Combination therapy of tiotropium and ciclesonide attenuates airway inflammation and remodeling in a guinea pig model of chronic asthma

Loes E M Kistemaker, I Sophie T Bos, Mark H Menzen, Harm Maarsingh, Herman Meurs, Reinoud Gosens, Loes E M Kistemaker, I Sophie T Bos, Mark H Menzen, Harm Maarsingh, Herman Meurs, Reinoud Gosens

Abstract

Background: The long-acting anticholinergic tiotropium has recently been registered for the treatment of asthma, and its use is associated with a reduction in exacerbation frequency. Anti-inflammatory and anti-remodeling effects of tiotropium have been demonstrated in in vitro and in vivo models. Because tiotropium treatment is used in combination with inhaled corticosteroids, potential additive effects between the two would be clinically relevant. Therefore, the aim of this study was to investigate additive effects between tiotropium and ciclesonide on airway inflammation and remodeling in guinea pig models of asthma.

Methods: Guinea pigs (n = 3-8/group) were sensitized and challenged with ovalbumin in an acute (single challenge) and a chronic model (12 weekly challenges) of allergic asthma. Animals were treated with vehicle, nebulized tiotropium (0.01-0.3 mM) and/or intranasally instilled ciclesonide (0.001-1 mg/kg) before each challenge. Bronchoalveolar lavage fluid and lungs were collected for analysis of airway inflammation and remodeling.

Results: Tiotropium and ciclesonide treatment, alone or in combination, did not inhibit airway inflammation in the acute asthma model. In a dose-finding study, low doses of tiotropium and ciclesonide inhibited airway eosinophilia and airway smooth muscle thickening in the chronic asthma model. Threshold doses of 0.01 mM tiotropium (nebulizer concentration) and 0.01 mg/kg ciclesonide were selected to investigate potential additive effects between both drugs. At these doses, tiotropium and ciclesonide did not inhibit airway eosinophilia or airway smooth muscle thickening when administered alone, but significantly inhibited these allergen-induced responses when administered in combination.

Conclusions: Combined treatment with low doses of tiotropium and ciclesonide inhibits airway inflammation and remodeling in a guinea pig model of chronic asthma, suggesting that combined treatment with anticholinergics and corticosteroids may have anti-inflammatory and anti-remodeling activity in allergic airway diseases. Since tiotropium is registered as a therapy for asthma added on to corticosteroid treatment, these beneficial effects of the combination therapy may be clinically relevant.

Figures

Fig. 1
Fig. 1
Experimental procedure. a acute protocol, b chronic protocol. Male Dunkin Hartley guinea pigs (n = 3–8 animals per group, see Additional file 1: Table S1-S3 for overview of groups) were sensitized to ovalbumin (OVA) by intraperitoneal injection of 0.5 ml allergen solution containing 100 μg/ml ovalbumin and 100 mg/ml Al(OH)3, and intracutaneous injection of 0.5 ml allergen solution. Subsequently, guinea pigs were challenged with OVA (0.05–0.1 %) via inhalation of aerosolized solution. Tiotropium (tio; 0.01–0.3 mM; 3 min inhalation time) was administered via aerosol inhalation and ciclesonide (cicl; 0.001–1 mg/kg) via intranasal instillation, 24 h and/or 1 h before every challenge. In the acute protocol, a bronchoalveolar lavage was performed 25 h after the ovalbumin challenge. In the chronic protocol, lungs were harvested for tissue sections 24 h after the last of 12 weekly OVA challenges
Fig. 2
Fig. 2
Inflammatory cell numbers in bronchoalveolar lavage fluid in response to a single ovalbumin (OVA) challenge and treatment with saline (sal; control), tiotropium (tio; 0.1 mM; nebulizer concentration), and/or ciclesonide (cicl; 1 mg/kg). Guinea pigs were treated as described in Fig. 1a. A bronchoalveolar lavage was performed 25 h after OVA challenge and inflammatory cells were determined. a total cells, b eosinophils, c macrophages, d lymphocytes, e neutrophils. ** p < 0.01, *** p < 0.001. Data represent mean ± s.e.m. of 5–8 animals per group
Fig. 3
Fig. 3
Airway eosinophilia in response to chronic ovalbumin (OVA) challenge and treatment with saline (sal; control), tiotropium (tio; 0.01 and 0.03 mM; nebulizer concencenrations) or ciclesonide (cicl; 0.01 and 0.1 mg/kg). Guinea pigs were treated as described in Fig. 1b. Lungs were collected 24 h after the last OVA challenge and eosinophil numbers were determined by H&E staining in the submucosa (a,c) and adventitia (b,d) of the non-cartilaginous (a,b) and cartilaginous airways (c,d). * p < 0.05, ** p < 0.01, *** p < 0.001. Data represent mean ± s.e.m. of 4 animals per group
Fig. 4
Fig. 4
Airway smooth muscle mass thickening in response to chronic ovalbumin (OVA) challenge and treatment with saline (sal, control), tiotropium (tio; 0.01 and 0.03 mM; nebulizer concentration) or ciclesonide (cicl; 0.01 and 0.1 mg/kg). Guinea pigs were treated as described in Fig. 1b. Lungs were collected 24 h after the last OVA challenge and airway smooth muscle mass was determined by α-sm-myosin antibody staining of non-cartilaginous (a) and cartilaginous airways (b). * p < 0.05. Data represent mean ± s.e.m. of 4 animals per group
Fig. 5
Fig. 5
Airway eosinophilia in response to chronic ovalbumin (OVA) challenge and treatment with saline (sal; control), tiotropium (tio; 0.01 mM; nebulizer concentration) and/or ciclesonide (cicl; 0.01 mg/kg). Guinea pigs were treated as described in Fig. 1b. Lungs were collected 24 h after the last OVA challenge and eosinophil numbers were determined by H&E staining in the submucosa (a, i) and adventitia (b,j) of non-cartilaginous (a,b) and cartilaginous (i,j) airways. Representative images are shown in panels c-h for non-cartilaginous airways (magnification 200x) and in panels K-P for cartilaginous airways (maginification 100x). * p < 0.05, ** p < 0.01. Data represent mean ± s.e.m. of 8 animals per group
Fig. 6
Fig. 6
Airway smooth muscle mass thickening in response to chronic ovalbumin (OVA) challenge and treatment with saline (sal; control), tiotropium (tio; 0.01 mM; nebulizer concentration) and/or ciclesonide (cicl; 0.01 mg/kg). Guinea pigs were treated as described in Fig. 1b. Lungs were collected 24 h after the last OVA challenge and airway smooth muscle mass was determined by α-sm-myosin antibody staining. Quantification is shown in figure a for non-cartilaginous airways and representative images are shown in panels b-g (magnification 200x). * p < 0.05. Data represent mean ± s.e.m. of 8 animals per group

References

    1. Global Initiative for Asthma (GINA). GINA Report, Global Burden of Asthma. Available from: . May, 2004; June 4, 2014.
    1. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. 2008;8(3):183–92. doi: 10.1038/nri2254.
    1. An SS, Bai TR, Bates JH, Black JL, Brown RH, Brusasco V, et al. Airway smooth muscle dynamics: a common pathway of airway obstruction in asthma. Eur Respir J. 2007;29(5):834–60. doi: 10.1183/09031936.00112606.
    1. Pare PD, Roberts CR, Bai TR, Wiggs BJ. The functional consequences of airway remodeling in asthma. Monaldi Arch Chest Dis. 1997;52(6):589–596.
    1. Kerstjens HA, Engel M, Dahl R, Paggiaro P, Beck E, Vandewalker M, et al. Tiotropium in asthma poorly controlled with standard combination therapy. N Engl J Med. 2012;367(13):1198–207. doi: 10.1056/NEJMoa1208606.
    1. Kerstjens HA, Casale TB, Bleecker ER, Meltzer EO, Pizzichini E, Schmidt O, et al. Tiotropium or salmeterol as add-on therapy to inhaled corticosteroids for patients with moderate symptomatic asthma: two replicate, double-blind, placebo-controlled, parallel-group, active-comparator, randomised trials. Lancet Respir Med. 2015;3(5):367–76. doi: 10.1016/S2213-2600(15)00031-4.
    1. Beeh KM, Moroni-Zentgraf P, Ablinger O, Hollaenderova Z, Unseld A, Engel M, et al. Tiotropium Respimat(R) in asthma: a double-blind, randomised, dose-ranging study in adult patients with moderate asthma. Respir Res. 2014;15(1):61. doi: 10.1186/1465-9921-15-61.
    1. Kistemaker LE, Gosens R. Acetylcholine beyond bronchoconstriction: roles in inflammation and remodeling. Trends Pharmacol Sci. 2015;36(3):164–71. doi: 10.1016/j.tips.2014.11.005.
    1. Kistemaker LE, Oenema TA, Meurs H, Gosens R. Regulation of airway inflammation and remodeling by muscarinic receptors: perspectives on anticholinergic therapy in asthma and COPD. Life Sci. 2012;91(21-22):1126–33. doi: 10.1016/j.lfs.2012.02.021.
    1. Razani-Boroujerdi S, Behl M, Hahn FF, Pena-Philippides JC, Hutt J, Sopori ML. Role of muscarinic receptors in the regulation of immune and inflammatory responses. J Neuroimmunol. 2008;194(1-2):83–88. doi: 10.1016/j.jneuroim.2007.11.019.
    1. Buhling F, Lieder N, Kuhlmann UC, Waldburg N, Welte T. Tiotropium suppresses acetylcholine-induced release of chemotactic mediators in vitro. Respir Med. 2007;101(11):2386–394. doi: 10.1016/j.rmed.2007.06.009.
    1. Profita M, Bonanno A, Siena L, Ferraro M, Montalbano AM, Pompeo F, et al. Acetylcholine mediates the release of IL-8 in human bronchial epithelial cells by a NFkB/ERK-dependent mechanism. Eur J Pharmacol. 2008;582(1-3):145–153. doi: 10.1016/j.ejphar.2007.12.029.
    1. Gosens R, Rieks D, Meurs H, Ninaber DK, Rabe KF, Nanninga J, et al. Muscarinic M3 receptor stimulation increases cigarette smoke-induced IL-8 secretion by human airway smooth muscle cells. Eur Respir J. 2009;34(1399-3003; 0903-1936; 6):1436-1443.
    1. Gosens R, Dueck G, Rector E, Nunes RO, Gerthoffer WT, Unruh H, et al. Cooperative regulation of GSK-3 by muscarinic and PDGF receptors is associated with airway myocyte proliferation. Am J Physiol Lung Cell Mol Physiol. 2007;293(5):L1348–L1358. doi: 10.1152/ajplung.00346.2007.
    1. Matthiesen S, Bahulayan A, Kempkens S, Haag S, Fuhrmann M, Stichnote C, et al. Muscarinic receptors mediate stimulation of human lung fibroblast proliferation. Am J Respir Cell Mol Biol. 2006;35(6):621–27. doi: 10.1165/rcmb.2005-0343RC.
    1. Milara J, Serrano A, Peiro T, Gavalda A, Miralpeix M, Morcillo EJ, et al. Aclidinium inhibits human lung fibroblast to myofibroblast transition. Thorax. 2012;67(3):229–237. doi: 10.1136/thoraxjnl-2011-200376.
    1. Haag S, Matthiesen S, Juergens UR, Racke K. Muscarinic receptors mediate stimulation of collagen synthesis in human lung fibroblasts. Eur Respir J. 2008;32(3):555–62. doi: 10.1183/09031936.00129307.
    1. Oenema TA, Mensink G, Smedinga L, Halayko AJ, Zaagsma J, Meurs H, et al. Cross-talk between transforming growth factor-beta(1) and muscarinic M(2) receptors augments airway smooth muscle proliferation. Am J Respir Cell Mol Biol. 2013;49(1):18–27. doi: 10.1165/rcmb.2012-0261OC.
    1. Ohta S, Oda N, Yokoe T, Tanaka A, Yamamoto Y, Watanabe Y, et al. Effect of tiotropium bromide on airway inflammation and remodelling in a mouse model of asthma. Clin Exp Allergy. 2010;40(1365-2222; 0954-7894; 8):1266-275.
    1. Kistemaker LE, Bos ST, Mudde WM, Hylkema MN, Hiemstra PS, Wess J, et al. Muscarinic m3 receptors contribute to allergen-induced airway remodeling in mice. Am J Respir Cell Mol Biol. 2014;50(4):690–8. doi: 10.1165/rcmb.2013-0220OC.
    1. Gosens R, Bos IS, Zaagsma J, Meurs H. Protective effects of tiotropium bromide in the progression of airway smooth muscle remodeling. Am J Respir Crit Care Med. 2005;171(10):1096–102. doi: 10.1164/rccm.200409-1249OC.
    1. Bos IS, Gosens R, Zuidhof AB, Schaafsma D, Halayko AJ, et al. Inhibition of allergen-induced airway remodelling by tiotropium and budesonide: a comparison. Eur Respir J. 2007;30(4):653–61. doi: 10.1183/09031936.00004907.
    1. Pahl A, Bauhofer A, Petzold U, Cnota PJ, Maus J, Brune K, et al. Synergistic effects of the anti-cholinergic R, R-glycopyrrolate with anti-inflammatory drugs. Biochem Pharmacol. 2006;72(12):1690–696. doi: 10.1016/j.bcp.2006.07.025.
    1. Meurs H, Santing RE, Remie R, van der Mark TW, Westerhof FJ, Zuidhof AB, et al. A guinea pig model of acute and chronic asthma using permanently instrumented and unrestrained animals. Nat Protoc. 2006;1(2):840–7. doi: 10.1038/nprot.2006.144.
    1. Bosnjak B, Tilp C, Tomsic C, Dekan G, Pieper MP, Erb KJ, et al. Tiotropium bromide inhibits relapsing allergic asthma in BALB/c mice. Pulm Pharmacol Ther. 2014;27(1):44–51. doi: 10.1016/j.pupt.2013.09.004.
    1. Peters SP, Kunselman SJ, Icitovic N, Moore WC, Pascual R, Ameredes BT, et al. National Heart, Lung, and Blood Institute Asthma Clinical Research Network: Tiotropium bromide step-up therapy for adults with uncontrolled asthma. N Engl J Med. 2010;363(18):1715–26. doi: 10.1056/NEJMoa1008770.
    1. Barnes PJ. Glucocorticosteroids: current and future directions. Br J Pharmacol. 2011;163(1):29–43. doi: 10.1111/j.1476-5381.2010.01199.x.
    1. Cortijo J, Mata M, Milara J, Donet E, Gavalda A, Miralpeix M, et al. Aclidinium inhibits cholinergic and tobacco smoke-induced MUC5AC in human airways. Eur Respir J. 2011;37(2):244–254. doi: 10.1183/09031936.00182009.
    1. Kistemaker LE, Hiemstra PS, Bos IS, Bouwman S, van den Berge M, Hylkema MN, et al. Tiotropium attenuates IL-13-induced goblet cell metaplasia of human airway epithelial cells. Thorax. 2015;70(7):668–76. doi: 10.1136/thoraxjnl-2014-205731.
    1. Gosens R, Grainge C. Bronchoconstriction and airway biology: potential impact and therapeutic opportunities. Chest. 2015;147(3):798–803. doi: 10.1378/chest.14-1142.
    1. Noble PB, Pascoe CD, Lan B, Ito S, Kistemaker LE, Tatler AL. at al. Airway smooth muscle in asthma: Linking contraction and mechanotransduction to disease pathogenesis and remodelling. Pulm Pharmacol Ther. 2014;29(2):96–107. doi: 10.1016/j.pupt.2014.07.005.
    1. Grainge CL, Lau LC, Ward JA, Dulay V, Lahiff G, Wilson S, et al. Effect of bronchoconstriction on airway remodeling in asthma. N Engl J Med. 2011;364(21):2006–15. doi: 10.1056/NEJMoa1014350.
    1. Fisher JT, Vincent SG, Gomeza J, Yamada M, Wess J. Loss of vagally mediated bradycardia and bronchoconstriction in mice lacking M2 or M3 muscarinic acetylcholine receptors. FASEB J. 2004;18(6):711–3.
    1. Oenema TA, Maarsingh H, Smit M, Groothuis GM, Meurs H, Gosens R. Bronchoconstriction Induces TGF-beta Release and Airway Remodelling in Guinea Pig Lung Slices. PLoS One. 2013;8(6) doi: 10.1371/journal.pone.0065580.
    1. Smit M, Zuidhof AB, Bos SI, Maarsingh H, Gosens R, Zaagsma J, et al. Bronchoprotection by olodaterol is synergistically enhanced by tiotropium in a guinea pig model of allergic asthma. J Pharmacol Exp Ther. 2014;348(2):303–10. doi: 10.1124/jpet.113.208439.
    1. Racke K, Juergens UR, Matthiesen S. Control by cholinergic mechanisms. Eur J Pharmacol. 2006;533(1-3):57–68. doi: 10.1016/j.ejphar.2005.12.050.
    1. Raemdonck K, de Alba J, Birrell MA, Grace M, Maher SA, Irvin CG, et al. A role for sensory nerves in the late asthmatic response. Thorax. 2012;67(1):19–25. doi: 10.1136/thoraxjnl-2011-200365.
    1. Gosens R, Roscioni SS, Dekkers BG, Pera T, Schmidt M, Schaafsma D, et al. Pharmacology of airway smooth muscle proliferation. Eur J Pharmacol. 2008;585(2-3):385–97. doi: 10.1016/j.ejphar.2008.01.055.

Source: PubMed

3
Subscribe