Baseline type 2 biomarker levels and response to tezepelumab in severe asthma

Jonathan Corren, Tuyet-Hang Pham, Esther Garcia Gil, Kinga Sałapa, Pin Ren, Jane R Parnes, Gene Colice, Janet M Griffiths, Jonathan Corren, Tuyet-Hang Pham, Esther Garcia Gil, Kinga Sałapa, Pin Ren, Jane R Parnes, Gene Colice, Janet M Griffiths

Abstract

Background: Tezepelumab is a human monoclonal antibody that blocks activity of thymic stromal lymphopoietin (TSLP). In the phase IIb PATHWAY study (NCT02054130), tezepelumab significantly reduced annualized asthma exacerbation rates (AAERs) versus placebo in adults with severe, uncontrolled asthma. We evaluated the effects of tezepelumab in reducing type 2 (T2) inflammatory biomarker levels in the PATHWAY population, and the relationship between baseline T2 biomarker levels and AAER.

Methods: Adults with severe, uncontrolled asthma (n = 550) were randomized to tezepelumab (70 mg or 210 mg every 4 weeks, or 280 mg every 2 weeks) or placebo for 52 weeks. Blood eosinophil count, fractional exhaled nitric oxide (FeNO), and serum total immunoglobulin (Ig)E, interleukin (IL)-5, IL-13, periostin, thymus and activation-regulated chemokine (TARC), and TSLP were measured at baseline and over 52 weeks. AAERs were analyzed by baseline threshold (high/low) biomarker levels.

Results: Positive correlations were observed between T2 inflammatory biomarkers (blood eosinophil count, FeNO, IL-5, IL-13 and periostin) at baseline. At Week 52, treatment with tezepelumab 210 mg reduced all biomarker levels measured from baseline versus placebo. Exacerbations were reduced by 55-83% in the pooled tezepelumab cohort versus placebo, irrespective of baseline blood eosinophil count, FeNO, or serum total IgE, IL-5, IL-13, periostin, TARC, or TSLP, when these biomarkers were assessed individually.

Conclusion: At baseline, positive correlations between specific T2 inflammatory biomarkers were observed. Tezepelumab reduced multiple T2 inflammatory biomarkers, which indicates decreased airway inflammation, and reduced exacerbations irrespective of baseline T2 biomarker profiles in patients with severe asthma.

Keywords: asthma; biomarkers; inflammation.

Conflict of interest statement

Dr. Corren received grants from AstraZeneca during the conduct of the study. He has also received grants from Genentech, Novartis, Optinose, Regeneron, Sanofi, and Teva, and has received personal fees from AstraZeneca, Genentech, Regeneron, and Sanofi. Ms Pham, Dr. Ren, Dr. Colice, and Dr. Griffiths are employees of AstraZeneca and own stock and stock options in AstraZeneca. Dr. Garcia Gil was an employee of AstraZeneca at the time of this study; she is a present employee of Almirall. Ms Sałapa is an employee of AstraZeneca. Dr. Parnes is an employee of Amgen Inc., and owns stock and stock options in Amgen Inc.

© 2022 AstraZeneca and Amgen Inc. Allergy published by European Academy of Allergy and Clinical Immunology and John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Baseline correlations between serum levels of IL‐5 and IL‐13 and other biomarkers of inflammation. Spearman correlation coefficients (r) are shown. Data are presented on the log scale. Abbreviations: FeNO, fractional exhaled nitric oxide; IL, interleukin; ppb, parts per billion
FIGURE 2
FIGURE 2
Median percentage change from baseline in biomarker levels over 52 weeks. Abbreviations: FeNO, fractional exhaled nitric oxide; IgE, immunoglobulin E; IL, interleukin; MAD, median absolute deviation; Q4W, every 4 weeks; TARC, thymus and activation‐regulated chemokine
FIGURE 3
FIGURE 3
Median percentage change from baseline to Week 52 in biomarker levels. FeNO, fractional exhaled nitric oxide; IgE, immunoglobulin E; IL, interleukin; ppb, parts per billion; Q4W, every 4 weeks; TARC, thymus and activation‐regulated chemokine
FIGURE 4
FIGURE 4
AAER by baseline biomarker category at Week 52. AAER = the total number of asthma exacerbations/total person‐year follow‐up in each group. *p values are nominal. Gray shaded area shows the response of the overall population. an refers to the number of patients within each subgroup based on baseline status. bAAER reduction from baseline at Week 52 vs placebo was 62% with tezepelumab 70 mg Q4W, 71% with tezepelumab 210 mg Q4W, and 66% with tezepelumab 280 mg Q2W. cData based on tezepelumab 210 mg Q4W and 280 mg Q2W data only; IL‐5 and TARC were not measured in the tezepelumab 70 mg Q4W cohort. AAER, annualized asthma exacerbation rate; CI, confidence interval; FeNO, fractional exhaled nitric oxide; IgE, immunoglobulin E; IL, interleukin; ppb, parts per billion; Q2W, every 2 weeks; Q4W, every 4 weeks; RR, rate ratio; TARC, thymus and activation‐regulated chemokine; TSLP, thymic stromal lymphopoietin

References

    1. Chung KF, Wenzel SE, Brozek JL, et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J. 2014;43(2):343‐373.
    1. Global Initiative for Asthma . Global initiative for asthma management and prevention 2020 guidelines. . Assessed February 26, 2021.
    1. Hekking PW, Wener RR, Amelink M, Zwinderman AH, Bouvy ML, Bel EH. The prevalence of severe refractory asthma. J Allergy Clin Immunol. 2015;135(4):896‐902.
    1. Jackson DJ, Busby J, Pfeffer PE, et al. Characterisation of patients with severe asthma in the UK severe asthma registry in the biologic era. Thorax. 2021;76(3):220‐227.
    1. Busse WW, Maspero JF, Rabe KF, et al. Liberty asthma QUEST: phase 3 randomized, double‐blind, placebo‐controlled, parallel‐group study to evaluate dupilumab efficacy/safety in patients with uncontrolled, moderate‐to‐severe asthma. Adv Ther. 2018;35(5):737‐748.
    1. Busse WW, Bleecker ER, FitzGerald JM, et al. 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(1):46‐59.
    1. Normansell R, Walker S, Milan SJ, Walters EH, Nair P. Omalizumab for asthma in adults and children. Cochrane Database Syst Rev. 2014(1):CD003559.
    1. Allakhverdi Z, Comeau MR, Jessup HK, et al. 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(2):253‐258.
    1. Soumelis V, Reche PA, Kanzler H, et al. Human epithelial cells trigger dendritic cell–mediated allergic inflammation by producing TSLP. Nat Immunol. 2002;3(7):673‐680.
    1. Shikotra A, Choy DF, Ohri CM, et al. Increased expression of immunoreactive thymic stromal lymphopoietin in patients with severe asthma. J Allergy Clin Immunol. 2012;129(1):104‐111.
    1. Bleck B, Tse DB, Curotto de Lafaille MA, Zhang F, Reibman J. Diesel exhaust particle‐exposed human bronchial epithelial cells induce dendritic cell maturation and polarization via thymic stromal lymphopoietin. J Clin Immunol. 2008;28(2):147‐156.
    1. Calvén J, Yudina Y, Hallgren O, et al. Viral stimuli trigger exaggerated thymic stromal lymphopoietin expression by chronic obstructive pulmonary disease epithelium: role of endosomal TLR3 and cytosolic RIG‐I‐like helicases. Innate Immun. 2012;4(1):86‐99.
    1. Ying S, O’Connor B, Ratoff J, et al. Thymic stromal lymphopoietin expression is increased in asthmatic airways and correlates with expression of Th2‐attracting chemokines and disease severity. J Immunol. 2005;174(12):8183‐8190.
    1. Ying S, O'Connor B, Ratoff J, et al. Expression and cellular provenance of thymic stromal lymphopoietin and chemokines in patients with severe asthma and chronic obstructive pulmonary disease. J Immunol. 2008;181(4):2790‐2798.
    1. Kitajima M, Lee H‐C, Nakayama T, Ziegler SF. TSLP enhances the function of helper type 2 cells. Eur J Immunol. 2011;41(7):1862‐1871.
    1. Camelo A, Rosignoli G, Ohne Y, et al. IL‐33, IL‐25, and TSLP induce a distinct phenotypic and activation profile in human type 2 innate lymphoid cells. Blood Adv. 2017;1(10):577‐589.
    1. Gauvreau GM, O'Byrne PM, Boulet L‐P, et al. Effects of an anti‐TSLP antibody on allergen‐induced asthmatic responses. N Engl J Med. 2014;370(22):2102‐2110.
    1. Corren J, Parnes JR, Wang L, et al. Tezepelumab in adults with uncontrolled asthma. N Engl J Med. 2017;377(10):936‐946.
    1. Verstraete K, Peelman F, Braun H, et al. Structure and antagonism of the receptor complex mediated by human TSLP in allergy and asthma. Nat Commun. 2017;8:14937.
    1. 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.
    1. American Thoracic S, European RS. ATS/ERS recommendations for standardized procedures for the online and offline measurement of exhaled lower respiratory nitric oxide and nasal nitric oxide, 2005. Am J Respir Crit Care Med. 2005;171(8):912‐930.
    1. Castro M, Corren J, Pavord ID, et al. Dupilumab efficacy and safety in moderate‐to‐severe uncontrolled asthma. N Engl J Med. 2018;378(26):2486‐2496.
    1. Goldman M, Hirsch I, Zangrilli JG, Newbold P, Xu X. The association between blood eosinophil count and benralizumab efficacy for patients with severe, uncontrolled asthma: subanalyses of the phase III SIROCCO and CALIMA studies. Curr Med Res Opin. 2017;33(9):1605‐1613.
    1. Jeppegaard M, Veidal S, Sverrild A, Backer V, Porsbjerg C. Validation of ATS clinical practice guideline cut‐points for FeNO in asthma. Respir Med. 2018;144:22‐29.
    1. Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double‐blind, placebo‐controlled trial. Lancet. 2012;380(9842):651‐659.
    1. Soma T, Iemura H, Naito E, et al. Implication of fraction of exhaled nitric oxide and blood eosinophil count in severe asthma. Allergol Int. 2018;67S:S3‐S11.
    1. Cleveland W. Robust locally weighted regression and smoothing scatterplots. J Am Stat Assoc. 1979;74:829‐836.
    1. Janeway CA, Travers P, Walport M, Shlomchik M. Immunobiology: the immune system in health and disease, 5th Edition. Garland Science; 2001. The production of IgE. . Accessed July 19, 2020.
    1. Buhl R, Korn S, Menzies‐Gow A, et al. Prospective, single‐arm, longitudinal study of biomarkers in real‐world patients with severe asthma. J Allergy Clin Immunol Pract. 2020;8(8):2630‐2639 e2636.
    1. Truyen E, Coteur L, Dilissen E, et al. Evaluation of airway inflammation by quantitative Th1/Th2 cytokine mRNA measurement in sputum of asthma patients. Thorax. 2006;61(3):202‐208.
    1. Pham TH, Kearley J, Parnes JR, Leung L, Goleva E, Griffiths JM. Development of a highly sensitive assay to quantitate circulating thymic stromal lymphopoietin (TSLP) levels in blood. J Allergy Clin Immunol. 2020;145:AB30.
    1. Lee HC, Ziegler SF. Inducible expression of the proallergic cytokine thymic stromal lymphopoietin in airway epithelial cells is controlled by NFkappaB. Proc Natl Acad Sci USA. 2007;104(3):914‐919.
    1. Rochman Y, Dienger‐Stambaugh K, Richgels PK, et al. TSLP signaling in CD4(+) T cells programs a pathogenic T helper 2 cell state. Sci Signal. 2018;11(521):eaam8858.
    1. Omori M, Ziegler S. Induction of IL‐4 expression in CD4(+) T cells by thymic stromal lymphopoietin. J Immunol. 2007;178(3):1396‐1404.
    1. Schleich F, Demarche S, Louis R. Biomarkers in the management of difficult asthma. Curr Top Med Chem. 2016;16(14):1561‐1573.
    1. Dweik RA, Boggs PB, Erzurum SC, et al. An official ATS clinical practice guideline: interpretation of exhaled nitric oxide levels (FENO) for clinical applications. Am J Respir Crit Care Med. 2011;184(5):602‐615.
    1. Syed F, Huang CC, Li K, et al. Identification of interleukin‐13 related biomarkers using peripheral blood mononuclear cells. Biomarkers. 2007;12(4):414‐423.
    1. Li Y, Wang W, Lv Z, et al. 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(7):2253‐2262.
    1. Khaitov MR, Gaisina AR, Shilovskiy IP, et al. The role of interleukin‐33 in pathogenesis of bronchial asthma. New experimental data. Biochemistry (Mosc). 2018;83(1):13‐25.
    1. Bianchetti L, Marini MA, Isgro M, Bellini A, Schmidt M, Mattoli S. IL‐33 promotes the migration and proliferation of circulating fibrocytes from patients with allergen‐exacerbated asthma. Biochem Biophys Res Commun. 2012;426(1):116‐121.
    1. Beale J, Jayaraman A, Jackson DJ, et al. Rhinovirus‐induced IL‐25 in asthma exacerbation drives type 2 immunity and allergic pulmonary inflammation. Sci Transl Med. 2014;6(256):256ra134.
    1. Doran E, Cai F, Holweg CTJ, Wong K, Brumm J, Arron JR. Interleukin‐13 in asthma and other eosinophilic disorders. Front Med. 2017;4:139.
    1. Neelamegan R, Saka V, Tamilarasu K, Rajaram M, Selvarajan S, Chandrasekaran A. Clinical utility of fractional exhaled nitric oxide (FeNO) as a biomarker to predict severity of disease and response to inhaled corticosteroid (ICS) in asthma patients. J Clin Diagn Res. 2016;10(12):FC01‐FC06.
    1. Lommatzsch M, Klein M, Stoll P, Virchow JC. Impact of an increase in the inhaled corticosteroid dose on blood eosinophils in asthma. Thorax. 2019;74(4):417‐418.
    1. Diver S, Khalfaoui L, Emson C, et al. Effect of tezepelumab on airway inflammatory cells, remodelling, and hyperresponsiveness in patients with moderate‐to‐severe uncontrolled asthma (CASCADE): a double‐blind, randomised, placebo‐controlled, phase 2 trial. Lancet Respir Med. 2021;9(11):1299‐1312.
    1. Sverrild A, Hansen S, Hvidtfeldt M, et al. The effect of tezepelumab on airway hyperresponsiveness to mannitol in asthma (UPSTREAM). Eur Respir J. 2021:2101296.

Source: PubMed

3
Tilaa