An Inhaled PI3Kδ Inhibitor Improves Recovery in Acutely Exacerbating COPD Patients: A Randomized Trial

Anthony Cahn, J Nicole Hamblin, Jon Robertson, Malcolm Begg, Emily Jarvis, Robert Wilson, Gordon Dear, Claudia Leemereise, Yi Cui, Maki Mizuma, Mickael Montembault, Cedric Van Holsbeke, Wim Vos, Wilfried De Backer, Jan De Backer, Edith M Hessel, Anthony Cahn, J Nicole Hamblin, Jon Robertson, Malcolm Begg, Emily Jarvis, Robert Wilson, Gordon Dear, Claudia Leemereise, Yi Cui, Maki Mizuma, Mickael Montembault, Cedric Van Holsbeke, Wim Vos, Wilfried De Backer, Jan De Backer, Edith M Hessel

Abstract

Purpose: This study evaluated the safety and efficacy of inhaled nemiralisib, a phosphoinositide 3-kinase δ (PI3Kδ) inhibitor, in patients with an acute exacerbation of chronic obstructive pulmonary disease (COPD).

Methods: In this double-blind, placebo-controlled study, 126 patients (40-80 years with a post-bronchodilator forced expiratory volume in 1 sec (FEV1) ≤80% of predicted (previously documented)) were randomized 1:1 to once daily inhaled nemiralisib (1 mg) or placebo for 84 days, added to standard of care. The primary endpoint was specific imaging airway volume (siVaw) after 28 treatment days and was analyzed using a Bayesian repeated measures model (clintrials.gov: NCT02294734).

Results: A total of 126 patients were randomized to treatment; 55 on active treatment and 49 on placebo completed the study. When comparing nemiralisib and placebo-treated patients, an 18% placebo-corrected increase from baseline in distal siVaw (95% credible intervals (Cr I) (-1%, 42%)) was observed on Day 28. The probability that the true treatment ratio was >0% (Pr(θ>0)) was 96%, suggestive of a real treatment effect. Improvements were observed across all lung lobes. Patients treated with nemiralisib experienced a 107.3 mL improvement in posterior median FEV1 (change from baseline, 95% Cr I (-2.1, 215.5)) at day 84, compared with placebo. Adverse events were reported by 41 patients on placebo and 49 on nemiralisib, the most common being post-inhalation cough on nemiralisib (35%) vs placebo (3%).

Conclusion: These data show that addition of nemiralisib to usual care delivers more effective recovery from an acute exacerbation and improves lung function parameters including siVaw and FEV1. Although post-inhalation cough was identified, nemiralisib was otherwise well tolerated, providing a promising novel therapy for this acutely ill patient group.

Keywords: COPD; acute exacerbation; nemiralisib.

Conflict of interest statement

AC, JR, MB, GD, CL, YC, MMiz, MMont and EJ are GSK employees and all hold GSK shares. JNH, RW and EMH were GSK employees at the time of the study conduct, analysis and interpretation, and hold GSK shares. JNH and EMH are named on patents for compound GSK2269557 (nemiralisib). JNH reports patents WO2010125082A1 and WO2012055846A1 issued. EMH has a patent WO2015055691A1 issued. CVH, WV and JDB are employees of FLUIDDA, the company responsible for FRI analysis and interpretation. WV and JDB hold FLUIDDA shares. WDB reports costs from University Hospital Antwerp to perform the study. The current affiliation of JNH is Discovery, Charles River, Chesterford Research Park, Cambridge, UK. The current affiliation of RW is DLRC, Letchworth Garden City, UK. The current affiliation of WV is OncoRadiomics, Liège, Belgium. The current affiliation of EMH is Eligo Bioscience, Paris, France. The authors report no other conflicts of interest in this work.

© 2021 Cahn et al.

Figures

Figure 1
Figure 1
Study schematic (Panel A) and trial profile (Panel B).
Figure 2
Figure 2
Median % change from baseline in distal region siVaw at FRC after 28 days.
Figure 3
Figure 3
Placebo-corrected change in siVaw at FRC after 12 and 28 days of treatment with nemiralisib.
Figure 4
Figure 4
Change from baseline in FEV1 (Panel A) and sGaw (Panel B) in relation to the severity of the index exacerbation (FEV1 and sGaw measured at clinical visits at baseline, Day 28 and Day 84). Data presented as median ± 95% Cr I. Analyses of FEV1 data excludes data for one subject (281) due to erroneously large FEV1 recorded at baseline.
Figure 5
Figure 5
Number of patients (out of 63 per arm) with any new exacerbation eventsa during treatment and of the observed total number of severe exacerbationsb (28 in the placebo arm and 21 in the nemiralisib arm) the proportion requiring hospitalization. aNew exacerbation event based on observed treatment for an exacerbation; a new prescription for corticosteroids and/or antibiotics. bSevere exacerbations based on investigator-reported exacerbations resulting in hospitalization.

References

    1. From the Global Strategy for the Diagnosis, Management and Prevention of COPD. Global Initiative for Chronic Obstructive Lung Disease (GOLD) 2020. Available from: . Accessed May6, 2021.
    1. Wedzicha JA, Seemungal TAR. COPD exacerbations: defining their cause and prevention. Lancet. 2007;370(9589):786–796. doi:10.1016/S0140-6736(07)61382-8
    1. Bafadhel M, McKenna S, Terry S, et al. Acute exacerbations of chronic obstructive pulmonary disease: identification of biologic clusters and their biomarkers. Am J Respir Crit Care Med. 2011;184:662–671. doi:10.1164/rccm.201104-0597OC
    1. Miravitlles M, Ferrer M, Pont À, et al.; IMPAC Study Group. Exacerbations impair quality of life in patients with chronic obstructive pulmonary disease. A two-year follow-up study. Thorax. 2004;59:387–395. doi:10.1136/thx.2003.008730
    1. Niewoehner DE, Lokhnygina Y, Rice K, et al. Risk indexes for exacerbations and hospitalizations due to COPD. Chest. 2007;131:20–28. doi:10.1378/chest.06-1316
    1. Soler-Cataluña JJ, Martínez-García MA, Román Sánchez P, Salcedo E, Navarro M, Ochando R. Severe acute exacerbations and mortality in patients with chronic obstructive pulmonary disease. Thorax. 2005;60:925–931. doi:10.1136/thx.2005.040527
    1. Wilson R. Treatment of COPD exacerbations: antibiotics. Eur Respir Rev. 2005;14:32–38. doi:10.1183/09058180.05.00009404
    1. Hunter MH, King DE. COPD: management of acute exacerbations and chronic stable disease. Am Fam Physician. 2001;64:603–613.
    1. Seemungal TA, Donaldson GC, Bhowmik A, Jeffries DJ, Wedzicha JA. Time course and recovery of exacerbations in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2000;16:1608–1613. doi:10.1164/ajrccm.161.5.9908022
    1. Hurst JR, Donaldson GC, Quint JK, Goldring JJ, Baghai-Ravary R, Wedzicha J. Temporal clustering of exacerbations in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;179:369–374. doi:10.1164/rccm.200807-1067OC
    1. Suissa S, Dell’Aniello S, Ernst P. Long-term natural history of chronic obstructive pulmonary disease: severe exacerbations and mortality. Thorax. 2012;67:957–963. doi:10.1136/thoraxjnl-2011-201518
    1. Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K pathway in human disease. Cell. 2017;170:605–635.
    1. Down K, Amour A, Baldwin IR, et al. Optimization of novel indazoles as highly potent and selective inhibitors of phosphoinositide 3-kinase δ for the treatment of respiratory disease. J Med Chem. 2015;58(18):7381–7399. doi:10.1021/acs.jmedchem.5b00767
    1. Cahn A, Hamblin JN, Begg M, et al. Safety, pharmacokinetics and dose-response characteristics of GSK2269557, an inhaled PI3Kδ inhibitor under development for the treatment of COPD. Pulm Pharmacol Ther. 2017;46:69–77. doi:10.1016/j.pupt.2017.08.008
    1. Milara J, Lluch J, Almudever P, Freire J, Xiaozhong Q, Cortijo J. Roflumilast N-oxide reverses corticosteroid resistance in neutrophils from patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2014;134(2):314–322. doi:10.1016/j.jaci.2014.02.001
    1. To Y, Ito K, Kizawa Y, et al. Targeting phosphoinositide-3-kinase-delta with theophylline reverses corticosteroid insensitivity in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;182:897–904. doi:10.1164/rccm.200906-0937OC
    1. Angulo I, Vadas O, Garçon F, et al. Phosphoinositide 3-kinase gene mutation predisposes to respiratory infection and airway damage. Science. 2013;342(6160):866–871. doi:10.1126/science.1243292
    1. Stark AK, Chandra A, Chakraborty K, et al. PI3Kδ hyper-activation promotes development of B cells that exacerbate Streptococcus pneumoniae infection in an antibody-independent manner. Nat Commun. 2018;9:3174. doi:10.1038/s41467-018-05674-8
    1. Lucas CL, Kuehn HS, Zhao F, et al. Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110δ result in T cell senescence and human immunodeficiency. Nat Immunol. 2014;15(1):88–97. doi:10.1038/ni.2771
    1. Sapey E, Stockley JA, Greenwood H, et al. Behavioral and structural differences in migrating peripheral neutrophils from patients with chronic obstructive pulmonary disease. AM J Respir Crit Care Med. 2011;183:1176–1186. doi:10.1164/rccm.201008-1285OC
    1. Stark A-K, Sriskantharajah S, Hessel EM, Okkenhaug K. PI3K inhibitors in inflammation, autoimmunity and cancer. Curr Opin Pharmacol. 2015;23:82–91.
    1. Rangelov BA, Young AL, Jacob J, et al. Thoracic imaging at exacerbation of chronic obstructive pulmonary disease: a systematic review. Int J Chron Obstruct Pulmon Dis. 2020;15:1751–1787. doi:10.2147/COPD.S250746
    1. Vos W, Van Holsbeke C, Van Geffen W, et al. Changes in functional respiratory imaging (FRI) endpoints correlate with changes in patient reported outcomes (PRO) after recovering from acute COPD exacerbation. Eur Respir J. 2015;46(suppl 59):OA2948.
    1. Vos W, Van Holsbeke C, De Backer J, De Backer WA. Correlation between changes in FEV1 and functional respiratory imaging biomarkers in a large population of asthma and COPD patients. Am J Respir Crit Care Med. 2015;191:A2266.
    1. De Backer LA, Vos WG, Salgado R, et al. Functional imaging using computer methods to compare the effect of salbutamol and ipratropium bromide in patient-specific airway models of COPD. Int J Chron Obstruct Pulmon Dis. 2011;6:637–646. doi:10.2147/COPD.S21917
    1. Lanclus M, Clukers J, Van Holsbeke C, et al. Machine learning algorithms utilizing functional respiratory imaging may predict COPD exacerbations. Acad Radiol. 2019;26:1191–1199. doi:10.1016/j.acra.2018.10.022
    1. Quanjer PH, Tammeling GJ, Cotes JE, Pedersen OF, Peslin R, Yernault JC. Lung volumes and forced ventilatory flows. Report working party standardization of lung function tests, European Community for steel and coal. Official Statement of the European Respiratory Society. Eur Respir J Suppl. 1993;6(suppl 16):5–40. doi:10.1183/09041950.005s1693
    1. Jones PW, Harding G, Berry P, Wiklund I, Chen W-H, Kline Leidy N. Development and first validation of the COPD assessment test. Eur Respir J. 2009;34:648–654. doi:10.1183/09031936.00102509
    1. Bestall JC, Paul EA, Garrod R, Garnham R, Jones PW, Wedzicha JA. Usefulness of the Medical Research Council (MRC) dyspnoea scale as a measure of disability in patients with chronic obstructive pulmonary disease. Thorax. 1999;54:581–586. doi:10.1136/thx.54.7.581
    1. van Geffen WH, Hajian B, Vos W, et al. Functional respiratory imaging: heterogeneity of acute exacerbations of COPD. Int J Chron Obstruct Pulmon Dis. 2018;13:1783–1792. doi:10.2147/COPD.S152463
    1. Barbosa E, Ferreira F, Vos W, et al. Quantitative functional respiratory imaging may predict the mechanism of FEV1 decline after lung transplantation. Eur Respir J. 2017;50:PA2465.
    1. Vos W, Janssens A, Van Holsbeke C, et al. Functional respiratory imaging to predict post-operative FEV1 after lobectomy/pneumonectomy. Eur Respir J. 2014;44:P342. doi:10.1183/09031936.00011714
    1. Lins M, Vandevenne J, Thillai M, et al. Assessment of small pulmonary blood vessels in COVID-19 patients using HRCT. Acad Radiol. 2020;27:1449–1455. doi:10.1016/j.acra.2020.07.019
    1. Thillai M, Patvardhan C, Swietlik EM, et al. Functional respiratory imaging identifies redistribution of pulmonary blood flow in patients with COVID-19. Thorax. 2020. doi:10.1136/thoraxjnl-2020-215395
    1. Watz H, Tetzlaff K, Magnussen H, et al. Spirometric changes during exacerbations of COPD: a post hoc analysis of the WISDOM trial. Respir Res. 2018;19:251–266. doi:10.1186/s12931-018-0944-3
    1. Coutré SE, Barrientos JC, Brown JR, et al. Management of adverse events associated with idelalisib treatment: expert panel opinion. Leuk Lymphoma. 2015;56:2779–2786. doi:10.3109/10428194.2015.1022770
    1. Sánchez-Alegría K, Flores-León M, Avila-Muñoz E, Rodríguez-Corona N, Arias C. PI3K Signaling in neurons: a central node for the control of multiple functions. Int J Mol Sci. 2018;19:3725. doi:10.3390/ijms19123725
    1. Wilson R, Cahn A, Montembault M, et al. Safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of single (SD) and repeat (RD) inhaled doses of a novel phosphoinositide 3-kinase δ inhibitor (PI3Kδ), GSK2269557, administered to healthy smokers. Eur Respir J. 2014;44(Suppl 58):3411.

Source: PubMed

3
Suscribir