Exploring PI3Kδ Molecular Pathways in Stable COPD and Following an Acute Exacerbation, Two Randomized Controlled Trials

Malcolm Begg, J Nicole Hamblin, Emily Jarvis, Glyn Bradley, Stephen Mark, David Michalovich, Mark Lennon, Hannah E Wajdner, Augustin Amour, Robert Wilson, Ken Saunders, Rikako Tanaka, Saki Arai, Teresa Tang, Cedric Van Holsbeke, Jan De Backer, Wim Vos, Ingrid L Titlestad, J Mark FitzGerald, Kieran Killian, Jean Bourbeau, Claude Poirier, François Maltais, Anthony Cahn, Edith M Hessel, Malcolm Begg, J Nicole Hamblin, Emily Jarvis, Glyn Bradley, Stephen Mark, David Michalovich, Mark Lennon, Hannah E Wajdner, Augustin Amour, Robert Wilson, Ken Saunders, Rikako Tanaka, Saki Arai, Teresa Tang, Cedric Van Holsbeke, Jan De Backer, Wim Vos, Ingrid L Titlestad, J Mark FitzGerald, Kieran Killian, Jean Bourbeau, Claude Poirier, François Maltais, Anthony Cahn, Edith M Hessel

Abstract

Background: Inhibition of phosphoinositide 3-kinase δ (PI3Kδ) exerts corrective effects on the dysregulated migration characteristics of neutrophils isolated from patients with chronic obstructive pulmonary disease (COPD).

Objective: To develop novel, induced sputum endpoints to demonstrate changes in neutrophil phenotype in the lung by administering nemiralisib, a potent and selective inhaled PI3Kδ inhibitor, to patients with stable COPD or patients with acute exacerbation (AE) of COPD.

Methods: In two randomized, double-blind, placebo-controlled clinical trials patients with A) stable COPD (N=28, randomized 3:1) or B) AECOPD (N=44, randomized 1:1) received treatment with inhaled nemiralisib (1mg). Endpoints included induced sputum at various time points before and during treatment for the measurement of transcriptomics (primary endpoint), inflammatory mediators, functional respiratory imaging (FRI), and spirometry.

Results: In stable COPD patients, the use of nemiralisib was associated with alterations in sputum neutrophil transcriptomics suggestive of an improvement in migration phenotype; however, the same nemiralisib-evoked effects were not observed in AECOPD. Inhibition of sputum inflammatory mediators was also observed in stable but not AECOPD patients. In contrast, a placebo-corrected improvement in forced expiratory volume in 1 sec of 136 mL (95% Credible Intervals -46, 315mL) with a probability that the true treatment ratio was >0% (Pr(θ>0)) of 93% was observed in AECOPD. However, FRI endpoints remained unchanged.

Conclusion: We provide evidence for nemiralisib-evoked changes in neutrophil migration phenotype in stable COPD but not AECOPD, despite improving lung function in the latter group. We conclude that induced sputum can be used for measuring evidence of alteration of neutrophil phenotype in stable patients, and our study provides a data set of the sputum transcriptomic changes during recovery from AECOPD.

Keywords: COPD exacerbations; PI3Kdelta; nemiralisib; sputum; transcriptomics.

Conflict of interest statement

MB, EJ, GB, SM, DM, ML, HW, AA, KS, RT, SA, TT and AC are employees and shareholders of GSK. JNH, RW & EMH are former employees and shareholders of GSK. JNH, EMH and AA are named on patents related to compound GSK2269557 (nemiralisib). JNH has patents WO 2010125082 and WO 2012055846 issued. EMH has a patent WO2015055691A1 issued. AA has patents US20160263109, US20160256466, EP3057588, EP3057587, WO2015055691, and WO2015055690 issued to GSK. CVH and JDB are employees of FLUIDDA, the company responsible for FRI analysis and interpretation. WV is a former employee of FLUIDDA. WV and JDB hold FLUIDDA shares. JMF has received research funding paid directly to the University of British Columbia from AstraZeneca, GSK, Novartis, Sanofi-Regeneron, Canadian Institutes of Health Research (CIHR), NIH and AllerGen; and has received honoraria from AstraZeneca, GSK, TEVA, Sanofi for participation in Advisory Boards and speaker bureaus. JB has received grants from CIHR, Canadian Respiratory Research Network, Foundation of the McGill University Health Centre (MUHC), Aerocrine, AstraZeneca, GSK, Boehringer Ingelheim, Novartis, Grifols and Trudell; and personal fees from Canadian Thoracic Society, CHEST, AstraZeneca, GSK, Boehringer Ingelheim, Novartis, Grifols, Pfizer, and Trudell for consultancy and lectures. CP has received personal fees from GSK, Boehringer Ingelheim, Novartis, AstraZeneca and Apnee Sante. FM has received grants paid to his institution from AstraZeneca, GSK, Boehringer Ingelheim, Sanofi, Novartis and Grifols; has received personal fees from GSK, Boehringer Ingelheim, Grifols, and Novartis for serving on speaker bureaus; and is financially involved with Oxynov, a company developing an oxygen delivery system. The current affiliation of JNH is Discovery, Charles River, Chesterford Research Park, Cambridge, UK. The current affiliation of DM is Benevolent AI Ltd, London, United Kingdom. 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 Begg et al.

Figures

Figure 1
Figure 1
mRNA transcriptomic alterations in induced sputum from stable COPD patients (Study A). Transcriptomic changes in induced sputum taken from stable COPD patients dosed for 14 days with nemiralisib show a partial clustering by treatment (Panel A). Unbiased pathway analysis highlighted nemiralisib-evoked alterations in neutrophil phenotype and bacterial infection response genes (Panels B and C, left), which were not observed in the samples taken 7 days apart prior to randomization (Panels B and C, right).
Figure 2
Figure 2
mRNA transcriptomic alterations in induced sputum from AECOPD patients (Study B). Transcriptomic changes in induced sputum taken from acutely exacerbating COPD patients. Of the predefined neutrophil-related genes a small number were altered in both the nemiralisib and placebo-treated groups (Panel A). The entire unbiased gene array output also showed a similar number of genes changing in the nemiralisib and placebo-treated groups when comparing both day 12 or day 84 to the screening time point (Panel A, SCN v D12). There was a significant overlap in the genes changing between screening and day 12 in the nemiralisib and placebo-treated groups at day 12 and day 84, with almost all genes changing in the same direction (Panel B).
Figure 3
Figure 3
Inflammatory biomarkers in induced sputum from AECOPD patients (Study B) Sputum and circulating biomarkers measured were also not impacted by treatment with nemiralisib. Circulating levels of CRP decreased (Panel A, left), as did neutrophil count (Panel A, right), presented as mean (95% CI) at baseline, Day 12, Day 28, Day 56, Day 84 and at follow-up. Total sputum cell count and the inflammatory cytokines IL-8, IL-6 and TNFα were also not inhibited following treatment with nemiralisib (Panel B), presented as geometric mean (95% CI) at baseline, Day 12, Day 28 and Day 84.
Figure 4
Figure 4
Summary of statistical analysis of FEV1 and FVC in AECOPD patients (Study B). Treatment with nemiralisib showed an improvement in FEV1 and FVC at all time points. Panel A shows adjusted median change from baseline in FEV1 (95% Cr I), and Panel B shows adjusted median change from baseline in FVC (95% Cr I) measured at clinical visits at baseline, Day 12, Day 28, Day 56 and Day 84. Data presented as adjusted median ± 95% Credible Intervals.
Figure 5
Figure 5
Lung transcriptomics during exacerbation recovery (Study B). Combined sputum transcriptomic data shows partial clustering by time point (Panels A and B). Unbiased overlay analysis showed a highly significant overlap with data from ECLIPSE ., The degree of overlap and gene directionality for the matching genes are displayed in (Panel C).
Figure 6
Figure 6
Blood transcriptomics during exacerbation recovery (Study B). Combined blood transcriptomic data shows partial clustering by time point (Panels A and B). Unbiased overlay analysis showed a highly significant overlap with data from ECLIPSE.27,30 The degree of overlap and gene directionality for the matching genes are displayed in (Panel C).

References

    1. Dy R, Sethi S. The lung microbiome and exacerbations of COPD. Curr Opin Pulm Med. 2016;22:196–202. doi:10.1097/MCP.0000000000000268
    1. Ritchie AI, Wedzicha JA. Definition, causes, pathogenesis, and consequences of chronic obstructive pulmonary disease exacerbations. Clin Chest Med. 2020;41:421–438. doi:10.1016/j.ccm.2020.06.007
    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. Wilkinson TMA, Aris E, Bourne SC, et al. Drivers of year-to-year variation in exacerbation frequency of COPD: analysis of the AERIS cohort. ERJ Open Res. 2019;5:00248–2018. doi:10.1183/23120541.00248-2018
    1. AERIS Study Group; Wilkinson TMA, Aris E, Bourne S, et al. A prospective, observational cohort study of the seasonal dynamics of airway pathogens in the aetiology of exacerbations in COPD. Thorax. 2017;72:919–927. doi:10.1136/thoraxjnl-2016-209023
    1. Liu J, Pang Z, Wang G, et al. Advanced role of neutrophils in common respiratory diseases. J Immunol Res. 2017;2017:6710278. doi:10.1155/2017/6710278
    1. Sapey M, Rijkers GT, van Overveld FJ. Neutrophils and emerging targets for treatment in chronic obstructive pulmonary disease. Expert Rev Clin Immunol. 2013;9:1055–1068. doi:10.1586/1744666X.2013.851347
    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. 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;134:314–322. 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. Stark A-K, Sriskantharajah S, Hessel EM, Okkenhaug K. PI3K inhibitors in inflammation, autoimmunity and cancer. Curr Opin Pharmacol. 2015;2382–2391.
    1. Gupta V, Singh D. Critical assessment of the value of sputum neutrophils. COPD. 2013;10:107–114. doi:10.3109/15412555.2013.766105
    1. Tregay N, Begg M, Cahn A, et al. Use of autologous 99mTechnetium-labelled neutrophils to quantify lung neutrophil clearance in COPD. Thorax. 2019;74:659–666. doi:10.1136/thoraxjnl-2018-212509
    1. Sims P, Coffman RL, Hessel EM. Biomarkers measuring the activity of Toll-like receptor ligands in clinical development programs. Methods Mol Biol. 2009;517:415–440.
    1. Gauvreau GM, Hessel EM, Boulet LP, Coffman RL, O’Byrne PM. Immunostimulatory sequences regulate interferon-inducible genes but not allergic airway responses. Am J Respir Crit Care Med. 2006;174:15–20. doi:10.1164/rccm.200601-057OC
    1. Govoni M, Bassi M, Vezzoli S, et al. 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. doi:10.1186/s12931-020-1329-y
    1. Begg M, Wilson R, Hamblin JN, et al. Relationship between pharmacokinetics and pharmacodynamic responses in healthy smokers informs a once-daily dosing regimen for nemiralisib. J Pharmacol Exp Ther. 2019;369:337–344. doi:10.1124/jpet.118.255109
    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. 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. Global Initiative for Chronic Obstructive Lung Disease (GOLD). From the global strategy for the diagnosis, management and prevention of COPD; 2020. Available from: . Accessed May6, 2021.
    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. 1993;6(suppl 16):5–40. doi:10.1183/09041950.005s1693
    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;161:1608–1613. doi:10.1164/ajrccm.161.5.9908022
    1. Juss JK, House D, Amour A, et al. Acute respiratory distress syndrome neutrophils have a distinct phenotype and are resistant to phosphoinositide 3-kinase inhibition. Am J Respir Crit Care Med. 2016;194(8):961–973. doi:10.1164/rccm.201509-1818OC
    1. Vlahos R, Wark PA, Anderson GP, Bozinovski S. Glucocorticosteroids differentially regulate MMP-9 and neutrophil elastase in COPD. PLoS One. 2012;7:e33277. doi:10.1371/journal.pone.0033277
    1. Cahn A, Hamblin JN, Robertson J, et al. An inhaled PI3δ inhibitor improves recovery in acutely exacerbating COPD patients, a randomized trial. Int J Chron Obstruct Pulmon Dis. In press 2021.
    1. Butler CC, Gillespie D, White P, et al. C-reactive protein testing to guide antibiotic prescribing for COPD exacerbations. N Engl J Med. 2019;381:111–120. doi:10.1056/NEJMoa1803185
    1. ECLIPSE investigators; Vestbo J, Anderson W, Coxson HO, et al. Evaluation of COPD longitudinally to identify predictive surrogate end-points (ECLIPSE). Eur Respir J. 2008;31:869–873. doi:10.1183/09031936.00111707
    1. Reinhold D, Morrow JD, Jacobson S, et al. Meta-analysis of peripheral blood gene expression modules for COPD phenotypes. PLoS One. 2017;12:e0185682. doi:10.1371/journal.pone.0185682
    1. ECLIPSE Investigators; Singh D, Fox SM, Tal-Singer R, et al. Induced sputum genes associated with spirometric and radiological disease severity in COPD ex-smokers. Thorax. 2011;66(6):489–495. doi:10.1136/thx.2010.153767.
    1. Gupta V, Khan A, Higham A, et al. The effect of phosphatidylinositol-3 kinase inhibition on matrix metalloproteinase-9 and reactive oxygen species release from chronic obstructive pulmonary disease neutrophils. Int Immunopharmacol. 2016;35:155–162. doi:10.1016/j.intimp.2016.03.027
    1. Pavord ID, Jones PW, Burgel PR, Rabe KF. Exacerbations of COPD. Int J Chron Obstruct Pulmon Dis. 2016;11(speciss):21–30. doi:10.2147/COPD.S85978
    1. Singh D, Fox SM, Tal-Singer R, Bates S, Riley JH, Celli B. Altered gene expression in blood and sputum in COPD frequent exacerbators in the ECLIPSE cohort. PLoS One. 2014;9(9):e107381. doi:10.1371/journal.pone.0107381
    1. COPDMAP; Michaeloudes C, Kuo CH, Haji G, et al. Metabolic re-patterning in COPD airway smooth muscle cells. Eur Respir J. 2017;50:1700202. doi:10.1183/13993003.00202-2017
    1. Ryter SW, Rosas IO, Owen CA, et al. Mitochondrial dysfunction as a pathogenic mediator of chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Ann Am Thorac Soc. 2018;15:S266–S272. doi:10.1513/AnnalsATS.201808-585MG
    1. Cosio MG, Saetta M, Agusti A. Immunologic aspects of chronic obstructive pulmonary disease. N Engl J Med. 2009;360:2445–2454. doi:10.1056/NEJMra0804752
    1. Remels AH, Gosker HR, Schrauwen P, Langen RC, Schols AM. Peroxisome proliferator-activated receptors: a therapeutic target in COPD? Eur Respir J. 2008;31:502–508. doi:10.1183/09031936.00068207
    1. Wan ES, Silverman EK. Genetics of COPD and emphysema. Chest. 2009;136:859–866. doi:10.1378/chest.09-0555
    1. Stolk J, Stockley RA, Stoel BC, et al. Randomised controlled trial for emphysema with a selective agonist of the γ-type retinoic acid receptor. Eur Respir J. 2012;40:306–312. doi:10.1183/09031936.00161911
    1. Higham A, Lea S, Plumb J, et al. The role of the liver X receptor in chronic obstructive pulmonary disease. Respir Res. 2013;14:106. doi:10.1186/1465-9921-14-106
    1. Criner RN, Han MK. COPD care in the 21st century: a public health priority. Respir Care. 2018;63:591–600. doi:10.4187/respcare.06276

Source: PubMed

3
Abonneren