Sputum and blood transcriptomics characterisation of the inhaled PDE4 inhibitor CHF6001 on top of triple therapy in patients with chronic bronchitis

Mirco Govoni, Michele Bassi, Stefano Vezzoli, Germano Lucci, Aida Emirova, Marie Anna Nandeuil, Stefano Petruzzelli, Gera L Jellema, Ebenezer K Afolabi, Brendan Colgan, Brian Leaker, Oliver Kornmann, Kai Michael Beeh, Henrik Watz, Dave Singh, Mirco Govoni, Michele Bassi, Stefano Vezzoli, Germano Lucci, Aida Emirova, Marie Anna Nandeuil, Stefano Petruzzelli, Gera L Jellema, Ebenezer K Afolabi, Brendan Colgan, Brian Leaker, Oliver Kornmann, Kai Michael Beeh, Henrik Watz, Dave Singh

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.

Keywords: Biomarkers; Chronic obstructive pulmonary disease; Gene expression; Inflammation; Phosphodiesterase 4 inhibitors.

Conflict of interest statement

MG, MB, SV, GL, AE, MAN and SP are employees of Chiesi, the sponsor of this study.

GLJ and EKA are employees of the Almac group; Almac received funding from Chiesi as part of a consultation fee for the work reported in this manuscript.

BC and BL have nothing to disclose.

OK’s institution received fees from Chiesi for conducting this study as a participating site. He reports personal fees as speaker or Advisory Board member from AstraZeneca, Boehringer Ingelheim, GlaxoSmithKline, Sanofi and Novartis.

KMB declares that no personal payments were received from any pharmaceutical entity in the past 5 yrs. He is a full time employee of insaf Respiratory Research Institute. The institution has received compensation for services on advisory boards or consulting for Ablynx, Almirall, AstraZeneca, Berlin Chemie, Boehringer, Chiesi, Cytos, Mundipharma, Novartis, Pohl Boskamp, Zentiva. The institution has received compensation for speaker activities in scientific meetings supported by Almirall, AstraZeneca, Berlin Chemie, Boehringer, Cytos, ERT, GSK, Novartis, Pfizer, Pohl Boskamp, Takeda. The institution has received compensation for design and performance of clinical trials from Almirall, Altana/Nycomed, AstraZeneca, Boehringer, Cytos, GSK, Infinity, Medapharma, MSD, Mundipharma, Novartis, Parexel, Pearl Therapeutics, Pfizer, Revotar, Teva, Sterna, and Zentiva.

HW reports personal fees from Chiesi during the conduct of the study. Outside the submitted work, Dr. Watz reports personal fees from Bayer, personal fees from GSK, personal fees from Boehringer Ingelheim, personal fees from Novartis, personal fees from AstraZeneca, personal fees from BerlinChemie, and personal fees from Roche.

DS received personal fees from Chiesi during the conduct of this study. Outside the submitted work, he reports grants and personal fees from AstraZeneca, Boehringer Ingelheim, Chiesi, GlaxoSmithKline, Glenmark, Menarini, Mundipharma, Novartis, Pfizer, Pulmatrix, Therevance, and Verona, and personal fees from Cipla, Genentech and Peptinnovate.

Figures

Fig. 1
Fig. 1
Differentially expressed probe sets for CHF6001 800 μg and 1600 μg BID relative to placebo in (A) blood and (B) sputum. Volcano plot depicting all detected probe sets and coloured by fold change (FC) and adjusted p-values: orange, |FC| > 1.3; red, pFDR< 0.05; green, |FC| > 1.3 and pFDR< 0.05. Abbreviation: BID, twice daily
Fig. 2
Fig. 2
Log2 fold change with CHF6001 1600 μg BID vs. log2 fold change with CHF6001 800 μg BID. Equality line in red, fitted loess curve in blue. Only differentially expressed probe sets (pFDR

Fig. 3

Network of pro-inflammatory cytokines and…

Fig. 3

Network of pro-inflammatory cytokines and matrix metalloproteinases differentially expressed after treatment with CHF6001…

Fig. 3
Network of pro-inflammatory cytokines and matrix metalloproteinases differentially expressed after treatment with CHF6001 A) 800 μg and B) 1600 μg BID relative to placebo. Each node represents all the proteins produced by a single, protein-coding gene locus, edges represent proteins that jointly contribute to a shared function, and the information inside the circle describes protein structure. Edges: a red line indicates the presence of fusion evidence; a green line, neighbourhood evidence; a blue line, co-occurrence evidence; a magenta line, experimental evidence; a yellow line, text mining evidence; a light blue line, database evidence; a black line, co-expression evidence; a purple line, protein homology evidence. A green halo around the nodes: downregulation, a red halo: upregulation. Unmarked nodes: pFDRp < 0.05 and |FC| > 1.3

Fig. 4

Cytokines and inflammation response from…

Fig. 4

Cytokines and inflammation response from WikiPathways. Downregulated genes are coloured in different shades…

Fig. 4
Cytokines and inflammation response from WikiPathways. Downregulated genes are coloured in different shades of green

Fig. 5

Key pathophysiological pathways significantly modulated…

Fig. 5

Key pathophysiological pathways significantly modulated by CHF6001 in sputum with associated inhibition of…

Fig. 5
Key pathophysiological pathways significantly modulated by CHF6001 in sputum with associated inhibition of downstream inflammatory effectors. Created with Biorender.com
Fig. 3
Fig. 3
Network of pro-inflammatory cytokines and matrix metalloproteinases differentially expressed after treatment with CHF6001 A) 800 μg and B) 1600 μg BID relative to placebo. Each node represents all the proteins produced by a single, protein-coding gene locus, edges represent proteins that jointly contribute to a shared function, and the information inside the circle describes protein structure. Edges: a red line indicates the presence of fusion evidence; a green line, neighbourhood evidence; a blue line, co-occurrence evidence; a magenta line, experimental evidence; a yellow line, text mining evidence; a light blue line, database evidence; a black line, co-expression evidence; a purple line, protein homology evidence. A green halo around the nodes: downregulation, a red halo: upregulation. Unmarked nodes: pFDRp < 0.05 and |FC| > 1.3
Fig. 4
Fig. 4
Cytokines and inflammation response from WikiPathways. Downregulated genes are coloured in different shades of green
Fig. 5
Fig. 5
Key pathophysiological pathways significantly modulated by CHF6001 in sputum with associated inhibition of downstream inflammatory effectors. Created with Biorender.com

References

    1. Rennard SI, Calverley PMA, Goehring UM, Bredenbröker D, Martinez FJ. Reduction of exacerbations by the PDE4 inhibitor roflumilast - the importance of defining different subsets of patients with COPD. Respir Res. 2011;12:18. doi: 10.1186/1465-9921-12-18.
    1. Martinez FJ, Rabe KF, Sethi S, Pizzichini E, McIvor A, Anzueto A, et al. Effect of roflumilast and inhaled corticosteroid/long-acting β2-agonist on chronic obstructive pulmonary disease exacerbations (RE2SPOND). A randomized clinical trial. Am. J. Respir. Crit. Care Med. 2016;194:559–567. doi: 10.1164/rccm.201607-1349OC.
    1. Martinez FJ, Calverley PMA, Goehring U-M, Brose M, Fabbri LM, Rabe KF. Effect of roflumilast on exacerbations in patients with severe chronic obstructive pulmonary disease uncontrolled by combination therapy (REACT): a multicentre randomised controlled trial. Lancet Elsevier. 2015;385:857–866. doi: 10.1016/S0140-6736(14)62410-7.
    1. Gupta S. Side-effects of roflumilast. Lancet. 2012;379:710–711. doi: 10.1016/S0140-6736(12)60304-3.
    1. Kardos P, Mokros I, Sauer R, Vogelmeier CF. Health status in patients with COPD treated with roflumilast: two large noninterventional real-life studies: DINO and DACOTA. Int. J. Chron. Obstruct. Pulmon. Dis. 2018;13:1455–1468. doi: 10.2147/COPD.S159827.
    1. Mariotti F, Govoni M, Lucci G, Santoro D, Nandeuil MA. Safety, tolerability, and pharmacokinetics of single and repeat ascending doses of CHF6001, a novel inhaled phosphodiesterase-4 inhibitor: two randomized trials in healthy volunteers. Int. J. Chron. Obstruct. Pulmon. Dis. Dove Press. 2018;13:3399–3410. doi: 10.2147/COPD.S174156.
    1. Cenacchi V, Battaglia R, Cinato F, Riccardi B, Spinabelli D, Brogin G, et al. In vitro and in vivo metabolism of CHF 6001, a selective phosphodiesterase (PDE4) inhibitor. Xenobiotica. 2015;45:693–710. doi: 10.3109/00498254.2015.1014945.
    1. Villetti G, Carnini C, Battipaglia L, Preynat L, Bolzoni PT, Bassani F, et al. CHF6001 II: a novel phosphodiesterase 4 inhibitor, suitable for topical pulmonary administration - in vivo preclinical pharmacology profile defines a potent anti-inflammatory compound with a wide therapeutic window. J Pharmacol Exp Ther. 2015;352:568–578. doi: 10.1124/jpet.114.220558.
    1. Singh D, Nandeuil MA, Pigeon-Francisco C, Emirova A, Santoro D, Biondaro S, et al. Efficacy and safety of CHF6001, a novel inhaled PDE4 inhibitor in COPD: the PIONEER dose finding study. Am J Respir Crit Care Med. 2019;199:A4529. doi: 10.1164/rccm.201812-2345LE.
    1. Singh D, Beeh KM, Colgan B, Kornmann O, Leaker B, Watz H, et al. Effect of the inhaled PDE4 inhibitor CHF6001 on biomarkers of inflammation in COPD. Respir Res. 2019;20:180. doi: 10.1186/s12931-019-1142-7.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300. doi: 10.2307/2346101.
    1. Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. STRING v10: protein–protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43:D447–D452. doi: 10.1093/nar/gku1003.
    1. Ezkurdia I, Juan D, Rodriguez JM, Frankish A, Diekhans M, Harrow J, et al. Multiple evidence strands suggest that there may be as few as 19,000 human protein-coding genes. Hum Mol Genet. 2014;23:5866–5878. doi: 10.1093/hmg/ddu309.
    1. Slenter DN, Kutmon M, Hanspers K, Riutta A, Windsor J, Nunes N, et al. WikiPathways: a multifaceted pathway database bridging metabolomics to other omics research. Nucleic Acids Res. 2018;46:D661–D667. doi: 10.1093/nar/gkx1064.
    1. Barnes PJ. The cytokine network in asthma and chronic obstructive pulmonary disease. J Clin Invest. 2008;118:3546–3556. doi: 10.1172/JCI36130.
    1. Arulkumaran N, Unwin RJ, Tam FW. A potential therapeutic role for P2X7 receptor (P2X7R) antagonists in the treatment of inflammatory diseases. Expert Opin Investig Drugs. 2011;20:897–915. doi: 10.1517/13543784.2011.578068.
    1. Fagan KA, McMurtry IF, Rodman DM. Role of endothelin-1 in lung disease. Respir Res. 2001;2:90–101. doi: 10.1186/RR44.
    1. Roland M, Bhowmik A, Sapsford RJ, Seemungal TA, Jeffries DJ, Warner TD, et al. Sputum and plasma endothelin-1 levels in exacerbations of chronic obstructive pulmonary disease. Thorax. 2001;56:30–35. doi: 10.1136/THORAX.56.1.30.
    1. Westwood J-P, Mackay AJ, Donaldson G, Machin SJ, Wedzicha JA, Scully M. The role of complement activation in COPD exacerbation recovery. ERJ Open Research. 2016;2:00027–02016. doi: 10.1183/23120541.00027-2016.
    1. Barnes PJ, Drazen JM, Rennard SI, Thomson NC, editors. Asthma and COPD: Basic mechanisms and clinical management. 2nd ed. [Oxford, UK]: Academic Press; 2008.
    1. Liu R-M. Oxidative stress, plasminogen activator inhibitor 1, and lung fibrosis. Antioxid Redox Signal. 2008;10:303–319. doi: 10.1089/ars.2007.1903.
    1. Springer J, Scholz FR, Peiser C, Dinh QT, Fischer A, Quarcoo D, et al. Transcriptional down-regulation of suppressor of cytokine signaling (SOCS)-3 in chronic obstructive pulmonary disease. J Occup Med Toxicol. 2013;8:29. doi: 10.1186/1745-6673-8-29.
    1. UniProt Consortium. UniProt data [Internet]. [cited 2019 Mar 5]. Available from: .
    1. Prabhu AV, Luu W, Li D, Sharpe LJ, Brown AJ. DHCR7: a vital enzyme switch between cholesterol and vitamin D production. Prog. Lipid Res. 2016;64:138–151. doi: 10.1016/j.plipres.2016.09.003.
    1. Sundar IK, Hwang J-W, Wu S, Sun J, Rahman I. Deletion of vitamin D receptor leads to premature emphysema/COPD by increased matrix metalloproteinases and lymphoid aggregates formation. Biochem Biophys Res Commun. 2011;406:127–133. doi: 10.1016/j.bbrc.2011.02.011.
    1. Ishii M, Yamaguchi Y, Nakamura T, Akishita M. The vitamin D receptors may function as antiinflammatory effects in patients with COPD. Chest. 2015;148:690. doi: 10.1378/chest.2249480.
    1. García de Tena J, El Hachem Debek A, Hernández Gutiérrez C, Izquierdo Alonso JL. The role of vitamin D in chronic obstructive pulmonary disease, asthma and other respiratory diseases. Arch. Bronconeumol. 2014;50:179–184. doi: 10.1016/j.arbr.2014.03.015.
    1. Gerritsen WBM, Asin J, Zanen P, Bosch JMM va. den, Haas FJLM. Markers of inflammation and oxidative stress in exacerbated chronic obstructive pulmonary disease patients. Respir. Med. 2005;99:84–90. doi:10.1016/J.RMED.2004.04.017.
    1. Domej W, Oettl K, Renner W. Oxidative stress and free radicals in COPD--implications and relevance for treatment. Int. J. Chron. Obstruct. Pulmon. Dis. 2014;9:1207–1224. doi: 10.2147/COPD.S51226.
    1. Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2016;138:16–27. doi: 10.1016/j.jaci.2016.05.011.
    1. Nixon J, Newbold P, Mustelin T, Anderson GP, Kolbeck R. Monoclonal antibody therapy for the treatment of asthma and chronic obstructive pulmonary disease with eosinophilic inflammation. Pharmacol Ther. 2017;169:57–77. doi: 10.1016/J.PHARMTHERA.2016.10.016.
    1. George L, Brightling CE. Eosinophilic airway inflammation: role in asthma and chronic obstructive pulmonary disease. Ther Adv Chronic Dis. 2016;7:34–51. doi: 10.1177/2040622315609251.
    1. Sridhar S, Liu H, Pham T-H, Damera G, Newbold P. Modulation of blood inflammatory markers by benralizumab in patients with eosinophilic airway diseases. Respir Res. 2019;20:14. doi: 10.1186/s12931-018-0968-8.
    1. Alevy YG, Patel AC, Romero AG, Patel DA, Tucker J, Roswit WT, et al. IL-13-induced airway mucus production is attenuated by MAPK13 inhibition. J Clin Invest. 2012;122:4555–4568. doi: 10.1172/JCI64896.
    1. Beeh KM, Beier J, Kornmann O, Buhl R. Sputum matrix metalloproteinase-9, tissue inhibitor of metalloprotinease-1, and their molar ratio in patients with chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis and healthy subjects. Respir Med. 2003;97:634–639. doi: 10.1053/RMED.2003.1493.
    1. Groutas WC, Dou D, Alliston KR. Neutrophil elastase inhibitors. Expert Opin Ther Pat. 2011;21:339–354. doi: 10.1517/13543776.2011.551115.
    1. Hessel J, Heldrich J, Fuller J, Staudt MR, Radisch S, Hollmann C, et al. Intraflagellar transport gene expression associated with short cilia in smoking and COPD. PLoS One. 2014;9:e85453. doi: 10.1371/journal.pone.0085453.
    1. Yaghi A, Dolovich MB. Airway epithelial cell cilia and obstructive lung disease. Cells. 2016;5:40. doi: 10.3390/cells5040040.
    1. Srivastava PK, Dastidar SG, Ray A. Chronic obstructive pulmonary disease: role of matrix metalloproteases and future challenges of drug therapy. Expert Opin Investig Drugs. 2007;16:1069–1078. doi: 10.1517/13543784.16.7.1069.
    1. Costa C, Traves SL, Tudhope SJ, Fenwick PS, Belchamber KBR, Russell REK, et al. Enhanced monocyte migration to CXCR3 and CCR5 chemokines in COPD. Eur Respir J. 2016;47:1093–1102. doi: 10.1183/13993003.01642-2015.
    1. Gangemi S, Casciaro M, Trapani G, Quartuccio S, Navarra M, Pioggia G, et al. Association between HMGB1 and COPD: a systematic review. Mediat Inflamm. 2015;2015:164913. doi: 10.1155/2015/164913.
    1. Radsak MP, Taube C, Haselmayer P, Tenzer S, Salih HR, Wiewrodt R, et al. Soluble triggering receptor expressed on myeloid cells 1 is released in patients with stable chronic obstructive pulmonary disease. Clin Dev Immunol. 2007;2007:52040. doi: 10.1155/2007/52040.
    1. Petrusca DN, Gu Y, Adamowicz JJ, Rush NI, Hubbard WC, Smith PA, et al. Sphingolipid-mediated inhibition of apoptotic cell clearance by alveolar macrophages. J Biol Chem. 2010;285:40322–40332. doi: 10.1074/jbc.M110.137604.
    1. Chand S, Mehta N, Bahia MS, Dixit A, Silakari O. Protein kinase C-theta inhibitors: a novel therapy for inflammatory disorders. Curr Pharm Des. 2012;18:4725–4746. doi: 10.2174/138161212802651625.
    1. Pouwels SD, van Geffen WH, Jonker MR, Kerstjens HAM, Nawijn MC, Heijink IH. Increased neutrophil expression of pattern recognition receptors during COPD exacerbations. Respirology. 2017;22:401–404. doi: 10.1111/resp.12912.
    1. Kinose D, Ogawa E, Kudo M, Marumo S, Kiyokawa H, Hoshino Y, et al. Association of COPD exacerbation frequency with gene expression of pattern recognition receptors in inflammatory cells in induced sputum. Clin Respir J. 2016;10:11–21. doi: 10.1111/crj.12171.
    1. Pomerenke A, Lea SR, Herrick S, Lindsay MA, Singh D. Characterization of TLR-induced inflammatory responses in COPD and control lung tissue explants. Int J Chron Obstruct Pulmon Dis. 2016;11:2409–2417. doi: 10.2147/COPD.S105156.
    1. Yoshikawa T, Dent G, Ward J, Angco G, Nong G, Nomura N, et al. Impaired neutrophil chemotaxis in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;175:473–479. doi: 10.1164/rccm.200507-1152OC.
    1. Calabrese F, Baraldo S, Bazzan E, Lunardi F, Rea F, Maestrelli P, et al. IL-32, a novel proinflammatory cytokine in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;178:894–901. doi: 10.1164/rccm.200804-646OC.
    1. Cao J, Zhang L, Li D, Xu F, Huang S, Xiang Y, et al. IL-27 is elevated in patients with COPD and patients with pulmonary TB and induces human bronchial epithelial cells to produce CXCL10. Chest. 2012;141:121–130. doi: 10.1378/chest.10-3297.
    1. Vasquez RE, Xin L, Soong L. Effects of CXCL10 on dendritic cell and CD4 T-cell functions during Leishmania amazonensis infection. Infect Immun. 2008;76:161–169. doi: 10.1128/IAI.00825-07.
    1. Li X, O’Regan AW, Berman JS. IFN- γ induction of osteopontin expression in human monocytoid cells. J Interf Cytokine Res. 2004;23:259–265. doi: 10.1089/107999003321829971.
    1. Vlahos R, Bozinovski S. Role of alveolar macrophages in chronic obstructive pulmonary disease. Front Immunol. 2014;5:435. doi: 10.3389/fimmu.2014.00435.
    1. John-Schuster G, Conlon TM, Gunter S, Eickelberg O, Heikenwalder M, Yildirim AO. Lymphotoxin-β-receptor blockade prevents cigarette smoke-induced COPD. Am J Respir Crit Care Med. 2017;195:A4297.
    1. Betts JC, Mayer RJ, Tal-singer R, Warnock L, Clayton C, Bates S, et al. Gene expression changes caused by the p38 MAPK inhibitor dilmapimod in COPD patients : analysis of blood and sputum samples from a randomized, placebo-controlled clinical trial. Pharmacol Res Perspect. 2015;3:e00094. doi: 10.1002/prp2.94.
    1. van den Berge M, Steiling K, Timens W, Hiemstra PS, Sterk PJ, Heijink IH, et al. Airway gene expression in COPD is dynamic with inhaled corticosteroid treatment and reflects biological pathways associated with disease activity. Thorax. 2014;69:14–23. doi: 10.1136/thoraxjnl-2012-202878.
    1. Baines KJ, Wright TK, Gibson PG, Powell H, Hansbro PM, Simpson JL. Azithromycin treatment modifies airway and blood gene expression networks in neutrophilic COPD. ERJ Open Research. 2018;4:00031–02018. doi: 10.1183/23120541.00031-2018.

Source: PubMed

3
Abonnieren