Inhibition of LPS-induced airway neutrophilic inflammation in healthy volunteers with an oral CXCR2 antagonist

Brian R Leaker, Peter J Barnes, Brian O'Connor, Brian R Leaker, Peter J Barnes, Brian O'Connor

Abstract

Background: Inhaled lipopolysaccharide (LPS) induces a dose-dependent, acute neutrophilic response in the airways of healthy volunteers that can be quantified in induced sputum. Chemokines, such as CXCL1 and CXCL8, play an important role in neutrophilic inflammation in the lung through the activation of CXCR2 and small molecule antagonists of these receptors have now been developed. We investigated the effect of AZD8309, a CXCR2 antagonist, compared with placebo on LPS-induced inflammation measured in sputum of healthy volunteers.

Methods: Twenty healthy subjects were randomized in a double-blind placebo-controlled, cross-over study. AZD8309 (300 mg) or placebo was dosed twice daily orally for 3 days prior to challenge with inhaled LPS and induced sputum was collected 6 h later.

Results: Treatment with AZD8309 showed a mean 77% reduction in total sputum cells (p < 0.001) and 79% reduction in sputum neutrophils (p < 0.05) compared with placebo after LPS challenge. There was also a reduction in neutrophil elastase activity (p < 0.05) and CXCL1 (p < 0.05) and trends for reductions in sputum macrophages (47%), leukotriene B4 (39%) and CXCL8 (52%).

Conclusions: AZD8309 inhibited LPS-induced inflammation measured in induced sputum of normal volunteers, indicating that this treatment may be useful in the treatment of neutrophilic diseases of the airways, such as COPD, severe asthma and cystic fibrosis.

Trial registration: NCT00860821.

Figures

Figure 1
Figure 1
Study design. After a screening visit (visit 1) treatment period 1 was comprised of visits 2 and 3 and treatment period 2 was comprised of visits 4 and 5. Eligible randomized subjects returned to the clinic (visit 2) 7–21 days following visit 1 and were dosed with either 300 mg AZD8309 or placebo at 09:00 hours and were discharged with instructions for continued dosing at home. Subjects returned for LPS challenge the following morning (visit 3). Six hours post-completion of the LPS inhalation, subjects produced an induced sputum sample. There was a washout period between visits 3 and 4 of at least 21 days. Treatment period 2 was structured the same as treatment period 1.
Figure 2
Figure 2
Effect of AZD8309 on cell counts in sputum post LPS challenge. Total cell counts (top left plot) were performed by trypan blue exclusion with a hemocytometer. Differential cell counts for neutrophils (top right plot) and macrophages (bottom centre plot) were performed with cytospin preparations. Bars show the mean values of 15–16 patients. Open bars represent placebo and solid bars AZD8309. Results are expressed as geometric mean. CVs are shown in Table 2. Statistics by ANOVA, ** = P < 0.001 and * = P < 0.05.
Figure 3
Figure 3
Effect of AZ8309 on mediator concentrations in sputum supernatant. Sputum samples were collected 6 hours post challenge with LPS. There was a reduction in concentrations of CXCL8 (top left plot), CXCL1 (top right plot), neutrophil elastase activity (NEA) (bottom left plot) and leukotriene B4 (LTB4) (Bottom right plot); mean values of 13–16 patients are shown. Open bars represent placebo and solid bars AZD8309. Results are expressed as geometric mean. CVs are shown in Table 3. Statistics by ANOVA, * = P < 0.05.
Figure 4
Figure 4
Effect of AZD8309 on circulating blood neutrophils pre and post challenge with lipopolysaccharide. Blood neutrophils numbers were measured at predose (t = 0) and at 7 hours post dosing on day 1 and at trough (t = 0) and at 1, 3, 7, 12 and 25 hours post dosing on day 3. The solid black circles show the effects on placebo treatment and the open grey circles show effects after treatment with AZD8309. The arrow indicates the point at which subjects were challenged with LPS on day 3. Each point is presented as mean with s.e.m.
Figure 5
Figure 5
Effect of AZD8309 on FEV1 after inhaled lipopolysaccharide (LPS). There was a similar peak fall in FEV1 at 1 h after challenge, then a more rapid recovery after AZD8309 compared to placebo, with a significant reduction in area under the curve (P < 0.05). The average values for the coefficient of variances were 14.4% and 15% for AZD8309 and placebo respectively. Mean values of 16 patients are shown.

References

    1. Barnes PJ. New anti-inflammatory treatments for chronic obstructive pulmonary disease. Nat New Drug Disc. 2013;12:543–559. doi: 10.1038/nrd4025.
    1. Barnes PJ. Severe asthma: advances in current management and future therapy. J Allergy Clin Immunol. 2012;129:48–59. doi: 10.1016/j.jaci.2011.11.006.
    1. Keatings VM, Collins PD, Scott DM, Barnes PJ. Differences in interleukin-8 and tumor necrosis factor-α in induced sputum from patients with chronic obstructive pulmonary disease or asthma. Am J Respir Crit Care Med. 1996;153:530–534. doi: 10.1164/ajrccm.153.2.8564092.
    1. Beeh KM, Kornmann O, Buhl R, Culpitt SV, Giembycz MA, Barnes PJ. Neutrophil chemotactic activity of sputum from patients with COPD: role of interleukin 8 and leukotriene B4. Chest. 2003;123:1240–1247. doi: 10.1378/chest.123.4.1240.
    1. Traves SL, Culpitt S, Russell REK, Barnes PJ, Donnelly LE. Elevated levels of the chemokines GRO-α and MCP-1 in sputum samples from COPD patients. Thorax. 2002;57:590–595. doi: 10.1136/thorax.57.7.590.
    1. Qiu Y, Zhu J, Bandi V, Atmar R, Hattotuwa K, Guntapalli K, Jeffery P. Biopsy neutrophilia, chemokine and receptor gene expression in severe exacerbations of COPD. Am J Respir Crit Care Med. 2003;168:968–975. doi: 10.1164/rccm.200208-794OC.
    1. Jatakanon A, Uasaf C, Maziak W, Lim S, Chung KF, Barnes PJ. Neutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med. 1999;160:1532–1539. doi: 10.1164/ajrccm.160.5.9806170.
    1. Mackerness KJ, Jenkins GR, Bush A, Jose PJ. Characterisation of the range of neutrophil stimulating mediators in cystic fibrosis sputum. Thorax. 2008;63:614–620. doi: 10.1136/thx.2007.089359.
    1. Donnelly LE, Barnes PJ. Chemokine receptors as therapeutic targets in chronic obstructive pulmonary disease. Trends Pharmacol Sci. 2006;27:546–553. doi: 10.1016/j.tips.2006.08.001.
    1. Gaggar A, Jackson PL, Noerager BD, O’Reilly PJ, McQuaid DB, Rowe SM, Clancy JP, Blalock JE. A novel proteolytic cascade generates an extracellular matrix-derived chemoattractant in chronic neutrophilic inflammation. J Immunol. 2008;180:5662–5669.
    1. Xu X, Jackson PL, Tanner S, Hardison MT, Abdul RM, Blalock JE, Gaggar A. A self-propagating matrix metalloprotease-9 (MMP-9) dependent cycle of chronic neutrophilic inflammation. PLoS One. 2011;6:e15781. doi: 10.1371/journal.pone.0015781.
    1. Traves SL, Smith SJ, Barnes PJ, Donnelly LE. Increased migration of monocytes from COPD patients towards GROα is not mediated by an increase in CXCR2 receptor expression. Am J Resp Crit Care Med. 2003;165(Suppl):A82.
    1. Donnelly LE, Barnes PJ. Chemokine receptor CXCR2 antagonism to prevent airway inflammation. Drugs Future. 2011;36:465–472.
    1. Gernez Y, Tirouvanziam R, Chanez P. Neutrophils in chronic inflammatory airway diseases: can we target them and how? Eur Respir J. 2010;35:467–469. doi: 10.1183/09031936.00186109.
    1. Holz O, Khalilieh S, Ludwig-Sengpiel A, Watz H, Stryszak P, Soni P, Tsai M, Sadeh J, Magnussen H. SCH527123, a novel CXCR2 antagonist, inhibits ozone-induced neutrophilia in healthy subjects. Eur Respir J. 2010;35:564–570. doi: 10.1183/09031936.00048509.
    1. Lazaar AL, Sweeney LE, MacDonald AJ, Alexis NE, Chen C, Tal-Singer R. SB-656933, a novel CXCR2 selective antagonist, inhibits ex vivo neutrophil activation and ozone-induced airway inflammation in humans. Br J Clin Pharmacol. 2011;72:282–293. doi: 10.1111/j.1365-2125.2011.03968.x.
    1. Michel O, Nagy AM, Schroeven M, Duchateau J, Neve J, Fondu P, Sergysels R. Dose–response relationship to inhaled endotoxin in normal subjects. Am J Respir Crit Care Med. 1997;156:1157–1164. doi: 10.1164/ajrccm.156.4.97-02002.
    1. Nightingale JA, Rogers DF, Hart LA, Kharitonov SA, Chung KF, Barnes PJ. Effect of inhaled endotoxin on induced sputum in normal, atopic and asthmatic subjects. Thorax. 1998;53:563–571. doi: 10.1136/thx.53.7.563.
    1. Michel O. Systemic and local airways inflammatory response to endotoxin. Toxicology. 2000;152:25–30. doi: 10.1016/S0300-483X(00)00288-2.
    1. Loh LC, Vyas B, Kanabar V, Kemeny DM, O’Connor BJ. Inhaled endotoxin in healthy human subjects: a dose-related study on systemic effects and peripheral CD4+ and CD8+ T cells. Respir Med. 2006;100:519–528. doi: 10.1016/j.rmed.2005.06.003.
    1. Goncalves AS, Appelberg R. The involvement of the chemokine receptor CXCR2 in neutrophil recruitment in LPS-induced inflammation and in Mycobacterium avium infection. Scand J Immunol. 2002;55:585–591. doi: 10.1046/j.1365-3083.2002.01097.x.
    1. Chapman RW, Minnicozzi M, Celly CS, Phillips JE, Kung TT, Hipkin RW, Fan X, Rindgen D, Deno G, Bond R, Gonsiorek W, Billah MM, Fine JS, Hey JA. A novel, orally active CXCR1/2 receptor antagonist, Sch527123, inhibits neutrophil recruitment, mucus production, and goblet cell hyperplasia in animal models of pulmonary inflammation. J Pharmacol Exp Ther. 2007;322:486–493. doi: 10.1124/jpet.106.119040.
    1. Virtala R, Ekman AK, Jansson L, Westin U, Cardell LO. Airway inflammation evaluated in a human nasal lipopolysaccharide challenge model by investigating the effect of a CXCR2 inhibitor. Clin Exp Allergy. 2012;42:590–596. doi: 10.1111/j.1365-2222.2011.03921.x.
    1. O’Grady NP, Preas HL, Pugin J, Fiuza C, Tropea M, Reda D, Banks SM, Suffredini AF. Local inflammatory responses following bronchial endotoxin instillation in humans. Am J Respir Crit Care Med. 2001;163:1591–1598. doi: 10.1164/ajrccm.163.7.2009111.
    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. doi: 10.1189/jlb.1003495.
    1. Magnussen H, Watz H, Sauer M, Khanskaya I, Gann L, Stryszak P, Staudiger H, Sadeh H. Safety and efficacy of SCH527123, a novel CXCR2 antagonist, in patients with COPD. Eur Resp J. 2010;36(suppl):38S.
    1. Rennard SL, Dale DC, Donohue JF, Magnusseen H, Sutehrlans ER, Watyz H, Lu S, Strysak P, Rosenburg E, Staudinger H. CXCR2 Antagonist MK-7123 – a phase 2 proof of concept trial for chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;187:A6071.
    1. Nair P, Gaga M, Zervas E, Alagha K, Hargreave FE, O’Byrne PM, Stryszak P, Gann L, Sadeh J, Chanez P. Study investigators. Safety and efficacy of a CXCR2 antagonist in patients with severe asthma and sputum neutrophils: a randomized, placebo-controlled clinical trial. Clin Exp Allergy. 2012;42:1097–1103. doi: 10.1111/j.1365-2222.2012.04014.x.
    1. Chapman RW, Phillips JE, Hipkin RW, Curran AK, Lundell D, Fine JS. CXCR2 antagonists for the treatment of pulmonary disease. Pharmacol Ther. 2009;121:55–68. doi: 10.1016/j.pharmthera.2008.10.005.
    1. Widdowson KL, Elliott JD, Veber DF, Nie H, Rutledge MC, McCleland BW, Xiang JN, Jurewicz AJ, Hertzberg RP, Foley JJ, Griswold DE, Martin L, Lee JM, White JR, Sarau HM. Evaluation of potent and selective small-molecule antagonists for the CXCR2 chemokine receptor. J Med Chem. 2004;47:1319–1321. doi: 10.1021/jm034248l.
    1. Hansel TT, Barnes PJ. New drugs for exacerbations of chronic obstructive pulmonary disease. Lancet. 2009;374:744–755. doi: 10.1016/S0140-6736(09)61342-8.

Source: PubMed

3
구독하다