Low-dose endotoxin inhalation in healthy volunteers--a challenge model for early clinical drug development

Ole Janssen, Frank Schaumann, Olaf Holz, Bianca Lavae-Mokhtari, Lutz Welker, Carla Winkler, Heike Biller, Norbert Krug, Jens M Hohlfeld, Ole Janssen, Frank Schaumann, Olaf Holz, Bianca Lavae-Mokhtari, Lutz Welker, Carla Winkler, Heike Biller, Norbert Krug, Jens M Hohlfeld

Abstract

Background: Inhalation of endotoxin (LPS) induces a predominantly neutrophilic airway inflammation and has been used as model to test the anti-inflammatory activity of novel drugs. In the past, a dose exceeding 15-50 μg was generally needed to induce a sufficient inflammatory response. For human studies, regulatory authorities in some countries now request the use of GMP-grade LPS, which is of limited availability. It was therefore the aim of this study to test the effect and reproducibility of a low-dose LPS challenge (20,000 E.U.; 2 μg) using a flow- and volume-controlled inhalation technique to increase LPS deposition.

Methods: Two to four weeks after a baseline sputum induction, 12 non-smoking healthy volunteers inhaled LPS on three occasions, separated by at least 4 weeks. To modulate the inflammatory effect of LPS, a 5-day PDE4 inhibitor (Roflumilast) treatment preceded the last challenge. Six hours after each LPS inhalation, sputum induction was performed.

Results: The low-dose LPS inhalation was well tolerated and increased the mean percentage of sputum neutrophils from 25% to 72%. After the second LPS challenge, 62% neutrophils and an increased percentage of monocytes were observed. The LPS induced influx of neutrophils and the cumulative inflammatory response compared with baseline were reproducible. Treatment with Roflumilast for 5 days did not have a significant effect on sputum composition.

Conclusion: The controlled inhalation of 2 μg GMP-grade LPS is sufficient to induce a significant neutrophilic airway inflammation in healthy volunteers. Repeated low-dose LPS challenges potentially result in a small shift of the neutrophil/monocyte ratio; however, the cumulative response is reproducible, enabling the use of this model for "proof-of-concept" studies for anti-inflammatory compounds during early drug development.

Trial registration: ClinicalTrials.gov NCT01400568.

Figures

Figure 1
Figure 1
Study design. LPS: inhalation of 2 μg (20,000 E.U.) nebulized Lipopolysaccharide. Treat: Oral administration (500 μg/day) of the PDE-4 inhibitor Roflumilast. FACS: Flow cytometry of sputum cells was performed. In a separate study performed >56 days after the end of the LPS challenge trial, 11 subjects underwent a follow-up sputum induction. (Visits 4, 6, and 9 refer to phone calls done 24 h after the respective challenges).
Figure 2
Figure 2
Procedures performed on challenge days LPS 1, LPS 2 and LPS Tx (Figure 1). FEV1 = lung function measurement, EBT = exhaled breath temperature, BT = body temperature, LPS = low dose lipopolysaccharide challenge (20,000 E.U.), 1 blood samples were not taken at visit 3 (LPS1). Lung function was also monitored by a portable AM1 detector hourly for 6 h, as well as 9, 11, 13, and 24 h after LPS challenge. Lung function results and the subject’s symptoms at 24 h were assessed by phone call.
Figure 3
Figure 3
Sputum neutrophils (left) and the sum of sputum monocytes and small macrophages (right). Individual data and mean values of percent sputum leukocytes are displayed. BL: baseline, LPS 1: first LPS challenge, LPS 2: second LPS challenge at least 4 weeks after LPS 1, LPS Tx: third LPS challenge at least 4 weeks after LPS 2 and after 5 days of treatment with Roflumilast (500 μg /day). For statistical details please refer to Table 2. ** p < 0.01, *** p < 0.001 compared with baseline; # p < 0.05 compared with LPS 1.
Figure 4
Figure 4
Concentrations of IL-8 and MPO in sputum supernatants. Individual data and mean values of log transformed values are displayed. BL: baseline, LPS 1: first LPS challenge, LPS: second LPS challenge at least 4 weeks after LPS 1, LPS Tx: third LPS challenge at least 4 weeks after LPS 2 and after 5 days of treatment with roflumilast (500 μg /day). For statistical details please refer to Table 2. *** p < 0.001 compared with baseline.
Figure 5
Figure 5
Reproducibility of the low dose LPS induced inflammatory response (LPS 1 vs. LPS 2). Top row (A, B): Change in the percentage of sputum neutrophils compared with baseline. Bottom row (C, D): Change in the percentage of the sum of neutrophils, monocytes and small macrophages compared with baseline. On the left (A, C) Bland-Altman plots with lines indicating the mean and 2*SD of the differences and on the right (B, D) the respective correlations with the line of identity.

References

    1. Rylander R. Endotoxin in the environment–exposure and effects. J Endotoxin Res. 2002;8:241–252.
    1. Hasday JD, Bascom R, Costa JJ, Fitzgerald T, Dubin W. Bacterial endotoxin is an active component of cigarette smoke. Chest. 1999;115:829–835. doi: 10.1378/chest.115.3.829.
    1. Carty CL, Gehring U, Cyrys J, Bischof W, Heinrich J. Seasonal variability of endotoxin in ambient fine particulate matter. J Environ Monit. 2003;5:953–958. doi: 10.1039/b308488d.
    1. Hohlfeld JM, Schoenfeld K, Lavae-Mokhtari M, Schaumann F, Mueller M, Bredenbroeker D, Krug N, Hermann R. Roflumilast attenuates pulmonary inflammation upon segmental endotoxin challenge in healthy subjects: a randomized placebo-controlled trial. Pulm Pharmacol Ther. 2008;21:616–623. doi: 10.1016/j.pupt.2008.02.002.
    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. 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. Thorn J. The inflammatory response in humans after inhalation of bacterial endotoxin: a review. Inflamm Res. 2001;50:254–261. doi: 10.1007/s000110050751.
    1. Alexis NE, Lay JC, Almond M, Bromberg PA, Patel DD, Peden DB. Acute LPS inhalation in healthy volunteers induces dendritic cell maturation in vivo. J Allergy Clin Immunol. 2005;115:345–350. doi: 10.1016/j.jaci.2004.11.040.
    1. Hernandez ML, Harris B, Lay JC, Bromberg PA, Diaz-Sanchez D, Devlin RB, Kleeberger SR, Alexis NE, Peden DB. Comparative airway inflammatory response of normal volunteers to ozone and lipopolysaccharide challenge. Inhal Toxicol. 2010;22:648–656. doi: 10.3109/08958371003610966.
    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. Michel O, Ginanni R, Le BB, Content J, Duchateau J, Sergysels R. Inflammatory response to acute inhalation of endotoxin in asthmatic patients. Am Rev Respir Dis. 1992;146:352–357. doi: 10.1164/ajrccm/146.2.352.
    1. Alexis NE, Brickey WJ, Lay JC, Wang Y, Roubey RA, Ting JP, Peden DB. Development of an inhaled endotoxin challenge protocol for characterizing evoked cell surface phenotype and genomic responses of airway cells in allergic individuals. Ann Allergy Asthma Immunol. 2008;100:206–215. doi: 10.1016/S1081-1206(10)60444-9.
    1. Kitz R, Rose MA, Borgmann A, Schubert R, Zielen S. Systemic and bronchial inflammation following LPS inhalation in asthmatic and healthy subjects. J Endotoxin Res. 2006;12:367–374. doi: 10.1179/096805106X153934.
    1. Aul R, Armstrong J, Duvoix A, Lomas D, Hayes B, Miller BE, Jagger C, Singh D. Inhaled LPS challenges in smokers: a study of pulmonary and systemic effects. Br J Clin Pharmacol. 2012;74(6):1023–32. doi: 10.1111/j.1365-2125.2012.04287.x.
    1. Maris NA, de Vos AF, Dessing MC, Spek CA, Lutter R, Jansen HM, van der Zee JS, Bresser P, van der Poll T. Antiinflammatory effects of salmeterol after inhalation of lipopolysaccharide by healthy volunteers. Am J Respir Crit Care Med. 2005;172:878–884. doi: 10.1164/rccm.200503-451OC.
    1. Wallin A, Pourazar J, Sandstrom T. LPS-induced bronchoalveolar neutrophilia; effects of salmeterol treatment. Respir Med. 2004;98:1087–1092. doi: 10.1016/j.rmed.2004.03.025.
    1. Michel O, Dentener M, Cataldo D, Cantinieaux B, Vertongen F, Delvaux C, Murdoch RD. Evaluation of oral corticosteroids and phosphodiesterase-4 inhibitor on the acute inflammation induced by inhaled lipopolysaccharide in human. Pulm Pharmacol Ther. 2007;20:676–683. doi: 10.1016/j.pupt.2006.08.002.
    1. Doyen V, Kassengera Z, Dinh DH, Michel O. Time course of endotoxin-induced airways’ inflammation in healthy subjects. Inflammation. 2010;35:33–38.
    1. Djukanovic R, Sterk PJ, Fahy JV, Hargreave FE. Standardised methodology of sputum induction and processing. Eur Respir J. 2002;20:1S–55S.
    1. Brand P, Friemel I, Meyer T, Schulz H, Heyder J, Haubetainger K. Total deposition of therapeutic particles during spontaneous and controlled inhalations. J Pharm Sci. 2000;89:724–731. doi: 10.1002/(SICI)1520-6017(200006)89:6<724::AID-JPS3>;2-B.
    1. Holz O, Tal-Singer R, Kanniess F, Simpson KJ, Gibson A, Vessey RS, Janicki S, Magnussen H, Jorres RA, Richter K. Validation of the human ozone challenge model as a tool for assessing anti-inflammatory drugs in early development. J Clin Pharmacol. 2005;45:498–503. doi: 10.1177/0091270004273527.
    1. Fleiss JL. Design and Analysis of Clinical Experiments. New York: Wiley-Interscience; 1986. pp. 8–13.
    1. Lay JC, Peden DB, Alexis NE. Flow cytometry of sputum: assessing inflammation and immune response elements in the bronchial airways. Inhal Toxicol. 2011;23:392–406. doi: 10.3109/08958378.2011.575568.
    1. Fan H, Cook JA. Molecular mechanisms of endotoxin tolerance. J Endotoxin Res. 2004;10:71–84.
    1. West MA, Heagy W. Endotoxin tolerance: A review. Crit Care Med. 2002;30:S64–S73. doi: 10.1097/00003246-200201001-00009.
    1. Kharitonov SA, Sjobring U. Lipopolysaccharide challenge of humans as a model for chronic obstructive lung disease exacerbations. Contrib Microbiol. 2007;14:83–100.
    1. Loh LC. Tolerance to repeat exposure of inhaled endotoxin: an observation in healthy humans. Eur Respir J. 2005;26:363–364. doi: 10.1183/09031936.05.00058405.
    1. Lahu G, Hunnemeyer A, Diletti E, Elmlinger M, Ruth P, Zech K, McCracken N, Facius A. Population pharmacokinetic modelling of roflumilast and roflumilast N-oxide by total phosphodiesterase-4 inhibitory activity and development of a population pharmacodynamic-adverse event model. Clin Pharmacokinet. 2010;49:589–606. doi: 10.2165/11536600-000000000-00000.
    1. Hermann R, Siegmund W, Giessmann T, Westphal K, Weinbrenner A, Hauns B, Reutter F, Lahu G, Zech K, Bethke TD. The oral, once-daily phosphodiesterase 4 inhibitor roflumilast lacks relevant pharmacokinetic interactions with inhaled budesonide. J Clin Pharmacol. 2007;47:1005–1013. doi: 10.1177/0091270007300950.
    1. Seehase S, Lauenstein HD, Schlumbohm C, Switalla S, Neuhaus V, Forster C, Fieguth HG, Pfennig O, Fuchs E, Kaup FJ. LPS-induced lung inflammation in marmoset monkeys - an acute model for anti-inflammatory drug testing. PLoS One. 2012;7:e43709. doi: 10.1371/journal.pone.0043709.
    1. Grootendorst DC, Gauw SA, Verhoosel RM, Sterk PJ, Hospers JJ, Bredenbroker D, Bethke TD, Hiemstra PS, Rabe KF. Reduction in sputum neutrophil and eosinophil numbers by the PDE4 inhibitor roflumilast in patients with COPD. Thorax. 2007;62:1081–1087. doi: 10.1136/thx.2006.075937.
    1. Gauvreau GM, Boulet LP, Schmid-Wirlitsch C, Cote J, Duong M, Killian KJ, Milot J, Deschesnes F, Strinich T, Watson RM. Roflumilast attenuates allergen-induced inflammation in mild asthmatic subjects. Respir Res. 2011;12:140. doi: 10.1186/1465-9921-12-140.

Source: PubMed

3
Abonnieren