal antibody therapy compared to placebo. AUTHORS' CONCLUSIONS We found that mepolizumab and benralizumab probably reduce the rate of moderate and severe exacerbations in the highly selected group of people who have both COPD and higher levels of blood eosinophils. This highlights the importance of disease phenotyping in COPD, and may play a role in the personalised treatment strategy in disease management. Further research is needed to elucidate the role of monoclonal antibodies in the management of COPD in clinical practice. In particular, it is not clear whether there is a threshold blood eosinophil level above which these drugs may be effective. Studies including cost effectiveness analysis may be beneficial given the high cost of these therapies, to support use if appropriate.
Collapse
Affiliation(s)
- Tim Donovan
- Medical and Sport Sciences, University of Cumbria, Lancaster, UK
| | | | - Ran Wang
- Department of Respiratory Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Patrick Bradley
- Department of Respiratory Medicine, North West Lung Centre, Wythenshawe Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | | |
Collapse
|
26
|
Shastri MD, Chong WC, Dua K, Peterson GM, Patel RP, Mahmood MQ, Tambuwala M, Chellappan DK, Hansbro NG, Shukla SD, Hansbro PM. Emerging concepts and directed therapeutics for the management of asthma: regulating the regulators. Inflammopharmacology 2020; 29:15-33. [PMID: 33152094 DOI: 10.1007/s10787-020-00770-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/17/2020] [Indexed: 12/19/2022]
Abstract
Asthma is a common, heterogeneous and serious disease, its prevalence has steadily risen in most parts of the world, and the condition is often inadequately controlled in many patients. Hence, there is a major need for new therapeutic approaches. Mild-to-moderate asthma is considered a T-helper cell type-2-mediated inflammatory disorder that develops due to abnormal immune responses to otherwise innocuous allergens. Prolonged exposure to allergens and persistent inflammation results in myofibroblast infiltration and airway remodelling with mucus hypersecretion, airway smooth muscle hypertrophy, and excess collagen deposition. The airways become hyper-responsive to provocation resulting in the characteristic wheezing and obstructed airflow experienced by patients. Extensive research has progressed the understanding of the underlying mechanisms and the development of new treatments for the management of asthma. Here, we review the basis of the disease, covering new areas such as the role of vascularisation and microRNAs, as well as associated potential therapeutic interventions utilising reports from animal and human studies. We also cover novel drug delivery strategies that are being developed to enhance therapeutic efficacy and patient compliance. Potential avenues to explore to improve the future of asthma management are highlighted.
Collapse
Affiliation(s)
- Madhur D Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia.,Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gregory M Peterson
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Rahul P Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Malik Q Mahmood
- Faculty of Health, School of Medicine, Deakin University, Melbourne, Australia
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Belfast, Northern Ireland, UK
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia. .,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia. .,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
27
|
Zanandrea R, Bonan CD, Campos MM. Zebrafish as a model for inflammation and drug discovery. Drug Discov Today 2020; 25:2201-2211. [PMID: 33035664 DOI: 10.1016/j.drudis.2020.09.036] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/17/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022]
Abstract
Zebrafish is a small teleost (bony) fish used in many areas of pharmacology and toxicology. This animal model has advantages for the discovery of anti-inflammatory drugs, such as the potential for real-time assessment of cell migration mechanisms. Additionally, zebrafish display a repertoire of inflammatory cells, mediators, and receptors that are similar to those in mammals, including humans. Inflammatory disease modeling in either larvae or adult zebrafish represents a promising tool for the screening of new anti-inflammatory compounds, contributing to our understanding of the mechanisms involved in chronic inflammatory conditions. In this review, we provide an overview of the characterization of inflammatory responses in zebrafish, emphasizing its relevance for drug discovery in this research area.
Collapse
Affiliation(s)
- Rodrigo Zanandrea
- Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Laboratório de Neuroquímica e Psicofarmacologia, Porto Alegre, RS, Brazil
| | - Carla D Bonan
- Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Laboratório de Neuroquímica e Psicofarmacologia, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Programa de Pós-Graduação em Biologia Celular e Molecular, Porto Alegre, RS, Brazil
| | - Maria M Campos
- Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Medicina, Programa de Pós-Graduação em Medicina e Ciências da Saúde, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Programa de Pós-Graduação em Biologia Celular e Molecular, Porto Alegre, RS, Brazil; Pontifícia Universidade Católica do Rio Grande do Sul, Escola de Ciências da Saúde e da Vida, Centro de Pesquisa em Toxicologia e Farmacologia, Porto Alegre, RS, Brazil.
| |
Collapse
|
28
|
Sevoflurane modulates AQPs (1,5) expression and endoplasmic reticulum stress in mice lung with allergic airway inflammation. Biosci Rep 2020; 39:221068. [PMID: 31710085 PMCID: PMC6879378 DOI: 10.1042/bsr20193282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/29/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023] Open
Abstract
Sevoflurane was found to show protective roles in mice with asthma, however, the mechanism of which needs further exploring. Aquaporins (AQPs) have been demonstrated to be involved in the pathogenesis of asthma, while endoplasmic reticulum stress has been reported to be related to many inflammatory diseases and involved in protein processing, including AQPs. The present study aimed to determine the role of sevoflurane in AQPs (AQP1,3,4,5) expression in mice with allergic airway inflammation and the probable mechanism. The increased number of inflammatory cells infiltrating the lung tissue, and the elevated levels of tumor necrosis factor-α (TNF-α) and interleukin (IL) 13 (IL-13) were all decreased after sevoflurane treatment (all P<0.05). Meanwhile, mRNA levels of AQP1 and AQP5 but not AQP3 and AQP4 were decreased in ovalbumin (OVA)-induced allergic mice lung. Both the decreased mRNA expression and protein levels of AQP1 and AQP5 in allergic lung tissues were reversed by sevoflurane treatment. Furthermore, we established that sevoflurane inhibited the OVA-induced protein increase in the endoplasmic reticulum (ER) stress markers BiP and C/EBP homologous protein (CHOP). Collectively, these findings suggested that sevoflurane modulated the expression and protein level of AOPs (AQP1, AQP5) as well as inhibited ER stress response in OVA-induced allergic airway inflammation of mice.
Collapse
|
29
|
Mohamed MME, Nicklin AD, Stover CM. The Value of Targeting Complement Components in Asthma. ACTA ACUST UNITED AC 2020; 56:medicina56080405. [PMID: 32806638 PMCID: PMC7466339 DOI: 10.3390/medicina56080405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 01/09/2023]
Abstract
Asthma is an important respiratory illness. Though pharmacological and biological treatment is well established and is staged according to endotypes and their responses to treatment, novel avenues are being explored. Our focus is complement. In this viewpoint, we evaluate the approach to target complement in this complex hypersensitivity reaction that develops chronicity and has a personal—as well as a societal—cost.
Collapse
|
30
|
Cong J, Gong J, Yang C, Xia Z, Zhang H. miR-22 Suppresses Tumor Invasion and Metastasis in Colorectal Cancer by Targeting NLRP3. Cancer Manag Res 2020; 12:5419-5429. [PMID: 32753959 PMCID: PMC7351629 DOI: 10.2147/cmar.s255125] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose This study aimed to investigate the effects of microRNA (miR)-22 on biological behaviors of colon cancer cells and to explore the relationship between miR-22 and NLRP3. Materials and Methods First, human colon cancer HCT116 cells were transfected with a miR-22 mimic, miR-22 inhibitor, control mimic, and control inhibitor, respectively. CCK8, colony formation, and transwell assays were performed to observe cell proliferation, migration, and invasion. Western blotting was used to analyze the expression of recombinant NLRP3 (NLR family, pyrin domain-containing protein 3) and epithelial–mesenchymal transformation (EMT)-related proteins. The target relationship between miR-22 and NLRP3 was verified by double luciferase report. Second, an NLRP3 inhibitor and NLRP3 mimic were transfected into HCT116 cells, and the biological behaviors and EMT-related proteins were again observed. Finally, a nude mouse xenograft model was constructed to verify the above results. Results In vitro, compared with the control group, administration of the miR-22 mimic significantly decreased proliferation, migration, and invasion of HCT116 cells, whereas the miR-22 inhibitor markedly increased their proliferation and invasion (p<0.05). Levels of NLRP3, interleukin-1β (IL-1β), matrix metalloproteinase-9 (MMP-9), MMP-2, N-cadherin, and vimentin were significantly reduced after miR-22 mimic transfection (p<0.05). Furthermore, silencing of NLRP3, a downstream gene of miR-22 in HCT116 cells, suppressed proliferation, migration, and invasion of HCT116 cells. However, overexpression of NLRP3 weakened the effects of the miR-22 mimic. In vivo, overexpression of miR-22 slowed the growth rate of tumors and reduced Ki-67 expression in tumor tissues compared with the model group (p<0.05). In tumor tissues, overexpression of miR-22 also decreased expression of NLRP3, IL-1β, MMP-9, MMP-2, N-cadherin, and vimentin compared with the model group (p<0.05). Overexpression of NLRP3 weakened the role of miR-22 overexpression in vivo. Conclusion miR-22 suppresses cell proliferation, migration, and invasion in colorectal cancer by targeting NLRP3.
Collapse
Affiliation(s)
- Jinchun Cong
- Department of Colorectal Tumor Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Jian Gong
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Chuanjia Yang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110016, People's Republic of China
| | - Zhixiu Xia
- Department of Colorectal Tumor Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Hong Zhang
- Department of Colorectal Tumor Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| |
Collapse
|
31
|
Govoni M, Bassi M, Vezzoli S, Lucci G, Emirova A, Nandeuil MA, Petruzzelli S, Jellema GL, Afolabi EK, Colgan B, Leaker B, Kornmann O, Beeh KM, Watz H, Singh D. Sputum and blood transcriptomics characterisation of the inhaled PDE4 inhibitor CHF6001 on top of triple therapy in patients with chronic bronchitis. Respir Res 2020; 21:72. [PMID: 32197620 PMCID: PMC7085203 DOI: 10.1186/s12931-020-1329-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background Although phosphodiesterase-4 (PDE4) inhibitors have been shown to reduce COPD exacerbation rate, their biological mechanism of action is not completely elucidated at the molecular level. We aimed to characterise the whole genome gene expression profile of the inhaled PDE4-inhibitor CHF6001 on top of triple therapy in sputum cells and whole blood of patients with COPD and chronic bronchitis. Methods Whole genome gene expression analysis was carried out by microarray in 54 patients before and after 32 days treatment with CHF6001 800 and 1600 μg and placebo twice daily (BID) in a randomised crossover study. Results CHF6001 had a strong effect in sputum, with 1471 and 2598 significantly differentially-expressed probe-sets relative to placebo (p-adjusted for False Discovery Rate < 0.05) with 800 and 1600 μg BID, respectively. Functional enrichment analysis showed significant modulation of key inflammatory pathways involved in cytokine activity, pathogen-associated-pattern-recognition activity, oxidative stress and vitamin D with associated inhibition of downstream inflammatory effectors. A large number of pro-inflammatory genes coding for cytokines and matrix-metalloproteinases were significantly differentially expressed for both doses; the majority (> 87%) were downregulated, including macrophage inflammatory protein-1-alpha and 1-beta, interleukin-27-beta, interleukin-12-beta, interleukin-32, tumour necrosis factor-alpha-induced-protein-8, ligand-superfamily-member-15, and matrix-metalloproteinases-7,12 and 14. The effect in blood was not significant. Conclusions Inhaled PDE4 inhibition by CHF6001 on top of triple therapy in patients with COPD and chronic bronchitis significantly modulated key inflammatory targets and pathways in the lung but not in blood. Mechanistically these findings support a targeted effect in the lung while minimising unwanted systemic class-effects. Trial registration ClinicalTrial.gov, EudraCT, 2015–005550-35. Registered 15 July 2016.
Collapse
Affiliation(s)
- Mirco Govoni
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy.
| | - Michele Bassi
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Stefano Vezzoli
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Germano Lucci
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Aida Emirova
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Marie Anna Nandeuil
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Stefano Petruzzelli
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | | | | | | | | | - Oliver Kornmann
- IKF Pneumologie Frankfurt, Clinical Research Centre Respiratory Diseases, Frankfurt, Germany
| | | | - Henrik Watz
- Pulmonary Research Institute at Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Dave Singh
- Medicines Evaluation Unit, The University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
32
|
Albertson TE, Chenoweth JA, Pearson SJ, Murin S. The pharmacological management of asthma-chronic obstructive pulmonary disease overlap syndrome (ACOS). Expert Opin Pharmacother 2020; 21:213-231. [PMID: 31955671 DOI: 10.1080/14656566.2019.1701656] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Asthma-chronic obstructive pulmonary disease overlap syndrome (ACOS) is a disease phenotype that shares T helper lymphocyte cell Th1/neutrophilic/non-Type-2 Inflammation pathways thought to be key in COPD and Th2/eosinophilic/Type-2 inflammatory pathways of asthma. The pharmacology of treating ACOS is challenging in severe circumstances.Areas covered: This review evaluates the stepwise treatment of ACOS using pharmacological treatments used in both COPD and asthma. The most common medications involve the same inhalers used to treat COPD and asthma patients. Advanced stepwise therapies for ACOS patients are based on patient characteristics and biomarkers. Very few clinical trials exist that focus specifically on ACOS patients.Expert opinion: After inhalers, advanced therapies including phosphodiesterase inhibitors, macrolides, N-acetylcysteine and statin therapy for those ACOS patients with a COPD appearance and exacerbations are available. In atopic ACOS patients with exacerbations, advanced asthma therapies (leukotriene receptor antagonists and synthesis blocking agents.) are used. ACOS patients with elevated blood eosinophil/IgE levels are considered for immunotherapy or therapeutic monoclonal antibodies blocking specific Th2/Type-2 interleukins or IgE. Symptom control, stabilization/improvement in pulmonary function and reduced exacerbations are the metrics of success. More pharmacological trials of ACOS patients are needed to better understand which patients benefit from specific treatments.Abbreviations: 5-LOi: 5-lipoxygenase inhibitor; ACOS: asthma - COPD overlap syndrome; B2AR: Beta2 adrenergic receptors; cAMP: cyclic adenosine monophosphate; cGMP: cyclic guanosine monophosphate; CI: confidence interval; COPD: chronic obstructive pulmonary disease; CRS : chronic rhinosinusitis; cys-LT: cysteinyl leukotrienes; DPI: dry powder inhaler; EMA: European Medicines Agency; FDA: US Food and Drug Administration; FDC: fixed-dose combination; FeNO: exhaled nitric oxide; FEV1: forced expiratory volume in one second; FVC: forced vital capacity; GM-CSF: granulocyte-macrophage colony-stimulating factor; ICS : inhaled corticosteroids; IL: interleukin; ILC2: Type 2 innate lymphoid cells; IP3: Inositol triphosphate; IRR: incidence rate ratio; KOLD: Korean Obstructive Lung Disease; LABA: long-acting B2 adrenergic receptor agonist; LAMA: long-acting muscarinic receptor antagonist; LRA: leukotriene receptor antagonist; LT: leukotrienes; MDI: metered-dose inhalers; MN: M-subtype muscarinic receptors; MRA: muscarinic receptor antagonist; NAC: N-acetylcysteine; NEB: nebulization; OR: odds ratio; PDE: phosphodiesterase; PEFR: peak expiratory flow rate; PGD2: prostaglandin D2; PRN: as needed; RR: risk ratio; SABA: short-acting B2 adrenergic receptor agonist; SAMA: short-acting muscarinic receptor antagonist; SDMI: spring-driven mist inhaler; Th1: T helper cell 1 lymphocyte; Th2: T helper cell 2 lymphocytes; TNF-α: tumor necrosis factor alpha; US : United States.
Collapse
Affiliation(s)
- Timothy E Albertson
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California, Davis, Sacramento, CA, USA.,Department of Emergency Medicine, University of California, Davis, Sacramento, CA, USA.,Veterans Administration Northern California Health Care System, Department of Medicine, Mather, CA, USA
| | - James A Chenoweth
- Department of Emergency Medicine, University of California, Davis, Sacramento, CA, USA.,Veterans Administration Northern California Health Care System, Department of Medicine, Mather, CA, USA
| | - Skyler J Pearson
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California, Davis, Sacramento, CA, USA.,Veterans Administration Northern California Health Care System, Department of Medicine, Mather, CA, USA
| | - Susan Murin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California, Davis, Sacramento, CA, USA.,Veterans Administration Northern California Health Care System, Department of Medicine, Mather, CA, USA
| |
Collapse
|
33
|
Peterson NC, Mahalingaiah PK, Fullerton A, Di Piazza M. Application of microphysiological systems in biopharmaceutical research and development. LAB ON A CHIP 2020; 20:697-708. [PMID: 31967156 DOI: 10.1039/c9lc00962k] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Within the last 10 years, several tissue microphysiological systems (MPS) have been developed and characterized for retention of morphologic characteristics and specific gene/protein expression profiles from their natural in vivo state. Once developed, their utility is typically further tested by comparing responses to known toxic small-molecule pharmaceuticals in efforts to develop strategies for further toxicity testing of compounds under development. More recently, application of this technology in biopharmaceutical (large molecules) development is beginning to be more appreciated. In this review, we describe some of the advances made for tissue-specific MPS and outline the advantages and challenges of applying and further developing MPS technology in preclinical biopharmaceutical research.
Collapse
Affiliation(s)
- Norman C Peterson
- Clinical Pharmacology and Safety Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA.
| | | | | | - Matteo Di Piazza
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Rd, Ridgefield, CT 06877, USA
| |
Collapse
|
34
|
Hillas G, Fouka E, Papaioannou AI. Antibodies targeting the interleukin-5 signaling pathway used as add-on therapy for patients with severe eosinophilic asthma: a review of the mechanism of action, efficacy, and safety of the subcutaneously administered agents, mepolizumab and benralizumab. Expert Rev Respir Med 2020; 14:353-365. [PMID: 31958239 DOI: 10.1080/17476348.2020.1718495] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Since the discovery of eosinophils in the sputum of asthmatic patients, several studies have offered evidence on their prominent role in the pathology and severity of asthma. Blood eosinophils, are a useful biomarker for therapy selection in severe asthma patients. IL-5 plays crucial role on maturation, activation, recruitment, and survival of eosinophils and constitutes an important therapeutic target for patients with severe uncontrolled eosinophilic asthma.Areas covered: This review focuses on the similarities and differences on mechanisms of action, efficacy, and safety, of two subcutaneously(SC) administered agents, the anti-interleukin(IL)-5 monoclonal antibody mepolizumab and the IL-5 receptor-α(IL-5Rα)-directed cytolytic monoclonal antibody benralizumab. All information used was collected from PubMed using keywords such as severe asthma, eosinophils, IL-5, airway inflammation, asthma exacerbations, mepolizumab, benralizumab, anti-IL5, and anti-IL5R either as single terms or in several combinations.Expert opinion: Both mepolizumab and benralizumab are promising for the treatment of severe eosinophilic asthma resulting in asthma control improvement and exacerbations reduction and can serve as steroid-sparing agents. However, since no head-to-head comparisons exist, it is unknown whether their different mechanisms of action might be related to different efficacy in specific patients' sub-phenotypes. Long-term clinical observations will provide real-world evidence regarding their lasting effectiveness and safety.
Collapse
Affiliation(s)
- Georgios Hillas
- 5th Pulmonary Department, Sotiria Chest Diseases Hospital, Athens, Greece
| | - Evangelia Fouka
- Pulmonary Department of Aristotele University of Thessaloniki, G Papanikolaou Hospital, Thessaloniki, Greece
| | - Andriana I Papaioannou
- 2nd Respiratory Medicine Department, University of Athens, Attikon Hospital, Athens, Greece
| |
Collapse
|
35
|
Chen MH, Huang MT, Yu WK, Lee SS, Wang JH, Cheng TJR, Bowman MR, Hsieh SL. Antibody blockade of Dectin-2 suppresses house dust mite-induced Th2 cytokine production in dendritic cell- and monocyte-depleted peripheral blood mononuclear cell co-cultures from asthma patients. J Biomed Sci 2019; 26:97. [PMID: 31861989 PMCID: PMC6925444 DOI: 10.1186/s12929-019-0598-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Background Dectin-2, which is a C-type lectin, interacts with the house dust mite (HDM) Dermatophagoides pteronyssinus allergen. This study aimed to investigate whether Dectin-2 blockade by antagonistic monoclonal antibodies (MoAbs) attenuates HDM-induced allergic responses. Methods Two anti-Dectin-2 MoAbs were generated and validated for specific binding to Dectin-2 Fc fusion protein (Dectin-2.Fc) and inhibition of Dectin-2.Fc/HDM interaction. Patients with asthma exhibiting high titers of anti-D. pteronyssinus IgE were enrolled. Peripheral blood mononuclear cells with depleted CD14+ monocytes were obtained from these patients and co-cultured with autologous monocyte-derived conventional dendritic cells in the presence of D. pteronyssinus or its group 2 allergens (Der p 2). Interleukin (IL)-5 and IL-13 levels in the culture supernatants were determined using ELISA in the presence or absence of anti-Dectin-2 MoAbs. Results Two MoAbs, 6A4G7 and 17A1D10, showed specific binding to recombinant Dectin-2.Fc and inhibited HDM binding to Dectin-2.Fc. Both anti-Dectin-2 MoAbs inhibited IL-5 and IL-13 production in co-cultures with Der p 2 stimulation in a dose-dependent manner. 6A4G7 and 17A1D10 (3 μg/mL) significantly inhibited Der p 2-induced (3 μg/mL) IL-5 production by 69.7 and 86.4% and IL-13 production by 84.0 and 81.4%, respectively. Moreover, this inhibitory effect of the two MoAbs remained significant in the presence of D. pteronyssinus. Conclusions Anti-Dectin-2 MoAbs significantly inhibited HDM-induced allergic responses in vitro and therefore have the potential to become therapeutic agents in mite-induced allergic diseases.
Collapse
Affiliation(s)
- Ming-Han Chen
- Division of Allergy, Immunology & Rheumatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | - Wen-Kuang Yu
- Department of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shinn-Shing Lee
- Section of Allergy, Immunology, and Rheumatology, Department of Medicine, Cheng Hsin Rehabilitation Medical Center, Taipei, Taiwan
| | - Jia-Horng Wang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Critical Care, Far Eastern Memorial Hospital, Taipei, Taiwan
| | | | - Michael R Bowman
- Inflammation and Immunology Research Unit, Pfizer Inc, Cambridge, MA, USA.,Present address: Immunology and Inflammation Therapeutic Area, Sanofi, Cambridge, MA, USA
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Taipei, Taiwan. .,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan. .,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan. .,Institute for Cancer Biology and Drug Discovery, Taipei Medical University, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan.
| |
Collapse
|
36
|
Yao Y, Gu Y, Yang M, Cao D, Wu F. The Gene Expression Biomarkers for Chronic Obstructive Pulmonary Disease and Interstitial Lung Disease. Front Genet 2019; 10:1154. [PMID: 31824564 PMCID: PMC6879656 DOI: 10.3389/fgene.2019.01154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023] Open
Abstract
COPD (chronic obstructive pulmonary disease) and ILD (interstitial lung disease) are two common respiratory diseases. They share similar clinical traits but require different therapeutic treatments. Identifying the biomarkers that are differentially expressed between them will not only help the diagnosis of COPD and ILD, but also provide candidate drug targets that may facilitate the development of new treatment for COPD and ILD. Due to the irreversible complex pathological changes of COPD, there are very limited therapeutic options for COPD patients. In this study, we analyzed the gene expression profiles of two datasets: one training dataset that includes 144 COPD patients and 194 ILD patients, and one test dataset that includes 75 COPD patients and 61 ILD patients. Advanced feature selection methods, mRMR (minimal Redundancy Maximal Relevance) and incremental feature selection (IFS), were applied to identify the 38-gene biomarker. An SVM (support vector machine) classifier was built based on the 38-gene biomarker. Its accuracy, sensitivity, and specificity on training dataset evaluated by leave one out cross-validation were 0.905, 0.896, and 0.912, respectively. And on independent test dataset, the accuracy, sensitivity, and specificity on were as great as and were 0.904, 0.933, and 0.869, respectively. The biological function analysis of the 38 genes indicated that many of them can be potential treatment targets that may benefit COPD and ILD patients.
Collapse
Affiliation(s)
- Yangwei Yao
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Yangyang Gu
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Meng Yang
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Dakui Cao
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| | - Fengjie Wu
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Jiaxing, Jiaxing, China
| |
Collapse
|
37
|
Britto CJ, Cohn L. Escalating Mucus Inhibition to the Top of Our Priorities. Am J Respir Cell Mol Biol 2019; 61:275-276. [PMID: 31063695 PMCID: PMC6839933 DOI: 10.1165/rcmb.2019-0143ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Clemente J Britto
- Section of Pulmonary, Critical Care, and Sleep MedicineYale School of MedicineNew Haven, Connecticut
| | - Lauren Cohn
- Section of Pulmonary, Critical Care, and Sleep MedicineYale School of MedicineNew Haven, Connecticut
| |
Collapse
|
38
|
Donovan T, Crossingham I, Milan SJ, Wang R, Bradley P. Anti-IL5 therapies for chronic obstructive pulmonary disease. Hippokratia 2019. [DOI: 10.1002/14651858.cd013432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Tim Donovan
- University of Cumbria; Medical and Sport Sciences; Lancaster UK
| | | | | | - Ran Wang
- Manchester University NHS Foundation Trust; Department of Respiratory Medicine; Manchester UK
| | - Patrick Bradley
- East Lancashire Hospitals NHS Trust; Blackburn Lancashire UK
| |
Collapse
|
39
|
Al-Ayed M, Alshaybari K, Alshehri D, Jamaan A, Nasser I, Alaamri H, Alaseeri W, Mahfouz AA, Ali Alsareli S, Asaad AM, Ali Magzoub A, Qureshi MA, Shalayel MH. Obesity and childhood asthma in male schoolchildren in Saudi Arabia: Is there a role for leptin, interleukin-4, interleukin-5, and interleukin-21? Ann Saudi Med 2019; 39:295-301. [PMID: 31580718 PMCID: PMC6832322 DOI: 10.5144/0256-4947.2019.295] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Adiposity is associated with high serum levels of adipokines and chemokines which are possibly implicated in a co-existence of obesity and asthma. OBJECTIVES Elucidate the possible roles of leptin, interleukin (IL)-4, IL-5 and IL-21 in linking obesity with childhood asthma. DESIGN Cross-sectional, analytical. SETTING Population of schoolchildren in a small Saudi city. SUBJECTS AND METHODS The study included a representative sample of Saudi schoolchildren grouped as obese asthmatics, non-obese asthmatics, or obese nonasthmatics, with nonobese nonasthmatics as a control group. An asthma control test was done for the asthmatic groups. MAIN OUTCOME MEASURES Serum levels of leptin, IL-4, IL-5, and IL-21. SAMPLE SIZE 345 male schoolchildren with a mean (SD) age of 13.0 (2.3) years. RESULTS Median serum leptin concentrations in obese asthmatics were significantly higher than in nonobese asthmatics ( P<.001). Uncontrolled asthmatics also had significantly higher leptin levels than controlled asthmatic children ( P<.002). Leptin levels were weakly but significantly correlated with the cytokines IL-4, IL-5, and IL-21. CONCLUSIONS Leptin may contribute to a link between obesity and childhood asthma. Differences in IL-21 levels between nonobese and obese asthmatics suggest that the co-existence of asthma and obesity increased IL-21 levels. Leptin plus some proinflammatory cytokines especially IL-21 may be potential predictors for asthma control in children. LIMITATIONS Blood sampling at different stages of asthma might influence cytokine expression. CONFLICT OF INTEREST None.
Collapse
Affiliation(s)
- Mohammed Al-Ayed
- From the Department of Pediatrics, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Khalid Alshaybari
- From the Department of Pediatrics, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Dhafer Alshehri
- From the Department of Pediatrics, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Alzahrani Jamaan
- From the Department of Pediatrics, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Iman Nasser
- From the Department of Pediatrics, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Hamdan Alaamri
- From the Department of Pediatrics, Armed Forces Medical Services, Ministry of Defense, Najran, Saudi Arabia
| | - Wed Alaseeri
- From the Department of Pediatrics, Ministry of Health, Saudi Arabia
| | - Ahmed A Mahfouz
- From the Department of Family Community Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | | | - Ahmed Morad Asaad
- From the Department of Microbiology, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Aamir Ali Magzoub
- From the Department Physiology, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Mohamed Ansar Qureshi
- From the Department of Microbiology, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Mohammed Helmy Shalayel
- From the Department of Biochemistry, College of Medicine, Najran University, Najran, Saudi Arabia
| |
Collapse
|
40
|
Kim BG, Ghosh P, Ahn S, Rhee DK. Pneumococcal pep27 mutant immunization suppresses allergic asthma in mice. Biochem Biophys Res Commun 2019; 514:210-216. [PMID: 31029416 DOI: 10.1016/j.bbrc.2019.04.116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/16/2019] [Indexed: 10/26/2022]
Abstract
Asthma is an allergic airway disease (AAD) characterized by eosinophilic inflammation, mucus hypersecretion, and airway hyper responsiveness, and it is caused by dysregulated immune responses. Conversely, regulatory T cells (Tregs) control aberrant immune responses and maintain homeostasis. Recent evidence suggests that Streptococcus pneumoniae, including its components as well as a live attenuated mutant, and pneumococcal infection induce Tregs and can thus potentially be harnessed therapeutically for asthma treatment. Previously, a pep27 deletion mutant (Δpep27) demonstrated a significantly attenuated virulence in a sepsis model, and Δpep27 immunization induced serotype-nonspecific protection against S. pneumoniae infection, as well as influenza virus, possibly via an immune tolerance mechanism. Here, the potential of Δpep27 immunization for asthma protection was studied. Mice were immunized intranasally with Δpep27 before or after ovalbumin sensitization and subsequent challenge. Δpep27 immunization suppressed hallmark features of AAD, including antigen-specific type 2 helper T cell cytokine and antibody responses, peripheral and pulmonary eosinophil accumulation, and goblet cell hyperplasia. Thus, a Δpep27 vaccine may be highly feasible as a preventive or therapeutic agent for asthma.
Collapse
Affiliation(s)
- Bo-Gyeong Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Prachetash Ghosh
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Saemi Ahn
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
41
|
Yuan F, Liu R, Hu M, Rong X, Bai L, Xu L, Mao Y, Hasimu H, Sun Y, He J. JAX2, an ethanol extract of Hyssopus cuspidatus Boriss, can prevent bronchial asthma by inhibiting MAPK/NF-κB inflammatory signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:305-314. [PMID: 30807985 DOI: 10.1016/j.phymed.2018.12.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/18/2018] [Accepted: 12/29/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Hyssopus cuspidatus Boriss has been used to treat bronchial asthma for many years in Uighur medicine. JAX2, an ethanol extract from this plant, has effectiveness against bronchial asthma. However, the molecular basis for the anti-inflammatory effects of JAX2 remains unclear. PURPOSE This study aimed to evaluate the mechanism of JAX2 against bronchial asthma. METHODS We established an asthma model in rats using ovalbumin (OVA), and an inflammatory model in RAW264.7 cells using lipopolysaccharide (LPS) stimulation. To evaluate the anti-inflammatory effects of JAX2, the concentrations of tumor necrosis factor (TNF)-α, interleukin (IL)-4, IL-6, IL-17, eotaxin and immunoglobulin (Ig)E were measured by enzyme-linked immunosorbent assay (ELISA). Cell viability was investigated by the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)2H-tetrazolium, inner salt (MTS) assay. Further, nitric oxide (NO) and reactive oxygen species (ROS) were determined using Griess reagent and 2,7-dichlorofluorescein diacetate. The phosphorylation of p-extracellular signal-regulated kinase (ERK), p-Jun-NH2-terminal kinase (JNK), p38 kinases (p38) and p-inhibitor of nuclear factor kappa-B kinase (IKK), and nuclear translocation of p-p65 kinases (p-p65) were determined by immunofluorescence to uncover the effects of JAX2 on the Mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) signaling pathways. RESULTS After JAX2 administration to rats, Interferon (IFN)-γ concentrations in BALF increased significantly. Further, the concentrations of TNF-α, IL-4, IL-6, IL-17 and eotaxin in BALF, and IgE in serum decreased. JAX2 decreased TNF-α, IL-6 and NO in cell supernatant, and reduced ROS intracellularly. Concurrently, IFN-γ concentrations increased in cell supernatant significantly. In LPS-induced RAW264.7 cells, JAX2 inhibited phosphorylation of p-ERK, p-JNK and p-38 MAPK. The subsequent phosphorylation of p-IKK and nuclear translocation of the p-p65 subunit of NF-κB were also suppressed. CONCLUSION Based on these findings, we believe that JAX2 has both preventive and treatment effects in bronchial asthma. Furthermore, in the RAW264.7 cell inflammatory model, JAX2 also inhibited NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Fengjuan Yuan
- Xinjiang Institute of Materia Medica, Urumqi 830004, China; Xinjiang Uygur Autonomous Region for Food and Drug Control, Urumqi 830002, China
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050,China
| | - Mengying Hu
- Xinjiang Institute of Materia Medica, Urumqi 830004, China
| | - Xiaojuan Rong
- Xinjiang Institute of Materia Medica, Urumqi 830004, China
| | - Liping Bai
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050,China
| | - Lei Xu
- Xinjiang Institute of Materia Medica, Urumqi 830004, China
| | - Yan Mao
- Xinjiang Institute of Materia Medica, Urumqi 830004, China
| | | | - Yuhua Sun
- Xinjiang Institute of Materia Medica, Urumqi 830004, China.
| | - Jinhua He
- Xinjiang Institute of Materia Medica, Urumqi 830004, China.
| |
Collapse
|
42
|
Ashoor DN, Ben Khalaf N, Bourguiba-Hachemi S, Marzouq MH, Fathallah MD. Engineering of the upper hinge region of human IgG1 Fc enhances the binding affinity to FcγIIIa (CD16a) receptor isoform. Protein Eng Des Sel 2019; 31:205-212. [PMID: 30299461 DOI: 10.1093/protein/gzy019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 08/04/2018] [Indexed: 11/12/2022] Open
Abstract
The interaction between antibodies and Immune cells surface FcγRIIIa (CD16a) receptor triggers a variety of immune responses including antibody-dependent cell-mediated cytotoxicity, antibody neutralization, phagocytosis, inflammation and tissue injury. Recent studies showed that IgG1 upper hinge region and FcγRs polymorphism play a major role in the interaction with Fcγ receptors and in the stability of the immune complex hence, in mounting strong inflammatory response. To further investigate this issue, we developed a tool box of IgG1 Fc isoforms to depict the affinity between mutated IgG1 Fc regions and extracellular domain variants (V158F) of CD16a. Our strategy consisted of designing different random upper-hinge mutated variants of IgG1 Fc domain, reproducing the naturally occurring two variants of CD16a and producing all of them as recombinant fusion proteins in Pichia Pastoris. The interactions were assayed using the Surface Plasmon Resonance (Biacore) method along with an in silico analysis to identify the major interaction and key residues that underline the affinity between the Fc region and CD16a variants. Our data showed that the affinity of the Fc region to the CD16a is strongly correlated to polar interactions. This molecular engineering approach yielded an IgG1Fc mutant with enhanced binding affinity to CD16a F158 variant.
Collapse
Affiliation(s)
- Dana N Ashoor
- Health Biotechnology Program, Department of Life Sciences, College of Graduate Studies, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Noureddine Ben Khalaf
- Health Biotechnology Program, Department of Life Sciences, College of Graduate Studies, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Sonia Bourguiba-Hachemi
- Health Biotechnology Program, Department of Life Sciences, College of Graduate Studies, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Maryam H Marzouq
- Health Biotechnology Program, Department of Life Sciences, College of Graduate Studies, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - M Dahmani Fathallah
- Health Biotechnology Program, Department of Life Sciences, College of Graduate Studies, Arabian Gulf University, Manama, Kingdom of Bahrain
| |
Collapse
|
43
|
Sridhar S, Liu H, Pham TH, Damera G, Newbold P. Modulation of blood inflammatory markers by benralizumab in patients with eosinophilic airway diseases. Respir Res 2019; 20:14. [PMID: 30658649 PMCID: PMC6339432 DOI: 10.1186/s12931-018-0968-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 12/19/2018] [Indexed: 01/01/2023] Open
Abstract
Background Benralizumab, a humanized, afucosylated, monoclonal antibody that targets interleukin-5 receptor α, depletes eosinophils and basophils by enhanced antibody-dependent cell-mediated cytotoxicity. It demonstrated efficacy for patients with moderate to severe asthma and, in a Phase IIa trial, for chronic obstructive pulmonary disease (COPD) with eosinophilic inflammation. We investigated effects of benralizumab 100 mg every 8 weeks (first three doses every 4 weeks) subcutaneous on blood inflammatory markers through proteomic and gene-expression analyses collected during two Phase II studies of patients with eosinophilic asthma and eosinophilic COPD. Methods Serum samples for proteomic analysis and whole blood for gene expression analysis were collected at baseline and 52 weeks (asthma study) or 32 weeks (COPD study) post-treatment. Proteomic analyses were conducted on a custom set of 90 and 147 Rules-Based Medicine analytes for asthma and COPD, respectively. Gene expression was profiled by Affymetrix Human Genome U133 plus 2 arrays (~ 54 K probes). Gene set variation analysis (GSVA) was used to determine transcriptomic activity of immune signatures. Treatment-related differences between analytes, genes, and gene signatures were analyzed for the overall population and for patient subgroups stratified by baseline blood eosinophil count (eosinophil-high [≥300 cells/μL] and eosinophil-low [< 300 cells/μL]) via t-test and repeated measures analysis of variance. Results Eosinophil chemokines eotaxin-1 and eotaxin-2 were significantly upregulated (false discovery rate [FDR] < 0.05) by approximately 2.1- and 1.4-fold in the asthma study and by 2.3- and 1.7-fold in the COPD study following benralizumab treatment. Magnitude of upregulation of these two chemokines was greater for eosinophil-high patients than eosinophil-low patients in both studies. Benralizumab was associated with significant reductions (FDR < 0.05) in expression of genes associated with eosinophils and basophils, such as CLC, IL-5Rα, and PRSS33; immune-signaling complex genes (FCER1A); G-protein–coupled receptor genes (HRH4, ADORA3, P2RY14); and further immune-related genes (ALOX15 and OLIG2). The magnitude of downregulation of gene expression was greater for eosinophil-high than eosinophil-low patients. GSVA on immune signatures indicated significant treatment reductions (FDR < 0.05) in eosinophil-associated signatures. Conclusions Benralizumab is highly selective, modulating blood proteins or genes associated with eosinophils or basophils. Modulated protein and gene expression patterns are most prominently altered in eosinophil-high vs. eosinophil-low patients. Trial registration NCT01227278 and NCT01238861. Electronic supplementary material The online version of this article (10.1186/s12931-018-0968-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sriram Sridhar
- MedImmune LLC, One MedImmune Way, #4552B, Gaithersburg, MD, USA
| | - Hao Liu
- MedImmune LLC, One MedImmune Way, #4552B, Gaithersburg, MD, USA
| | - Tuyet-Hang Pham
- MedImmune LLC, One MedImmune Way, #4552B, Gaithersburg, MD, USA
| | - Gautam Damera
- MedImmune LLC, One MedImmune Way, #4552B, Gaithersburg, MD, USA
| | - Paul Newbold
- MedImmune LLC, One MedImmune Way, #4552B, Gaithersburg, MD, USA.
| |
Collapse
|
44
|
Fernandez Romero GA, Beros J, Criner G. Mepolizumab for the prevention of chronic obstructive pulmonary disease exacerbations. Expert Rev Respir Med 2018; 13:125-132. [DOI: 10.1080/17476348.2019.1561287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
| | - Joanna Beros
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Gerard Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
45
|
Yoo SM, Chung SH. Targets of monoclonal antibodies for immunological diseases. Arch Pharm Res 2018; 42:293-304. [PMID: 30426387 DOI: 10.1007/s12272-018-1087-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/31/2018] [Indexed: 01/28/2023]
Abstract
Immunological disorders such as allergy, autoimmune diseases, auto-inflammatory syndromes and immunological deficiency syndromes are difficult to treat with chemical drugs. Recently, many monoclonal antibodies targeting various molecules including interleukin, tumor necrosis factor-α, cluster of differentiation, integrins, complement C5 and B lymphocyte stimulator are clinically available and give a hope to patients suffering from these intractable diseases. Here, we selected twenty-seven monoclonal antibodies approved by US FDA since 1997 and they are classified according to their target molecules. Although these biomedicines possessed some restrictions such as high cost and susceptible to infectious disease, these drawbacks can be overcome through cost-cutting innovations including biosimilars and careful monitoring. New targets are emerging rapidly and more effective biomedicines with acceptable side effects are in the pipeline for next decade.
Collapse
Affiliation(s)
- Seon Min Yoo
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Sung Hyun Chung
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea.
| |
Collapse
|
46
|
Henriksen DP, Bodtger U, Sidenius K, Maltbaek N, Pedersen L, Madsen H, Andersson EA, Norgaard O, Madsen LK, Chawes BL. Efficacy, adverse events, and inter-drug comparison of mepolizumab and reslizumab anti-IL-5 treatments of severe asthma - a systematic review and meta-analysis. Eur Clin Respir J 2018; 5:1536097. [PMID: 30533206 PMCID: PMC6282428 DOI: 10.1080/20018525.2018.1536097] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/28/2018] [Accepted: 10/06/2018] [Indexed: 12/25/2022] Open
Abstract
Background: New, complex, and expensive therapies targeting Interleukin-5 (IL-5) to treat severe eosinophilic asthma are emerging. Objective: To assess efficacy, adverse events, and inter-drug comparison of mepolizumab and reslizumab for treating severe eosinophilic asthma. Design: A systematic review and meta-analysis on randomized, placebo-controlled, clinical trials elucidating two critical (exacerbation rate and oral corticosteroid (OCS) use) and six important clinical outcomes on the efficacy and safety of mepolizumab and reslizumab. Results: Five studies (N = 2197) contributed with data for exacerbation rate, showing a reduction of 53% (95% CI 46; 59) in favour of anti-IL-5, corresponding to -0.94 annual exacerbations (95% CI -1.08;-0.82), thus exceeding the predefined minimal clinical important difference (MCID) of 25% reduction of the estimated ≥2 annual exacerbations. Quality of evidence was considered moderate, with low heterogeneity in study findings (I2 = 0%). One study (N = 135) contributed with data on percentage of patients experiencing ≥50% reduction inoral corticosteroid treatment, showing an effect of 20% (95% CI 2.3;47) in favour of anti-IL-5 treatment (mepolizumab), thus exceeding the predefined MCID of 10%. Quality of evidence was considered low. Compared to placebo, anti-IL-5 showed significant improvements in lung function, asthma control, and asthma-related quality of life, but below the MCIDs. No differences were observed for serious adverse events and number of patients, who dropped out. No studies evaluating sickleave or head-to-head comparisons were identified. By indirect comparison, we found no significant difference between mepolizumab and reslizumab in any ofthe predefined clinical outcomes. OCS treatment reduction could not be compared due to lack of reslizumab studies investigating this outcome. Conclusions: Mepolizumab and reslizumab provide significant and clinically relevant improvements in exacerbation rate and OCS reduction. Indirect, inter-study comparisons revealed no differences between the anti-IL-5 drugs in efficacy or safety measures.
Collapse
Affiliation(s)
- Daniel P. Henriksen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Uffe Bodtger
- Department of Respiratory Medicine, Næstved Hospital, Næstved, Denmark
| | | | - Niels Maltbaek
- Department of Medicine, Zealand University Hospital, Roskilde, Denmark
| | - Lars Pedersen
- Department of Respiratory Medicine, Bispebjerg Hospital, Copenhagen, Denmark
| | - Hanne Madsen
- Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark
| | | | - Ole Norgaard
- The Danish Medicines Council Secretariat, Copenhagen, Denmark
| | | | - Bo L. Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Cheng SL. Blood eosinophils and inhaled corticosteroids in patients with COPD: systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis 2018; 13:2775-2784. [PMID: 30233168 PMCID: PMC6132232 DOI: 10.2147/copd.s175017] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background COPD is a highly heterogeneous disease. Potential biomarkers to identify patients with COPD who will derive the greatest benefit from inhaled corticosteroid (ICS) treatment are needed. Blood eosinophil count can serve as a predictive biomarker for the efficacy of ICS treatment. The aim of this systematic review and meta-analysis was to assess whether a blood eosinophil count of ≥2% in patients undergoing ICS therapy was associated with a greater reduction in COPD exacerbation rate and pneumonia incidence. Materials and methods An electronic search was performed using the keywords "COPD", "eosinophil", and "clinical trial" in the PubMed and EMBASE databases to retrieve articles, up to 2017, relevant to our focus. Data were extracted, and a meta-analysis was conducted using RevMan 5 (version 5.3.5). Results Five studies comprising 12,496 patients with moderate-to-very severe COPD were included. At baseline, 60% of the patients had ≥2% blood eosinophils. Our meta-analysis showed a 17% reduction in exacerbation of moderate/severe COPD in patients with ≥2% blood eosinophils undergoing ICS therapy compared to the non-ICS/ICS withdrawal/placebo group. The difference between the two types of treatment was significant (risk ratio [RR], 0.816; 95% CI, 0.67-0.99; P=0.03). Furthermore, the risk of pneumonia-related events was significantly increased in the subgroup with eosinophil count ≥2% undergoing ICS-containing treatments (RR, 1.969; 95% CI, 1.369-2.833; P<0.001). There was no significant difference in the subgroup with eosinophil count <2% (RR, 1.29; 95% CI, 0.888-1.879; P<0.181). Conclusion The results of our meta-analysis suggest that the 2% threshold for blood eosinophils could accurately predict ICS treatment response in patients with COPD, but increased the risk of pneumonia.
Collapse
Affiliation(s)
- Shih-Lung Cheng
- Department of Internal Medicine, Far Eastern Memorial Hospital, Taipei, Taiwan, .,Department of Chemical Engineering and Materials Science, Yuan-Ze University, Taoyuan, Taiwan,
| |
Collapse
|
48
|
Shirai T, Hirai K, Gon Y, Maruoka S, Mizumura K, Hikichi M, Holweg C, Itoh K, Inoue H, Hashimoto S. Combined Assessment of Serum Periostin and YKL-40 May Identify Asthma-COPD Overlap. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 7:134-145.e1. [PMID: 29981861 DOI: 10.1016/j.jaip.2018.06.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/02/2018] [Accepted: 06/07/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Asthma-chronic obstructive pulmonary disease (COPD) overlap (ACO) has been proposed as a different diagnosis from asthma and COPD. However, little is known about the role of serum biomarkers in ACO. OBJECTIVE To evaluate serum periostin, a type 2 biomarker, and serum chitinase-3-like protein 1 (YKL-40), a useful biomarker for COPD, in Japanese patients with asthma, ACO, or COPD, and investigate the role of these biomarkers in identifying ACO. METHODS Subjects included Japanese patients with asthma (n = 177), ACO (n = 115), or COPD (n = 61). Serum periostin, YKL-40, and total IgE, blood eosinophils, and fractional exhaled nitric oxide were measured and compared among the patients. RESULTS Serum periostin was high in both asthma and ACO, but not in COPD, whereas serum YKL-40 was high in both COPD and ACO, but not in asthma. Serum periostin levels correlated weakly with eosinophil counts in asthma, ACO, and COPD. Multivariate linear regression analysis revealed that older age, lower body mass index, higher eosinophil counts, higher total IgE, and the absence of the diagnosis of COPD were significantly associated with higher periostin levels. Based on cutoff values derived by receiver operating characteristic analysis (periostin: 55.1 ng/mL; YKL-40: 61.3 ng/mL), patients were classified into high or low groups. The proportion of patients with both high serum periostin and YKL-40 levels was significantly higher in ACO than in asthma or COPD. CONCLUSIONS Serum periostin levels were comparable between asthma and ACO, whereas YKL-40 was comparable between ACO and COPD. Combined assessment of serum periostin and YKL-40 may identify ACO.
Collapse
Affiliation(s)
- Toshihiro Shirai
- Department of Respiratory Medicine, Shizuoka General Hospital, Shizuoka, Japan.
| | - Keita Hirai
- Department of Clinical Pharmacology and Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan; Laboratory of Clinical Pharmacogenomics, Shizuoka General Hospital, Shizuoka, Japan
| | - Yasuhiro Gon
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Shuichiro Maruoka
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kenji Mizumura
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Mari Hikichi
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | | | - Kunihiko Itoh
- Department of Clinical Pharmacology and Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan; Laboratory of Clinical Pharmacogenomics, Shizuoka General Hospital, Shizuoka, Japan
| | - Hiromasa Inoue
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Shu Hashimoto
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
49
|
|
50
|
Vesicle-associated membrane protein 7-mediated eosinophil degranulation promotes allergic airway inflammation in mice. Commun Biol 2018; 1:83. [PMID: 30271964 PMCID: PMC6123774 DOI: 10.1038/s42003-018-0081-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/03/2018] [Indexed: 01/05/2023] Open
Abstract
Eosinophil degranulation is a determining factor in allergy-mediated airway pathology. Receptor-mediated degranulation in eosinophils requires vesicle-associated membrane protein 7 (VAMP-7), a principal component of the SNARE fusion machinery. The specific contribution of eosinophil degranulation to allergen-induced airway responses remains poorly understood. We generated mice with VAMP-7 gene deficiency exclusively in eosinophils (eoCRE/V7) from a cross using eosinophil-specific Cre recombinase-expressing mice crossed with VAMP-7f/f mice. Eosinophils from eoCRE/V7 mice showed deficient degranulation responses in vitro, and responses continued to be decreased following ex vivo intratracheal adoptive transfer of eoCRE/V7 eosinophils into IL-5/hE2/EPX−/− mice. Consistent with diminished degranulation responses, reduced airway hyperresponsiveness was observed in ovalbumin-sensitized and challenged eoCRE/V7 mice following methacholine inhalation. Therefore, VAMP-7 mediates eosinophil degranulation both in vitro and ex vivo, and this event augments airway hyperresponsiveness. Lian Willetts et al. demonstrate that vesicle-associated membrane protein 7 (VAMP 7), a principal component of the membrane fusion machinery, promotes eosinophil degranulation in allergic airway inflammation. This study suggests VAMP7 as a therapeutic target for ameliorating asthma.
Collapse
|