145
Views
3
CrossRef citations to date
0
Altmetric
Review

Leukocyte Function in COPD: Clinical Relevance and Potential for Drug Therapy

&
Pages 2227-2242 | Published online: 30 Jul 2021

References

  • Belchamber KBR, Donnelly LE. Macrophage dysfunction in respiratory disease. Results Probl Cell Differ. 2017;62:299–313.
  • Hill AT, Campbell EJ, Hill SL, Bayley DL, Stockley RA. Association between airway bacterial load and markers of airway inflammation in patients with stable chronic bronchitis. Am J Med. 2000;109(4):288–295.
  • Donaldson GC, Seemungal TA, Bhowmik A, Wedzicha JA. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax. 2002;57(10):847–852.
  • Cai Y, Sugimoto C, Arainga M, Alvarez X, Didier ES, Kuroda MJ. In vivo characterization of alveolar and interstitial lung macrophages in rhesus macaques: implications for understanding lung disease in humans. J Immunol. 2014;192(6):2821–2829.
  • Perez-Rial S, Del Puerto-Nevado L, Terron-Exposito R, Giron-Martinez A, Gonzalez-Mangado N, Peces-Barba G. Role Oo recently migrated monocytes in cigarette smoke-induced lung inflammation in different strains of mice. PLoS One. 2013;8(9):e72975.
  • Evren E, Ringqvist E, Tripathi KP, et al. Distinct developmental pathways from blood monocytes generate human lung macrophage diversity. Immunity. 2020;54(2):259–275
  • Gomez Perdiguero E, Klapproth K, Schulz C, et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 2015;518(7540):547–551.
  • Tan SY, Krasnow MA. Developmental origin of lung macrophage diversity. Development. 2016;143(8):1318–1327.
  • Tomita K, Caramori G, Lim S, et al. Increased p21(CIP1/WAF1) and B cell lymphoma leukemia-x(L) expression and reduced apoptosis in alveolar macrophages from smokers. Am J Respir Crit Care Med. 2002;166(5):724–731.
  • Kulikauskaite J, Wack A. Teaching old dogs new tricks? The plasticity of lung alveolar macrophage subsets. Trends Immunol. 2020;41(10):864–877.
  • Culpitt SV, Rogers DF, Shah P, et al. Impaired inhibition by dexamethasone of cytokine release by alveolar macrophages from patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2003;167(1):24–31.
  • Russell RE, Culpitt SV, DeMatos C, et al. Release and activity of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 by alveolar macrophages from patients with chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2002;26(5):602–609.
  • Davies LC, Rice CM, Palmieri EM, Taylor PR, Kuhns DB, McVicar DW. Peritoneal tissue-resident macrophages are metabolically poised to engage microbes using tissue-niche fuels. Nat Commun. 2017;8(1):2074.
  • Ushach I, Zlotnik A. Biological role of granulocyte macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) on cells of the myeloid lineage. J Leukoc Biol. 2016;100(3):481–489.
  • Finlay GA, O’Driscoll LR, Russell KJ, et al. Matrix metalloproteinase expression and production by alveolar macrophages in emphysema. Am J Respir Crit Care Med. 1997;156(1):240–247.
  • Russell RE, Thorley A, Culpitt SV, et al. Alveolar macrophage-mediated elastolysis: roles of matrix metalloproteinases, cysteine, and serine proteases. Am J Physiol Lung Cell Mol Physiol. 2002;283(4):L867–L873.
  • Berenson CS, Garlipp MA, Grove LJ, Maloney J, Sethi S. Impaired phagocytosis of nontypeable haemophilus influenzae by human alveolar macrophages in chronic obstructive pulmonary disease. J Infect Dis. 2006;194(10):1375–1384.
  • Taylor AE, Finney-Hayward TK, Quint JK, et al. Defective macrophage phagocytosis of bacteria in COPD. Eur Respir J. 2010;35(5):1039–1047.
  • Bewley MA, Belchamber KB, Chana KK, et al. Differential effects of p38, MAPK, PI3K or Rho Kinase Inhibitors on bacterial phagocytosis and efferocytosis by macrophages in COPD. PLoS One. 2016;11(9):e0163139.
  • Hodge S, Hodge G, Scicchitano R, Reynolds PN, Holmes M. Alveolar macrophages from subjects with chronic obstructive pulmonary disease are deficient in their ability to phagocytose apoptotic airway epithelial cells. Immunol Cell Biol. 2003;81(4):289–296.
  • Wrench C, Belchamber KBR, Bercusson A, et al. Reduced clearance of fungal spores by chronic obstructive pulmonary disease GM-CSF- and M-CSF-derived macrophages. Am J Respir Cell Mol Biol. 2018;58(2):271–273.
  • Birrell MA, Wong S, Catley MC, Belvisi MG. Impact of tobacco-smoke on key signaling pathways in the innate immune response in lung macrophages. J Cell Physiol. 2008;214(1):27–37.
  • Oliver BG, Lim S, Wark P, et al. Rhinovirus exposure impairs immune responses to bacterial products in human alveolar macrophages. Thorax. 2008;63(6):519–525.
  • Finney LJ, Belchamber KBR, Fenwick PS, et al. Human rhinovirus impairs the innate immune response to bacteria in alveolar macrophages in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2019;199(12):1496–1507.
  • Belchamber KBR, Singh R, Batista CM, et al. Defective bacterial phagocytosis is associated with dysfunctional mitochondria in COPD macrophages. Eur Respir J. 2019;54:4.
  • Bewley MA, Preston JA, Mohasin M, et al. Impaired mitochondrial microbicidal responses in chronic obstructive pulmonary disease macrophages. Am J Respir Crit Care Med. 2017;196(7):845–855.
  • Chana KK, Fenwick P, Nicholson AG, Barnes PJ, Donnelly LE. Identification of a distinct glucocorticoid-insensitive pulmonary macrophage phenotype in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2014;133:1.
  • Traves SL, Smith SJ, Barnes PJ, Donnelly LE. Specific CXC but not CC chemokines cause elevated monocyte migration in COPD: a role for CXCR2. J Leukoc Biol. 2004;76:441–450.
  • Costa C, Traves SL, Tudhope SJ, et al. Enhanced monocyte migration to CXCR3 and CCR5 chemokines in COPD. Eur Respir J. 2016;47(4):1093–1102.
  • Faurschou M, Borregaard N. Neutrophil granules and secretory vesicles in inflammation. Microbes Infect. 2003;5(14):1317–1327.
  • Barnes PJ. Inflammatory endotypes in COPD. Allergy. 2019;74(7):1249–1256.
  • Patel AA, Ginhoux F, Yona S. Monocytes, macrophages, dendritic cells and neutrophils: an update on lifespan kinetics in health and disease. Immunology. 2021;163(3):250–261.
  • Sheshachalam A, Srivastava N, Mitchell T, Lacy P, Eitzen G. Granule protein processing and regulated secretion in neutrophils. Front Immunol. 2014;5:448.
  • Metzemaekers M, Gouwy M, Proost P. Neutrophil chemoattractant receptors in health and disease: double-edged swords. Cell Mol Immunol. 2020;17(5):433–450.
  • Hughes MJ, Sapey E, Stockley R. Neutrophil phenotypes in chronic lung disease. Expert Rev Respir Med. 2019;13(10):951–967.
  • Dunne AE, Kawamatawong T, Fenwick PS, et al. Direct inhibitory effect of the PDE4 inhibitor roflumilast on neutrophil migration in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2019;60(4):445–453.
  • Burnett D, Chamba A, Hill SL, Stockley RA. Neutrophils from subjects with chronic obstructive lung disease show enhanced chemotaxis and extracellular proteolysis. Lancet. 1987;2(8567):1043–1046.
  • Sapey E, Stockley JA, Greenwood H, et al. Behavioral and structural differences in migrating peripheral neutrophils from patients with COPD. Am J Respir Crit Care Med. 2011;183(9):176–186.
  • Butler A, Walton GM, Sapey E. Neutrophilic inflammation in the pathogenesis of chronic obstructive pulmonary disease. COPD. 2018;15(4):392–404.
  • Shanmugam L, Ravinder S, Johnson P, Padmavathi R, Rajagopalan B, Kindo A. Assessment of phagocytic activity of neutrophils in chronic obstructive pulmonary disease. Lung India. 2015;32(5):437–440.
  • Uddin M, Watz H, Malmgren A, Pedersen F. NETopathic inflammation in chronic obstructive pulmonary disease and severe asthma. Front Immunol. 2019;10:47.
  • Keatings VM, Collins PD, Scott DM, Barnes PJ. Differences in interleukin-8 and tumor necrosis factor-alpha in induced sputum from patients with chronic obstructive pulmonary disease or asthma. Am J Respir Crit Care Med. 1996;153(2):530–534.
  • Traves SL, Culpitt SV, Russell RE, Barnes PJ, Donnelly LE. Increased levels of the chemokines GROα and MCP-1 in sputum samples from patients with COPD. Thorax. 2002;57(7):590–595.
  • Mori Y, Iwasaki H, Kohno K, et al. Identification of the human eosinophil lineage-committed progenitor: revision of phenotypic definition of the human common myeloid progenitor. J Exp Med. 2009;206(1):183–193.
  • Olsson A, Salomonis N, Singh H, Grimes HL. Identification of the origin of eosinophils. Blood. 2015;126(23):886.
  • Vedel-Krogh S. The search for the “healthy” blood eosinophil count. Eur Respir J. 2020;55(5):2000473.
  • Pavord ID. Eosinophilic phenotypes of airway disease. Ann Am Thorac Soc. 2013;10(Suppl):S143–149.
  • Thompson-Souza GA, Gropillo I, Neves JS. Cysteinyl leukotrienes in eosinophil biology: functional roles and therapeutic perspectives in eosinophilic disorders. Front Med (Lausanne). 2017;4:106.
  • Leigh R, Pizzichini MMM, Morris MM, Maltais F, Hargreave FE, Pizzichini E. Stable COPD: predicting benefit from high-dose inhaled corticosteroid treatment. Eur Respir J. 2006;27(5):964–971.
  • Felton JM, Lucas CD, Rossi AG, Dransfield I. Eosinophils in the lung – modulating apoptosis and efferocytosis in airway inflammation. Front Immunol. 2014;5:302.
  • Jeffery PK. Comparison of the structural and inflammatory features of COPD and asthma. Giles F. Filley Lecture. Chest. 2000;117(5Suppl 1):251S–260S.
  • David B, Bafadhel M, Koenderman L, De Soyza A. Eosinophilic inflammation in COPD: from an inflammatory marker to a treatable trait. Thorax. 2020;76(2):188–195.
  • Bafadhel M, McKenna S, Terry S, et al. Blood eosinophils to direct corticosteroid treatment of exacerbations of chronic obstructive pulmonary disease: a randomized placebo-controlled trial. Am J Respir Crit Care Med. 2012;186(1):48–55.
  • Tashkin DP, Wechsler ME. Role of eosinophils in airway inflammation of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2018;13:335–349.
  • Littner MR, Ilowite JS, Tashkin DP, et al. Long-acting bronchodilation with once-daily dosing of tiotropium (Spiriva) in stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2000;161(4 Pt 1):1136–1142.
  • 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(6):662–671.
  • Méndez-Enríquez E, Hallgren J. Mast cells and their progenitors in allergic asthma. Front Immunol. 2019;10:821.
  • Födinger M, Fritsch G, Winkler K, et al. Origin of human mast cells: development from transplanted hematopoietic stem cells after allogeneic bone marrow transplantation. Blood. 1994;84(9):2954–2959.
  • Valent P, Akin C, Hartmann K, et al. Mast cells as a unique hematopoietic lineage and cell system: from Paul Ehrlich’s visions to precision medicine concepts. Theranostics. 2020;10(23):10743–10768.
  • Sonoda T, Hayashi C, Kitamura Y. Presence of mast cell precursors in the yolk sac of mice. Dev Biol. 1983;97(1):89–94.
  • Irani AA, Schechter NM, Craig SS, DeBlois G, Schwartz LB. Two types of human mast cells that have distinct neutral protease compositions. Proc Natl Acad Sci U S A. 1986;83(12):4464–4468.
  • Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ, Pavord ID. Mast-cell infiltration of airway smooth muscle in asthma. N Engl J Med. 2002;346(22):1699–1705.
  • Carroll NG, Mutavdzic S, James AL. Distribution and degranulation of airway mast cells in normal and asthmatic subjects. Eur Respir J. 2002;19(5):879–885.
  • Raible DG, Schulman ES, DiMuzio J, Cardillo R, Post TJ. Mast cell mediators prostaglandin-D2 and histamine activate human eosinophils. J Immunol. 1992;148(11):3536–3542.
  • Murakami D, Yamada H, Yajima T, Masuda A, Komune S, Yoshikai Y. Lipopolysaccharide inhalation exacerbates allergic airway inflammation by activating mast cells and promoting Th2 responses. Clin Exp Allergy. 2007;37(3):339–347.
  • Beckett EL, Stevens RL, Jarnicki AG, et al. A new short-term mouse model of chronic obstructive pulmonary disease identifies a role for mast cell tryptase in pathogenesis. J Allergy Clin Immun. 2013;131(3):752–762.
  • Andersson CK, Mori M, Bjermer L, Löfdahl C-G, Erjefält JS. Alterations in lung mast cell populations in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;181(3):206–217.
  • Ballarin A, Bazzan E, Zenteno RH, et al. Mast cell infiltration discriminates between histopathological phenotypes of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012;186(3):233–239.
  • López-Giraldo A, Cruz T, Molins L, et al. Characterization, localization and comparison of c-Kit+ lung cells in never smokers and smokers with and without COPD. BMC Pulm Med. 2018;18(1):123.
  • Swain SL, McKinstry KK, Strutt TM. Expanding roles for CD4(+) T cells in immunity to viruses. Nat Rev Immunol. 2012;12(2):136–148.
  • Kanhere A, Hertweck A, Bhatia U, et al. T-bet and GATA3 orchestrate Th1 and Th2 differentiation through lineage-specific targeting of distal regulatory elements. Nat Commun. 2012;3:1–2.
  • Hodge G, Nairn J, Holmes M, Reynolds PN, Hodge S. Increased intracellular T helper 1 proinflammatory cytokine production in peripheral blood, bronchoalveolar lavage and intraepithelial T cells of COPD subjects. Clin Exp Immunol. 2007;150(1):22–29.
  • Barcelo B, Pons J, Ferrer JM, Sauleda J, Fuster A, Agusti AG. Phenotypic characterisation of T-lymphocytes in COPD: abnormal CD4+CD25+ regulatory T-lymphocyte response to tobacco smoking. Eur Respir J. 2008;31(3):555–562.
  • Saetta M, Mariani M, Panina-Bordignon P, et al. Increased expression of the chemokine receptor CXCR3 and its ligand CXCL10 in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;165(10):1404–1409.
  • Di Stefano A, Caramori G, Capelli A, et al. STAT4 activation in smokers and patients with chronic obstructive pulmonary disease. Eur Respir J. 2004;24(1):78–85.
  • Schmidt A, Oberle N, Krammer PH. Molecular mechanisms of treg-mediated T cell suppression. Front Immunol. 2012;3:51.
  • Smyth LJ, Starkey C, Vestbo J, Singh D. CD4-regulatory cells in COPD patients. Chest. 2007;132(1):156–163.
  • Tesmer LA, Lundy SK, Sarkar S, Fox DA. Th17 cells in human disease. Immunol Rev. 2008;223:87–113.
  • Le Rouzic O, Pichavant M, Frealle E, Guillon A, Si-Tahar M, Gosset P. Th17 cytokines: novel potential therapeutic targets for COPD pathogenesis and exacerbations. Eur Respir J. 2017;50:4.
  • Di Stefano A, Caramori G, Gnemmi I, et al. T helper type 17-related cytokine expression is increased in the bronchial mucosa of stable chronic obstructive pulmonary disease patients. Clin Exp Immunol. 2009;157(2):316–324.
  • Prause O, Bozinovski S, Anderson GP, Linden A. Increased matrix metalloproteinase-9 concentration and activity after stimulation with interleukin-17 in mouse airways. Thorax. 2004;59(4):313–317.
  • Zhang N, Bevan MJ. CD8(+) T cells: foot soldiers of the immune system. Immunity. 2011;35(2):161–168.
  • Suzuki I, Martin S, Boursalian TE, Beers C, Fink PJ. Fas ligand costimulates the in vivo proliferation of CD8+ T cells. J Immunol. 2000;165(10):5537–5543.
  • Schmidt ME, Varga SM. The CD8 T cell response to respiratory virus infections. Front Immunol. 2018;9:678.
  • Saetta M, Di Stefano A, Turato G, et al. CD8+ T-lymphocytes in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157(3):822–826.
  • Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immun. 2016;138(1):16–27.
  • Roos-Engstrand E, Ekstrand-Hammarstrom B, Pourazar J, Behndig AF, Bucht A, Blomberg A. Influence of smoking cessation on airway T lymphocyte subsets in COPD. Copd. 2009;6(2):112–120.
  • Wang J, Urbanowicz RA, Tighe PJ, Todd I, Corne JM, Fairclough LC. Differential activation of killer cells in the circulation and the lung: a study of current smoking status and chronic obstructive pulmonary disease (COPD). PLoS One. 2013;8(3):e58556.
  • Freeman CM, Curtis JL, Chensue SW. CC chemokine receptor 5 and CXC chemokine receptor 6 expression by lung CD8+ cells correlates with chronic obstructive pulmonary disease severity. Am J Pathol. 2007;171(3):767–776.
  • Williams M, Todd I, Fairclough LC. The role of CD8 + T lymphocytes in chronic obstructive pulmonary disease: a systematic review. Inflamm Res. 2021;70(1):11–18.
  • Hodge G, Mukaro V, Reynolds PN, Hodge S. Role of increased CD8/CD28(null) T cells and alternative co-stimulatory molecules in chronic obstructive pulmonary disease. Clin Exp Immunol. 2011;166(1):94–102.
  • Kagi D, Ledermann B, Burki K, Zinkernagel RM, Hengartner H. Molecular mechanisms of lymphocyte-mediated cytotoxicity and their role in immunological protection and pathogenesis in vivo. Annu Rev Immunol. 1996;14:207–232.
  • De Cunto G, Lunghi B, Bartalesi B, et al. Severe reduction in number and function of peripheral T cells does not afford protection toward emphysema and bronchial remodeling induced in mice by cigarette smoke. Am J Pathol. 2016;186(7):1814–1824.
  • Hogg JC, Chu F, Utokaparch S, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350(26):2645–2653.
  • Gosman MM, Willemse BW, Jansen DF, et al. Increased number of B-cells in bronchial biopsies in COPD. Eur Respir J. 2006;27(1):60–64.
  • van der Strate BW, Postma DS, Brandsma CA, et al. Cigarette smoke-induced emphysema: a role for the B cell? Am J Respir Crit Care Med. 2006;173(7):751–758.
  • Ludwig RJ, Vanhoorelbeke K, Leypoldt F, et al. Mechanisms of autoantibody-induced pathology. Front Immunol. 2017;8:603.
  • Lee SH, Goswami S, Grudo A, et al. Antielastin autoimmunity in tobacco smoking-induced emphysema. Nat Med. 2007;13(5):567–569.
  • Nunez B, Sauleda J, Anto JM, et al. Anti-tissue antibodies are related to lung function in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2011;183(8):1025–1031.
  • Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9(5):503–510.
  • Culley FJ. Natural killer cells in infection and inflammation of the lung. Immunology. 2009;128(2):151–163.
  • Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends Immunol. 2001;22(11):633–640.
  • Huntington ND. The unconventional expression of IL-15 and its role in NK cell homeostasis. Immunol Cell Biol. 2014;92(3):210–213.
  • Freeman CM, Stolberg VR, Crudgington S, et al. Human CD56+ cytotoxic lung lymphocytes kill autologous lung cells in chronic obstructive pulmonary disease. PLoS One. 2014;9(7):e103840.
  • Robinson BW, Pinkston P, Crystal RG. Natural killer cells are present in the normal human lung but are functionally impotent. J Clin Invest. 1984;74(3):942–950.
  • Takeuchi M, Nagai S, Nakajima A, et al. Inhibition of lung natural killer cell activity by smoking: the role of alveolar macrophages. Respiration. 2001;68(3):262–267.
  • Hodge G, Mukaro V, Holmes M, Reynolds PN, Hodge S. Enhanced cytotoxic function of natural killer and natural killer T-like cells associated with decreased CD94 (Kp43) in the chronic obstructive pulmonary disease airway. Respirology. 2013;18(2):369–376.
  • Urbanowicz RA, Lamb JR, Todd I, Corne JM, Fairclough LC. Enhanced effector function of cytotoxic cells in the induced sputum of COPD patients. Respir Res. 2010;11:76.
  • Finch DK, Stolberg VR, Ferguson J, et al. Lung dendritic cells drive natural killer cytotoxicity in chronic obstructive pulmonary disease via IL-15Ralpha. Am J Respir Crit Care Med. 2018;198(9):1140–1150.
  • Hume P, Gibbings S, Jakubzick CV, et al. Localization of macrophages in the human lung via design-based stereology. Am J Respir Crit Care Med. 2020;201.
  • De Cunto G, Cavarra E, Bartalesi B, Lungarella G, Lucattelli M. Alveolar macrophage phenotype and compartmentalization drive different pulmonary changes in mouse strains exposed to cigarette smoke. COPD. 2020;17(4):429–443.
  • Theron AJ, Steel HC, Tintinger GR, Feldman C, Anderson R. Can the anti-inflammatory activities of β2-agonists be harnessed in the clinical setting? Drug Des Devel Ther. 2013;7:1387–1398.
  • Donnelly LE, Tudhope SJ, Fenwick PS, Barnes PJ. Effects of formoterol and salmeterol on cytokine release from monocyte-derived macrophages. Eur Respir J. 2010;36(1):178–186.
  • Haque R, Hakim A, Moodley T, et al. Inhaled long-acting β2 agonists enhance glucocorticoid receptor nuclear translocation and efficacy in sputum macrophages in COPD. J Allergy Clin Immun. 2013;132(5):1166–1173.
  • Perttunen H, Moilanen E, Zhang X, Barnes PJ, Kankaanranta H. Beta2-agonists potentiate corticosteroid-induced neutrophil survival. Copd. 2008;5(3):163–169.
  • Milara J, Contreras S, de Diego A, et al. In vitro anti-inflammatory effects of AZD8999, a novel bifunctional muscarinic acetylcholine receptor antagonist /β2-adrenoceptor agonist (MABA) compound in neutrophils from COPD patients. PLoS One. 2019;14(1):e0210188.
  • Calverley P, Rabe K, Goehring U, Kristiansen S, Fabbri L, Martinez F. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet. 2009;374:685–694.
  • Porpodis K, Domvri K, Zarogoulidis P, et al. Roflumilast, a phosphodiesterase-4 inhibitor, induces phagocytic activity in Greek COPD patients. Int J Chron Obstruct Pulmon Dis. 2015;10:1123–1128.
  • 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.e319.
  • Belchamber KB, Thomas CM, Dunne AE, Barnes PJ, Donnelly LE. Comparison of fluticasone propionate and budesonide on COPD macrophage and neutrophil function. Int J Chron Obstruct Pulmon Dis. 2018;13:2883–2897.
  • Hodge S, Hodge G, Brozyna S, Jersmann H, Holmes M, Reynolds PN. Azithromycin increases phagocytosis of apoptotic bronchial epithelial cells by alveolar macrophages. Eur Respir J. 2006;28(3):486–495.
  • Hodge S, Hodge G, Jersmann H, et al. Azithromycin improves macrophage phagocytic function and expression of mannose receptor in COPD. Am J Respir Crit Care Med. 2008;178(2):139–148.
  • Hodge S, Reynolds PN. Low-dose azithromycin improves phagocytosis of bacteria by both alveolar and monocyte-derived macrophages in chronic obstructive pulmonary disease subjects. Respirology. 2012;17(5):802–807.
  • Hodge S, Tran HB, Hamon R, et al. Nonantibiotic macrolides restore airway macrophage phagocytic function with potential anti-inflammatory effects in chronic lung diseases. Am J Physiol-Lung C. 2017;312(5):L678–L687.
  • Ji J, von Schéele I, Billing B, et al. Effects of budesonide on toll-like receptor expression in alveolar macrophages from smokers with and without COPD. Int J Chron Obstruct Pulmon Dis. 2016;11:1035–1043.
  • GOLD. Executive summary: global strategy for the diagnosis, management, and prevention of COPD; 2020. Available from: http://www.goldcopd.com/Guidelineitem.asp?l1=2&l2=1&intId=996. Accessed July 17, 2021.
  • Cosio BG, Tsaprouni L, Ito K, Jazrawi E, Adcock IM, Barnes PJ. Theophylline restores histone deacetylase activity and steroid responses in COPD macrophages. J Exp Med. 2004;200(5):689–695.
  • Higham A, Scott T, Li J, et al. Effects of corticosteroids on COPD lung macrophage phenotype and function. Clin Sci (Lond). 2020;134(7):751–763.
  • Hodge G, Hodge S. Steroid resistant CD8(+)CD28(null) NKT-like pro-inflammatory cytotoxic cells in chronic obstructive pulmonary disease. Front Immunol. 2016;7:617.
  • Donnelly LE, Barnes PJ. Chemokine receptors as therapeutic targets in chronic obstructive pulmonary disease. Trends Pharmacol Sci. 2006;27(10):546–553.
  • Rennard SI, Dale DC, Donohue JF, et al. CXCR2 antagonist MK-7123. A Phase 2 proof-of-concept trial for chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2015;191(9):1001–1011.
  • Kirsten AM, Förster K, Radeczky E, et al. The safety and tolerability of oral AZD5069, a selective CXCR2 antagonist, in patients with moderate-to-severe COPD. Pulm Pharmacol Ther. 2015;31:36–41.
  • Jurcevic S, Humfrey C, Uddin M, Warrington S, Larsson B, Keen C. The effect of a selective CXCR2 antagonist (AZD5069) on human blood neutrophil count and innate immune functions. Br J Clin Pharmacol. 2015;80(6):1324–1336.
  • Lazaar AL, Miller BE, Tabberer M, et al. Effect of the CXCR2 antagonist danirixin on symptoms and health status in COPD. Eur Respir J. 2018;52:4.
  • Lazaar AL, Miller BE, Donald AC, et al. CXCR2 antagonist for patients with chronic obstructive pulmonary disease with chronic mucus hypersecretion: a Phase 2b trial. Respir Res. 2020;21(1):149.
  • Keir HR, Richardson H, Fillmore C, et al. CXCL-8-dependent and -independent neutrophil activation in COPD: experiences from a pilot study of the CXCR2 antagonist danirixin. ERJ Open Res. 2020;6:4.
  • AZD2423 safety and tolerability study in patients with moderate and severe Chronic Obstructive Pulmonary Disease(COPD). Available from: https://ClinicalTrials.gov/show/NCT01215279. Accessed July 20, 2021.
  • Liu J, Fu Z, Li AR, et al. Optimization of a series of quinazolinone-derived antagonists of CXCR3. Bioorg Med Chem Lett. 2009;19(17):5114–5118.
  • Greenwood J, Steinman L, Zamvil SS. Statin therapy and autoimmune disease: from protein prenylation to immunomodulation. Nat Rev Immunol. 2006;6(5):358–370.
  • Walton GM, Stockley JA, Griffiths D, Sadhra CS, Purvis T, Sapey E. Repurposing treatments to enhance innate immunity can statins improve neutrophil functions and clinical outcomes in COPD? J Clin Med. 2016;5:10.
  • Sapey E, Patel JM, Greenwood H, et al. Simvastatin improves neutrophil function and clinical outcomes in pneumonia. A pilot randomized controlled clinical trial. Am J Respir Crit Care Med. 2019;200(10):1282–1293.
  • Bellosta S, Via D, Canavesi M, et al. HMG-CoA reductase inhibitors reduce MMP-9 secretion by macrophages. Arterioscl Throm Vas. 1998;18(11):1671–1678.
  • Tuomisto TT, Lumivuori H, Kansanen E, et al. Simvastatin has an anti-inflammatory effect on macrophages via upregulation of an atheroprotective transcription factor, Kruppel-like factor 2. Cardiovasc Res. 2008;78(1):175–184.
  • Singh D, Kolsum U, Brightling CE, et al. Eosinophilic inflammation in COPD: prevalence and clinical characteristics. Eur Respir J. 2014;44(6):1697–1700.
  • Wedzicha JA. Eosinophils as biomarkers of chronic obstructive pulmonary disease exacerbation risk maybe just for some? Am J Respir Crit Care Med. 2016;193(9):937–938.
  • Pavord ID, Chanez P, Criner GJ, et al. Mepolizumab for eosinophilic chronic obstructive pulmonary disease. N Engl J Med. 2017;377(17):1613–1629.
  • Criner GJ, Celli BR, Brightling CE, et al. Benralizumab for the prevention of COPD exacerbations. N Engl J Med. 2019;381(11):1023–1034.
  • Eich A, Urban V, Jutel M, et al. A randomized, placebo-controlled Phase 2 trial of CNTO 6785 in Chronic Obstructive Pulmonary Disease. Copd. 2017;14(5):476–483.
  • Yanagisawa H, Hashimoto M, Minagawa S, et al. Role of IL-17A in murine models of COPD airway disease. Am J Physiol Lung Cell Mol Physiol. 2017;312(1):L122–L130.
  • 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.
  • Tezepelumab COPD exacerbation study. Available from: https://ClinicalTrials.gov/show/NCT04039113. Accessed July 20, 2021.
  • Charron CE, Russell P, Ito K, et al. RV568, a narrow-spectrum kinase inhibitor with p38 MAPK-alpha and -gamma selectivity, suppresses COPD inflammation. Eur Respir J. 2017;50:4.
  • Watz H, Barnacle H, Hartley BF, Chan R. Efficacy and safety of the p38 MAPK inhibitor losmapimod for patients with chronic obstructive pulmonary disease: a randomised, double-blind, placebo-controlled trial. Lancet Respir Med. 2014;2(1):63–72.
  • Lomas DA, Lipson DA, Miller BE, et al. An oral inhibitor of p38 MAP kinase reduces plasma fibrinogen in patients with chronic obstructive pulmonary disease. J Clin Pharmacol. 2012;52(3):416–424.
  • Marks-Konczalik J, Costa M, Robertson J, McKie E, Yang S, Pascoe S. A post-hoc subgroup analysis of data from a six month clinical trial comparing the efficacy and safety of losmapimod in moderate-severe COPD patients with ≤2% and >2% blood eosinophils. Respir Med. 2015;109(7):860–869.
  • Patel NR, Cunoosamy DM, Fagerås M, et al. The development of AZD7624 for prevention of exacerbations in COPD: a randomized controlled trial. Int J Chron Obstruct Pulmon Dis. 2018;13:1009–1019.
  • Strâmbu IR, Kobalava ZD, Magnusson BP, MacKinnon A, Parkin JM. Phase II study of single/repeated doses of acumapimod (BCT197) to treat acute exacerbations of COPD. Copd. 2019;16(5–6):344–353.
  • Investigate the Safety, Tolerability and Pharmacokinetics of CHF6523 in Healthy and in COPD Subjects. Available from: https://ClinicalTrials.gov/show/NCT04032535. Accessed July 20, 2021.
  • Khindri S, Cahn A, Begg M, et al. A multicentre, randomized, double-blind, placebo-controlled, crossover study to investigate the efficacy, safety, tolerability, and pharmacokinetics of repeat doses of inhaled nemiralisib in adults with persistent, uncontrolled asthma. J Pharmacol Exp Ther. 2018;367(3):405–413.
  • 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(3):337–344.
  • Dose finding study of nemiralisib (GSK2269557) in subjects with an acute moderate or severe exacerbation of Chronic Obstructive Pulmonary Disease (COPD). Available from: https://ClinicalTrials.gov/show/NCT03345407. Accessed July 20, 2021.
  • Harvey CJ, Thimmulappa RK, Sethi S, et al. Targeting Nrf2 signaling improves bacterial clearance by alveolar macrophages in patients with COPD and in a mouse model. Sci Transl Med. 2011;3(78):78ra32.
  • Bewley MA, Budd RC, Ryan E, et al. Opsonic phagocytosis in chronic obstructive pulmonary disease is enhanced by Nrf2 agonists. Am J Respir Crit Care Med. 2018;198(6):739–750.
  • Wise RA, Holbrook JT, Criner G, et al. Lack of effect of oral sulforaphane administration on Nrf2 expression in COPD: a randomized, double-blind, placebo controlled trial. PLoS One. 2016;11(11):e0163716.
  • Boehm J, Davis R, Murar CE, et al. Discovery of a crystalline sulforaphane analog with good solid-state stability and engagement of the Nrf2 pathway in vitro and in vivo. Bioorg Med Chem. 2019;27(4):579–588.
  • Hybertson BM, Gao B, Bose SK, McCord JM. Oxidative stress in health and disease: the therapeutic potential of Nrf2 activation. Mol Aspects Med. 2011;32(4–6):234–246.