Skip to main content
Top
Published in: Respiratory Research 1/2020

01-12-2020 | Bronchoscopy | Study protocol

CASCADE: a phase 2, randomized, double-blind, placebo-controlled, parallel-group trial to evaluate the effect of tezepelumab on airway inflammation in patients with uncontrolled asthma

Authors: Claire Emson, Sarah Diver, Latifa Chachi, Ayman Megally, Cherrie Small, John Downie, Jane R. Parnes, Karin Bowen, Gene Colice, Chris E. Brightling

Published in: Respiratory Research | Issue 1/2020

Login to get access

Abstract

Background

Patients with severe, uncontrolled asthma, particularly those with a non-eosinophilic phenotype, have a great unmet need for new treatments that act on a broad range of inflammatory pathways in the airway. Tezepelumab is a human monoclonal antibody that blocks the activity of thymic stromal lymphopoietin, an epithelial cytokine. In the PATHWAY phase 2b study (NCT02054130), tezepelumab reduced exacerbations by up to 71% in adults with severe, uncontrolled asthma, irrespective of baseline eosinophilic inflammatory status. This article reports the design and objectives of the phase 2 CASCADE study.

Methods

CASCADE is an ongoing exploratory, phase 2, randomized, double-blind, placebo-controlled, parallel-group study aiming to assess the anti-inflammatory effects of tezepelumab 210 mg administered subcutaneously every 4 weeks for 28 weeks in adults aged 18–75 years with uncontrolled, moderate-to-severe asthma. The primary endpoint is the change from baseline to week 28 in airway submucosal inflammatory cells (eosinophils, neutrophils, T cells and mast cells) from bronchoscopic biopsies. Epithelial molecular phenotyping, comprising the three-gene-mean technique, will be used to assess participants’ type 2 (T2) status to enable evaluation of the anti-inflammatory effect of tezepelumab across the continuum of T2 activation. Other exploratory analyses include assessments of the impact of tezepelumab on airway remodelling, including reticular basement membrane thickening and airway epithelial integrity. At the onset of the COVID-19 pandemic, the protocol was amended to address the possibility that site visits would be limited. The amendment allowed for: at-home dosing of study drug by a healthcare professional, extension of the treatment period by up to 6 months so patients are able to attend an onsite visit to undergo the end-of-treatment bronchoscopy, and replacement of final follow-up visits with a virtual or telephone visit.

Discussion

CASCADE aims to determine the mechanisms by which tezepelumab improves clinical asthma outcomes by evaluating the effect of tezepelumab on airway inflammatory cells and remodelling in patients with moderate-to-severe, uncontrolled asthma. An important aspect of this study is the evaluation of the anti-inflammatory effect of tezepelumab across patients with differing levels of eosinophilic and T2 inflammation.

Trial registration

NCT03688074 (ClinicalTrials.gov). Registered 28 September 2018.
Literature
1.
go back to reference Chung KF, Wenzel SE, Brozek JL, Bush A, Castro M, Sterk PJ, Adcock IM, Bateman ED, Bel EH, Bleecker ER, et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J. 2014;43:343–73.PubMedCrossRef Chung KF, Wenzel SE, Brozek JL, Bush A, Castro M, Sterk PJ, Adcock IM, Bateman ED, Bel EH, Bleecker ER, et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J. 2014;43:343–73.PubMedCrossRef
2.
go back to reference Zeiger RS, Schatz M, Chen W, Li Q, Khatry DB, Tran TN. Adherent uncontrolled adult persistent asthma: characteristics and asthma outcomes. J Allergy Clin Immunol Pract. 2015;3:986–90.PubMedCrossRef Zeiger RS, Schatz M, Chen W, Li Q, Khatry DB, Tran TN. Adherent uncontrolled adult persistent asthma: characteristics and asthma outcomes. J Allergy Clin Immunol Pract. 2015;3:986–90.PubMedCrossRef
4.
go back to reference Chastek B, Korrer S, Nagar SP, Albers F, Yancey S, Ortega H, Forshag M, Dalal AA. Economic burden of illness among patients with severe asthma in a managed care setting. J Manag Care Spec Pharm. 2016;22:848–61.PubMed Chastek B, Korrer S, Nagar SP, Albers F, Yancey S, Ortega H, Forshag M, Dalal AA. Economic burden of illness among patients with severe asthma in a managed care setting. J Manag Care Spec Pharm. 2016;22:848–61.PubMed
5.
go back to reference Farne HA, Wilson A, Powell C, Bax L, Milan SJ. Anti-IL5 therapies for asthma. Cochrane Database Syst Rev. 2017;9:CD010834.PubMed Farne HA, Wilson A, Powell C, Bax L, Milan SJ. Anti-IL5 therapies for asthma. Cochrane Database Syst Rev. 2017;9:CD010834.PubMed
6.
go back to reference Normansell R, Walker S, Milan SJ, Walters EH, Nair P. Omalizumab for asthma in adults and children. Cochrane Database Syst Rev. 2014;(1):CD003559. Normansell R, Walker S, Milan SJ, Walters EH, Nair P. Omalizumab for asthma in adults and children. Cochrane Database Syst Rev. 2014;(1):CD003559.
7.
go back to reference Xiong XF, Zhu M, Wu HX, Fan LL, Cheng DY. Efficacy and safety of dupilumab for the treatment of uncontrolled asthma: a meta-analysis of randomized clinical trials. Respir Res. 2019;20:108.PubMedPubMedCentralCrossRef Xiong XF, Zhu M, Wu HX, Fan LL, Cheng DY. Efficacy and safety of dupilumab for the treatment of uncontrolled asthma: a meta-analysis of randomized clinical trials. Respir Res. 2019;20:108.PubMedPubMedCentralCrossRef
8.
go back to reference Zayed Y, Kheiri B, Banifadel M, Hicks M, Aburahma A, Hamid K, Bachuwa G, Chandran A. Dupilumab safety and efficacy in uncontrolled asthma: a systematic review and meta-analysis of randomized clinical trials. J Asthma. 2018;56:1–10. Zayed Y, Kheiri B, Banifadel M, Hicks M, Aburahma A, Hamid K, Bachuwa G, Chandran A. Dupilumab safety and efficacy in uncontrolled asthma: a systematic review and meta-analysis of randomized clinical trials. J Asthma. 2018;56:1–10.
9.
go back to reference Busse WW, Bleecker ER, FitzGerald JM, Ferguson GT, Barker P, Sproule S, Olsson RF, Martin UJ, Goldman M, Bora study investigators. Long-term safety and efficacy of benralizumab in patients with severe, uncontrolled asthma: 1-year results from the BORA phase 3 extension trial. Lancet Respir Med. 2019;7:46–59. Busse WW, Bleecker ER, FitzGerald JM, Ferguson GT, Barker P, Sproule S, Olsson RF, Martin UJ, Goldman M, Bora study investigators. Long-term safety and efficacy of benralizumab in patients with severe, uncontrolled asthma: 1-year results from the BORA phase 3 extension trial. Lancet Respir Med. 2019;7:46–59.
10.
go back to reference Swedin L, Saarne T, Rehnberg M, Glader P, Niedzielska M, Johansson G, Hazon P, Catley MC. Patient stratification and the unmet need in asthma. Pharmacol Ther. 2017;169:13–34.PubMedCrossRef Swedin L, Saarne T, Rehnberg M, Glader P, Niedzielska M, Johansson G, Hazon P, Catley MC. Patient stratification and the unmet need in asthma. Pharmacol Ther. 2017;169:13–34.PubMedCrossRef
11.
go back to reference Castro M, Corren J, Pavord ID, Maspero J, Wenzel S, Rabe KF, Busse WW, Ford L, Sher L, FitzGerald JM, et al. Dupilumab efficacy and safety in moderate-to-severe uncontrolled asthma. N Engl J Med. 2018;378:2486–96.PubMedCrossRef Castro M, Corren J, Pavord ID, Maspero J, Wenzel S, Rabe KF, Busse WW, Ford L, Sher L, FitzGerald JM, et al. Dupilumab efficacy and safety in moderate-to-severe uncontrolled asthma. N Engl J Med. 2018;378:2486–96.PubMedCrossRef
12.
go back to reference Hanania NA, Wenzel S, Rosen K, Hsieh HJ, Mosesova S, Choy DF, Lal P, Arron JR, Harris JM, Busse W. Exploring the effects of omalizumab in allergic asthma: an analysis of biomarkers in the EXTRA study. Am J Respir Crit Care Med. 2013;187:804–11.PubMedCrossRef Hanania NA, Wenzel S, Rosen K, Hsieh HJ, Mosesova S, Choy DF, Lal P, Arron JR, Harris JM, Busse W. Exploring the effects of omalizumab in allergic asthma: an analysis of biomarkers in the EXTRA study. Am J Respir Crit Care Med. 2013;187:804–11.PubMedCrossRef
13.
go back to reference Gauvreau GM, O'Byrne PM, Boulet LP, Wang Y, Cockcroft D, Bigler J, FitzGerald JM, Boedigheimer M, Davis BE, Dias C, et al. Effects of an anti-TSLP antibody on allergen-induced asthmatic responses. N Engl J Med. 2014;370:2102–10.PubMedCrossRef Gauvreau GM, O'Byrne PM, Boulet LP, Wang Y, Cockcroft D, Bigler J, FitzGerald JM, Boedigheimer M, Davis BE, Dias C, et al. Effects of an anti-TSLP antibody on allergen-induced asthmatic responses. N Engl J Med. 2014;370:2102–10.PubMedCrossRef
14.
go back to reference Corren J, Parnes JR, Wang L, Mo M, Roseti SL, Griffiths JM, van der Merwe R. Tezepelumab in adults with uncontrolled asthma. N Engl J Med. 2017;377:936–46.PubMedCrossRef Corren J, Parnes JR, Wang L, Mo M, Roseti SL, Griffiths JM, van der Merwe R. Tezepelumab in adults with uncontrolled asthma. N Engl J Med. 2017;377:936–46.PubMedCrossRef
15.
go back to reference Corren J, Garcia Gil E, Parnes J, Pham T, Griffiths J. Tezepelumab treatment effect on annualized rate of exacerbations by baseline biomarkers in uncontrolled severe asthma patients: phase 2b PATHWAY study. J Allergy Clin Immunol. 2019;199:A2621. Corren J, Garcia Gil E, Parnes J, Pham T, Griffiths J. Tezepelumab treatment effect on annualized rate of exacerbations by baseline biomarkers in uncontrolled severe asthma patients: phase 2b PATHWAY study. J Allergy Clin Immunol. 2019;199:A2621.
19.
go back to reference Gauvreau GM, Sehmi R, Ambrose CS, Griffiths JM. Thymic stromal lymphopoietin: its role and potential as a therapeutic target in asthma. Expert Opin Ther Targets. 2020;1–16. Gauvreau GM, Sehmi R, Ambrose CS, Griffiths JM. Thymic stromal lymphopoietin: its role and potential as a therapeutic target in asthma. Expert Opin Ther Targets. 2020;1–16.
22.
go back to reference Choy DF, Modrek B, Abbas AR, Kummerfeld S, Clark HF, Wu LC, Fedorowicz G, Modrusan Z, Fahy JV, Woodruff PG, Arron JR. Gene expression patterns of Th2 inflammation and intercellular communication in asthmatic airways. J Immunol. 2011;186:1861–9.PubMedCrossRef Choy DF, Modrek B, Abbas AR, Kummerfeld S, Clark HF, Wu LC, Fedorowicz G, Modrusan Z, Fahy JV, Woodruff PG, Arron JR. Gene expression patterns of Th2 inflammation and intercellular communication in asthmatic airways. J Immunol. 2011;186:1861–9.PubMedCrossRef
23.
go back to reference Bhakta NR, Solberg OD, Nguyen CP, Nguyen CN, Arron JR, Fahy JV, Woodruff PG. A qPCR-based metric of Th2 airway inflammation in asthma. Clin Transl Allergy. 2013;3:24.PubMedPubMedCentralCrossRef Bhakta NR, Solberg OD, Nguyen CP, Nguyen CN, Arron JR, Fahy JV, Woodruff PG. A qPCR-based metric of Th2 airway inflammation in asthma. Clin Transl Allergy. 2013;3:24.PubMedPubMedCentralCrossRef
24.
go back to reference Woodruff PG, Modrek B, Choy DF, Jia G, Abbas AR, Ellwanger A, Koth LL, Arron JR, Fahy JV. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med. 2009;180:388–95.PubMedPubMedCentralCrossRef Woodruff PG, Modrek B, Choy DF, Jia G, Abbas AR, Ellwanger A, Koth LL, Arron JR, Fahy JV. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med. 2009;180:388–95.PubMedPubMedCentralCrossRef
25.
go back to reference Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, Sousa A, Marshall RP, Bradding P, Green RH, Wardlaw AJ, Pavord ID. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med. 2009;360:973–84.PubMedPubMedCentralCrossRef Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, Sousa A, Marshall RP, Bradding P, Green RH, Wardlaw AJ, Pavord ID. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med. 2009;360:973–84.PubMedPubMedCentralCrossRef
26.
go back to reference Laviolette M, Gossage DL, Gauvreau G, Leigh R, Olivenstein R, Katial R, Busse WW, Wenzel S, Wu Y, Datta V, et al. Effects of benralizumab on airway eosinophils in asthmatic patients with sputum eosinophilia. J Allergy Clin Immunol. 2013;132:1086–96 e1085.PubMedPubMedCentralCrossRef Laviolette M, Gossage DL, Gauvreau G, Leigh R, Olivenstein R, Katial R, Busse WW, Wenzel S, Wu Y, Datta V, et al. Effects of benralizumab on airway eosinophils in asthmatic patients with sputum eosinophilia. J Allergy Clin Immunol. 2013;132:1086–96 e1085.PubMedPubMedCentralCrossRef
27.
go back to reference Djukanovic R, Wilson SJ, Kraft M, Jarjour NN, Steel M, Chung KF, Bao W, Fowler-Taylor A, Matthews J, Busse WW, et al. Effects of treatment with anti-immunoglobulin E antibody omalizumab on airway inflammation in allergic asthma. Am J Respir Crit Care Med. 2004;170:583–93.PubMedCrossRef Djukanovic R, Wilson SJ, Kraft M, Jarjour NN, Steel M, Chung KF, Bao W, Fowler-Taylor A, Matthews J, Busse WW, et al. Effects of treatment with anti-immunoglobulin E antibody omalizumab on airway inflammation in allergic asthma. Am J Respir Crit Care Med. 2004;170:583–93.PubMedCrossRef
28.
go back to reference Riccio AM, Dal Negro RW, Micheletto C, De Ferrari L, Folli C, Chiappori A, Canonica GW. Omalizumab modulates bronchial reticular basement membrane thickness and eosinophil infiltration in severe persistent allergic asthma patients. Int J Immunopathol Pharmacol. 2012;25:475–84.PubMedCrossRef Riccio AM, Dal Negro RW, Micheletto C, De Ferrari L, Folli C, Chiappori A, Canonica GW. Omalizumab modulates bronchial reticular basement membrane thickness and eosinophil infiltration in severe persistent allergic asthma patients. Int J Immunopathol Pharmacol. 2012;25:475–84.PubMedCrossRef
29.
go back to reference van Rensen EL, Evertse CE, van Schadewijk WA, van Wijngaarden S, Ayre G, Mauad T, Hiemstra PS, Sterk PJ, Rabe KF. Eosinophils in bronchial mucosa of asthmatics after allergen challenge: effect of anti-IgE treatment. Allergy. 2009;64:72–80.PubMedCrossRef van Rensen EL, Evertse CE, van Schadewijk WA, van Wijngaarden S, Ayre G, Mauad T, Hiemstra PS, Sterk PJ, Rabe KF. Eosinophils in bronchial mucosa of asthmatics after allergen challenge: effect of anti-IgE treatment. Allergy. 2009;64:72–80.PubMedCrossRef
30.
go back to reference Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A, Ludwig MS, Barnes N, Robinson D, Kay AB. Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest. 2003;112:1029–36.PubMedPubMedCentralCrossRef Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A, Ludwig MS, Barnes N, Robinson D, Kay AB. Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest. 2003;112:1029–36.PubMedPubMedCentralCrossRef
31.
go back to reference Flood-Page PT, Menzies-Gow AN, Kay AB, Robinson DS. Eosinophil’s role remains uncertain as anti-interleukin-5 only partially depletes numbers in asthmatic airway. Am J Respir Crit Care Med. 2003;167:199–204.PubMedCrossRef Flood-Page PT, Menzies-Gow AN, Kay AB, Robinson DS. Eosinophil’s role remains uncertain as anti-interleukin-5 only partially depletes numbers in asthmatic airway. Am J Respir Crit Care Med. 2003;167:199–204.PubMedCrossRef
32.
go back to reference Menzies-Gow A, Flood-Page P, Sehmi R, Burman J, Hamid Q, Robinson DS, Kay AB, Denburg J. Anti-IL-5 (mepolizumab) therapy induces bone marrow eosinophil maturational arrest and decreases eosinophil progenitors in the bronchial mucosa of atopic asthmatics. J Allergy Clin Immunol. 2003;111:714–9.PubMedCrossRef Menzies-Gow A, Flood-Page P, Sehmi R, Burman J, Hamid Q, Robinson DS, Kay AB, Denburg J. Anti-IL-5 (mepolizumab) therapy induces bone marrow eosinophil maturational arrest and decreases eosinophil progenitors in the bronchial mucosa of atopic asthmatics. J Allergy Clin Immunol. 2003;111:714–9.PubMedCrossRef
33.
go back to reference Kelly EA, Esnault S, Liu LY, Evans MD, Johansson MW, Mathur S, Mosher DF, Denlinger LC, Jarjour NN. Mepolizumab attenuates airway eosinophil numbers, but not their functional phenotype, in asthma. Am J Respir Crit Care Med. 2017;196:1385–95.PubMedPubMedCentralCrossRef Kelly EA, Esnault S, Liu LY, Evans MD, Johansson MW, Mathur S, Mosher DF, Denlinger LC, Jarjour NN. Mepolizumab attenuates airway eosinophil numbers, but not their functional phenotype, in asthma. Am J Respir Crit Care Med. 2017;196:1385–95.PubMedPubMedCentralCrossRef
34.
go back to reference Russell RJ, Chachi L, FitzGerald JM, Backer V, Olivenstein R, Titlestad IL, Ulrik CS, Harrison T, Singh D, Chaudhuri R, et al. Effect of tralokinumab, an interleukin-13 neutralising monoclonal antibody, on eosinophilic airway inflammation in uncontrolled moderate-to-severe asthma (MESOS): a multicentre, double-blind, randomised, placebo-controlled phase 2 trial. Lancet Respir Med. 2018;6:499–510.PubMedCrossRef Russell RJ, Chachi L, FitzGerald JM, Backer V, Olivenstein R, Titlestad IL, Ulrik CS, Harrison T, Singh D, Chaudhuri R, et al. Effect of tralokinumab, an interleukin-13 neutralising monoclonal antibody, on eosinophilic airway inflammation in uncontrolled moderate-to-severe asthma (MESOS): a multicentre, double-blind, randomised, placebo-controlled phase 2 trial. Lancet Respir Med. 2018;6:499–510.PubMedCrossRef
36.
go back to reference Greer AM, Matthay MA, Kukreja J, Bhakta NR, Nguyen CP, Wolters PJ, Woodruff PG, Fahy JV, Shin JS. Accumulation of BDCA1(+) dendritic cells in interstitial fibrotic lung diseases and Th2-high asthma. PLoS One. 2014;9:e99084.PubMedPubMedCentralCrossRef Greer AM, Matthay MA, Kukreja J, Bhakta NR, Nguyen CP, Wolters PJ, Woodruff PG, Fahy JV, Shin JS. Accumulation of BDCA1(+) dendritic cells in interstitial fibrotic lung diseases and Th2-high asthma. PLoS One. 2014;9:e99084.PubMedPubMedCentralCrossRef
37.
go back to reference Jatakanon A, Uasuf C, Maziak W, Lim S, Chung KF, Barnes PJ. Neutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med. 1999;160:1532–9.PubMedCrossRef Jatakanon A, Uasuf C, Maziak W, Lim S, Chung KF, Barnes PJ. Neutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med. 1999;160:1532–9.PubMedCrossRef
38.
go back to reference Laitinen LA, Laitinen A, Haahtela T. Airway mucosal inflammation even in patients with newly diagnosed asthma. Am Rev Respir Dis. 1993;147:697–704.PubMedCrossRef Laitinen LA, Laitinen A, Haahtela T. Airway mucosal inflammation even in patients with newly diagnosed asthma. Am Rev Respir Dis. 1993;147:697–704.PubMedCrossRef
39.
go back to reference Costa JJ, Weller PF, Galli SJ. The cells of the allergic response: mast cells, basophils, and eosinophils. JAMA. 1997;278:1815–22.PubMedCrossRef Costa JJ, Weller PF, Galli SJ. The cells of the allergic response: mast cells, basophils, and eosinophils. JAMA. 1997;278:1815–22.PubMedCrossRef
40.
go back to reference Wong CK, Hu S, Cheung PF, Lam CW. Thymic stromal lymphopoietin induces chemotactic and prosurvival effects in eosinophils: implications in allergic inflammation. Am J Respir Cell Mol Biol. 2010;43:305–15.PubMedCrossRef Wong CK, Hu S, Cheung PF, Lam CW. Thymic stromal lymphopoietin induces chemotactic and prosurvival effects in eosinophils: implications in allergic inflammation. Am J Respir Cell Mol Biol. 2010;43:305–15.PubMedCrossRef
41.
go back to reference Cook EB, Stahl JL, Schwantes EA, Fox KE, Mathur SK. IL-3 and TNFalpha increase thymic stromal lymphopoietin receptor (TSLPR) expression on eosinophils and enhance TSLP-stimulated degranulation. Clin Mol Allergy. 2012;10:8.PubMedPubMedCentralCrossRef Cook EB, Stahl JL, Schwantes EA, Fox KE, Mathur SK. IL-3 and TNFalpha increase thymic stromal lymphopoietin receptor (TSLPR) expression on eosinophils and enhance TSLP-stimulated degranulation. Clin Mol Allergy. 2012;10:8.PubMedPubMedCentralCrossRef
42.
go back to reference Li Y, Wang W, Lv Z, Li Y, Chen Y, Huang K, Corrigan CJ, Ying S. Elevated expression of IL-33 and TSLP in the airways of human asthmatics in vivo: a potential biomarker of severe refractory disease. J Immunol. 2018;200:2253–62.PubMedCrossRef Li Y, Wang W, Lv Z, Li Y, Chen Y, Huang K, Corrigan CJ, Ying S. Elevated expression of IL-33 and TSLP in the airways of human asthmatics in vivo: a potential biomarker of severe refractory disease. J Immunol. 2018;200:2253–62.PubMedCrossRef
43.
go back to reference Soumelis V, Reche PA, Kanzler H, Yuan W, Edward G, Homey B, Gilliet M, Ho S, Antonenko S, Lauerma A, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol. 2002;3:673–80.CrossRefPubMed Soumelis V, Reche PA, Kanzler H, Yuan W, Edward G, Homey B, Gilliet M, Ho S, Antonenko S, Lauerma A, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol. 2002;3:673–80.CrossRefPubMed
44.
go back to reference Allakhverdi Z, Comeau MR, Jessup HK, Yoon BR, Brewer A, Chartier S, Paquette N, Ziegler SF, Sarfati M, Delespesse G. Thymic stromal lymphopoietin is released by human epithelial cells in response to microbes, trauma, or inflammation and potently activates mast cells. J Exp Med. 2007;204:253–8.PubMedPubMedCentralCrossRef Allakhverdi Z, Comeau MR, Jessup HK, Yoon BR, Brewer A, Chartier S, Paquette N, Ziegler SF, Sarfati M, Delespesse G. Thymic stromal lymphopoietin is released by human epithelial cells in response to microbes, trauma, or inflammation and potently activates mast cells. J Exp Med. 2007;204:253–8.PubMedPubMedCentralCrossRef
45.
go back to reference Torii Y, Ito T, Amakawa R, Sugimoto H, Amuro H, Tanijiri T, Katashiba Y, Ogata M, Yokoi T, Fukuhara S. Imidazoquinoline acts as immune adjuvant for functional alteration of thymic stromal lymphopoietin-mediated allergic T cell response. J Immunol. 2008;181:5340–9.PubMedCrossRef Torii Y, Ito T, Amakawa R, Sugimoto H, Amuro H, Tanijiri T, Katashiba Y, Ogata M, Yokoi T, Fukuhara S. Imidazoquinoline acts as immune adjuvant for functional alteration of thymic stromal lymphopoietin-mediated allergic T cell response. J Immunol. 2008;181:5340–9.PubMedCrossRef
46.
go back to reference Tanaka J, Watanabe N, Kido M, Saga K, Akamatsu T, Nishio A, Chiba T. Human TSLP and TLR3 ligands promote differentiation of Th17 cells with a central memory phenotype under Th2-polarizing conditions. Clin Exp Allergy. 2009;39:89–100.PubMedCrossRef Tanaka J, Watanabe N, Kido M, Saga K, Akamatsu T, Nishio A, Chiba T. Human TSLP and TLR3 ligands promote differentiation of Th17 cells with a central memory phenotype under Th2-polarizing conditions. Clin Exp Allergy. 2009;39:89–100.PubMedCrossRef
47.
go back to reference Rochman I, Watanabe N, Arima K, Liu YJ, Leonard WJ. Cutting edge: direct action of thymic stromal lymphopoietin on activated human CD4+ T cells. J Immunol. 2007;178:6720–4.PubMedCrossRef Rochman I, Watanabe N, Arima K, Liu YJ, Leonard WJ. Cutting edge: direct action of thymic stromal lymphopoietin on activated human CD4+ T cells. J Immunol. 2007;178:6720–4.PubMedCrossRef
49.
50.
go back to reference Choy DF, Hart KM, Borthwick LA, Shikotra A, Nagarkar DR, Siddiqui S, Jia G, Ohri CM, Doran E, Vannella KM, et al. TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma. Sci Transl Med. 2015;7:301ra129.PubMedCrossRef Choy DF, Hart KM, Borthwick LA, Shikotra A, Nagarkar DR, Siddiqui S, Jia G, Ohri CM, Doran E, Vannella KM, et al. TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma. Sci Transl Med. 2015;7:301ra129.PubMedCrossRef
51.
go back to reference Brightling C, Wang M, Braddock M, Nordenmark L, Gottlow M, Colice G. MESOS: considerations in designing a mechanistic study for a biologic used to treat asthma. Clin Invest (Lond). 2015;5:713–22.CrossRef Brightling C, Wang M, Braddock M, Nordenmark L, Gottlow M, Colice G. MESOS: considerations in designing a mechanistic study for a biologic used to treat asthma. Clin Invest (Lond). 2015;5:713–22.CrossRef
52.
go back to reference James AL, Maxwell PS, Pearce-Pinto G, Elliot JG, Carroll NG. The relationship of reticular basement membrane thickness to airway wall remodeling in asthma. Am J Respir Crit Care Med. 2002;166:1590–5.PubMedCrossRef James AL, Maxwell PS, Pearce-Pinto G, Elliot JG, Carroll NG. The relationship of reticular basement membrane thickness to airway wall remodeling in asthma. Am J Respir Crit Care Med. 2002;166:1590–5.PubMedCrossRef
56.
go back to reference Datta A, Alexander R, Sulikowski MG, Nicholson AG, Maher TM, Scotton CJ, Chambers RC. Evidence for a functional thymic stromal lymphopoietin signaling axis in fibrotic lung disease. J Immunol. 2013;191:4867–79.PubMedCrossRef Datta A, Alexander R, Sulikowski MG, Nicholson AG, Maher TM, Scotton CJ, Chambers RC. Evidence for a functional thymic stromal lymphopoietin signaling axis in fibrotic lung disease. J Immunol. 2013;191:4867–79.PubMedCrossRef
57.
58.
go back to reference Mauri P, Riccio AM, Rossi R, Di Silvestre D, Benazzi L, De Ferrari L, Dal Negro RW, Holgate ST, Canonica GW. Proteomics of bronchial biopsies: galectin-3 as a predictive biomarker of airway remodelling modulation in omalizumab-treated severe asthma patients. Immunol Lett. 2014;162:2–10.PubMedCrossRef Mauri P, Riccio AM, Rossi R, Di Silvestre D, Benazzi L, De Ferrari L, Dal Negro RW, Holgate ST, Canonica GW. Proteomics of bronchial biopsies: galectin-3 as a predictive biomarker of airway remodelling modulation in omalizumab-treated severe asthma patients. Immunol Lett. 2014;162:2–10.PubMedCrossRef
59.
go back to reference Riccio AM, Mauri P, De Ferrari L, Rossi R, Di Silvestre D, Benazzi L, Chiappori A, Dal Negro RW, Micheletto C, Canonica GW. Galectin-3: an early predictive biomarker of modulation of airway remodeling in patients with severe asthma treated with omalizumab for 36 months. Clin Transl Allergy. 2017;7:6.PubMedPubMedCentralCrossRef Riccio AM, Mauri P, De Ferrari L, Rossi R, Di Silvestre D, Benazzi L, Chiappori A, Dal Negro RW, Micheletto C, Canonica GW. Galectin-3: an early predictive biomarker of modulation of airway remodeling in patients with severe asthma treated with omalizumab for 36 months. Clin Transl Allergy. 2017;7:6.PubMedPubMedCentralCrossRef
60.
go back to reference Ly N, Zheng Y, Griffiths JM, van der Merwe R, Agoram B, Roskos L. Exposure-response analysis of tezepelumab in patients with severe asthma to guide phase 3 dose selection. Eur Respir J. 2018;52:PA1688. Ly N, Zheng Y, Griffiths JM, van der Merwe R, Agoram B, Roskos L. Exposure-response analysis of tezepelumab in patients with severe asthma to guide phase 3 dose selection. Eur Respir J. 2018;52:PA1688.
61.
go back to reference Fleming TR, Labriola D, Wittes J. Conducting clinical research during the COVID-19 pandemic: protecting scientific integrity. JAMA. 2020;324:33–4. Fleming TR, Labriola D, Wittes J. Conducting clinical research during the COVID-19 pandemic: protecting scientific integrity. JAMA. 2020;324:33–4.
Metadata
Title
CASCADE: a phase 2, randomized, double-blind, placebo-controlled, parallel-group trial to evaluate the effect of tezepelumab on airway inflammation in patients with uncontrolled asthma
Authors
Claire Emson
Sarah Diver
Latifa Chachi
Ayman Megally
Cherrie Small
John Downie
Jane R. Parnes
Karin Bowen
Gene Colice
Chris E. Brightling
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Respiratory Research / Issue 1/2020
Electronic ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-020-01513-x

Other articles of this Issue 1/2020

Respiratory Research 1/2020 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.