Mucolytic Effectiveness of Tyloxapol in Chronic Obstructive Pulmonary Disease - A Double-Blind, Randomized Controlled Trial

Martin Koppitz, Charlotte Eschenburg, Emilia Salzmann, Martin Rosewich, Ralf Schubert, Stefan Zielen, Martin Koppitz, Charlotte Eschenburg, Emilia Salzmann, Martin Rosewich, Ralf Schubert, Stefan Zielen

Abstract

Objective: Mucoactive drugs should increase the ability to expectorate sputum and, ideally, have anti-inflammatory properties. The aim of the study was to evaluate the mucolytic activity of Tyloxapol compared to saline (0.9%) in COPD.

Design: A randomized, placebo-controlled, double-blinded crossover, clinical trial was carried out. Patients were randomly assigned to either inhale 5 ml Tyloxapol 1% or saline 0.9% solution three times daily for 3 weeks and vice versa for another 3 weeks. 28 patients (18 male, 10 female, 47 to 73 years old, median age 63.50) were screened, 21 were treated and 19 patients completed the study per protocol.

Results: A comparison of the two treatment phases showed that the primary endpoint sputum weight was statistically significant higher when patients inhaled Tyloxapol (mean 4.03 g, 95% CI: 2.34-5.73 g at week 3) compared to saline (mean 2.63 g, 95% CI: 1.73-3.53 g at week 3). The p-value at three weeks of treatment was 0.041 between treatment arms. Sputum cells decreased during the Tyloxapol treatment after 3 weeks, indicating that Tyloxapol might have some anti-neutrophilic properties. Lung function parameters (FVC, FEV1, RV, and RV/TLC) remained stable during the study, and no treatment effect was shown. Interestingly, there was a mean increase in all inflammatory cytokines (IL-1β, IL-6, and IL-8) during the saline treatment from day 1 to week 3, whereas during the Tyloxapol treatment, all cytokines decreased. Due to the small sample size and the large individual variation in sputum cytokines, these differences were not significant. However, analyses confirmed that Tyloxapol has significant anti-inflammatory properties in vitro. Despite the high number of inhalations (more than 1000), only 27 adverse events (20 during the Tyloxapol and seven during saline) were recorded. Eleven patients experienced AEs under Tyloxapol and six under saline treatment, which indicates that inhalation of saline or Tyloxapol is a very safe procedure.

Conclusion: Our study demonstrated that inhalation of Tyloxapol by patients with COPD is safe and superior to saline and has some anti-inflammatory effects.

Trial registration: ClinicalTrials.gov NCT02515799.

Conflict of interest statement

Competing Interests: The authors of this manuscript have the following competing interests: Stefan Zielen received fees for lectures and advisory boards from the following companies: bene-Arzneimittel GmbH (http://www.bene-arzneimittel.de), Vifor Pharma Deutschland GmbH (http://www.viforpharma.de), Novartis AG (https://www.novartis.com), GlaxoSmithKline GmbH (http://www.glaxosmithkline.de), ALK-Abelló Arzneimittel GmbH (http://www.alk-abello.com), Allergy Therapeutics (http://www.allergytherapeutics.com), Boehringer Ingelheim (https://www.boehringer-ingelheim.de), Allergopharma GmbH (https://www.allergopharma.de) and Biotest (https://www.biotest.com). Martin Rosewich received fees for lectures from the following companies: Novartis AG (https://www.novartis.com), GlaxoSmithKline (http://www.glaxosmithkline.de), Allergopharma GmbH (https://www.allergopharma.de) and Allergy Therapeutics (http://www.allergytherapeutics.com). This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Study design.
Fig 1. Study design.
Phase A patients were treated with either Tyloxapol or saline for 21 days at three inhalations a day. After a wash-out phase of 7 days, Phase B started. Patients were switched to the other medication for another 21 days.
Fig 2. Patient flow chart.
Fig 2. Patient flow chart.
28 patients were initially enrolled, 21 completed visit 1 and started treatment phase, 19 finished the study, 2 dropped out because of the following: intestinal bleeding (n = 1) and difficulties to inhale due to shortness of breath (n = 1).
Fig 3. Sputum weight before processing comparing…
Fig 3. Sputum weight before processing comparing Tyloxapol and saline.
The figures illustrate (A-C) the sputum weight of patients treated with Tyloxapol and saline before further processing at (A) day 1 (n = 19 vs. 18), (B) 1 week (n = 19 vs. 17) and (C) 3 weeks of inhalation (n = 18 vs. 16), and (D) the fitted mixed linear model for the weight of sputum secretion [g]. The black lines represent the predicted mean of each therapy arm. The grey lines represent the observed values by each test person at day 1, 7 and 21. *, p

Fig 4. Sputum weight and cells comparing…

Fig 4. Sputum weight and cells comparing smokers and non-smokers during treatment with Tyloxapol and…

Fig 4. Sputum weight and cells comparing smokers and non-smokers during treatment with Tyloxapol and saline.
(A, C and E) represent the Tyloxapol group, and (B, D and F) represent the saline group. (A-B) Sputum weight before processing. (C-D) the total cells and (E-F) the neutrophils. Sample size: A: n = 36 vs. 20, B: n = 33 vs. 18, C: n = 24 vs. 13, D: n = 22 vs. 12, E: n = 23 vs. 11, F: n = 22 vs. 9. Graphs show the pooled patients at all 3 visits and are separated by smoking status. Bars represent the median. ** p

Fig 5. Cytokine release after macrophage activation…

Fig 5. Cytokine release after macrophage activation test.

(A-D) Cytokine release, after 24-hr LPS-stimulation of…

Fig 5. Cytokine release after macrophage activation test.
(A-D) Cytokine release, after 24-hr LPS-stimulation of whole blood cells incubated with different concentrations of Tyloxapol in vitro as measured by the CBA. * p < 0.05; ** p < 0.01; *** p < 0.001.

Fig 6. Effect of Tyloxapol on cell…

Fig 6. Effect of Tyloxapol on cell viability.

Cell viability of LPS treated whole blood…

Fig 6. Effect of Tyloxapol on cell viability.
Cell viability of LPS treated whole blood cells in the presence of different concentrations of Tyloxapol (0, 0.001, 0.01, 0.1, 1.0 mg/ml) compared with hypertonic saline (3.0, 7.0%) after 24 hours was measured by 7AAD-staining using flow cytometry. (A-D) Histograms of flow cytometric analysis. (E) Quantitative analysis of four independent analysis. ** p
Similar articles
Cited by
References
    1. Vestbo J, Hogg JC. Convergence of the epidemiology and pathology of COPD. Thorax 2006; 61(1):86–8. - PMC - PubMed
    1. Fletcher C, Peto R. The natural history of chronic airflow obstruction. British medical journal 1977; 1(6077):1645–8. - PMC - PubMed
    1. Vestbo J, Prescott E, Lange P. Association of chronic mucus hypersecretion with FEV1 decline and chronic obstructive pulmonary disease morbidity. Copenhagen City Heart Study Group. American journal of respiratory and critical care medicine 1996; 153(5):1530–5. - PubMed
    1. de Marco R, Accordini S, Cerveri I, Corsico A, Antó JM, Künzli N et al. Incidence of chronic obstructive pulmonary disease in a cohort of young adults according to the presence of chronic cough and phlegm. American journal of respiratory and critical care medicine 2007; 175(1):32–9. - PubMed
    1. BRUMFITT W, WILLOUGHBY ML, BROMLEY LL. An evaluation of sputum examination in chronic bronchitis. Lancet (London, England) 1957; 273(7009):1306–9. - PubMed
Show all 57 references
Publication types
MeSH terms
Associated data
Grant support
This study was funded by bene-Arzneimittel GmbH, Munich, Germany, http://www.bene-arzneimittel.de. The authors declare that they have no other relevant conflicts of interest. The grant number was 100.000 €. The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig 4. Sputum weight and cells comparing…
Fig 4. Sputum weight and cells comparing smokers and non-smokers during treatment with Tyloxapol and saline.
(A, C and E) represent the Tyloxapol group, and (B, D and F) represent the saline group. (A-B) Sputum weight before processing. (C-D) the total cells and (E-F) the neutrophils. Sample size: A: n = 36 vs. 20, B: n = 33 vs. 18, C: n = 24 vs. 13, D: n = 22 vs. 12, E: n = 23 vs. 11, F: n = 22 vs. 9. Graphs show the pooled patients at all 3 visits and are separated by smoking status. Bars represent the median. ** p

Fig 5. Cytokine release after macrophage activation…

Fig 5. Cytokine release after macrophage activation test.

(A-D) Cytokine release, after 24-hr LPS-stimulation of…

Fig 5. Cytokine release after macrophage activation test.
(A-D) Cytokine release, after 24-hr LPS-stimulation of whole blood cells incubated with different concentrations of Tyloxapol in vitro as measured by the CBA. * p < 0.05; ** p < 0.01; *** p < 0.001.

Fig 6. Effect of Tyloxapol on cell…

Fig 6. Effect of Tyloxapol on cell viability.

Cell viability of LPS treated whole blood…

Fig 6. Effect of Tyloxapol on cell viability.
Cell viability of LPS treated whole blood cells in the presence of different concentrations of Tyloxapol (0, 0.001, 0.01, 0.1, 1.0 mg/ml) compared with hypertonic saline (3.0, 7.0%) after 24 hours was measured by 7AAD-staining using flow cytometry. (A-D) Histograms of flow cytometric analysis. (E) Quantitative analysis of four independent analysis. ** p
Similar articles
Cited by
References
    1. Vestbo J, Hogg JC. Convergence of the epidemiology and pathology of COPD. Thorax 2006; 61(1):86–8. - PMC - PubMed
    1. Fletcher C, Peto R. The natural history of chronic airflow obstruction. British medical journal 1977; 1(6077):1645–8. - PMC - PubMed
    1. Vestbo J, Prescott E, Lange P. Association of chronic mucus hypersecretion with FEV1 decline and chronic obstructive pulmonary disease morbidity. Copenhagen City Heart Study Group. American journal of respiratory and critical care medicine 1996; 153(5):1530–5. - PubMed
    1. de Marco R, Accordini S, Cerveri I, Corsico A, Antó JM, Künzli N et al. Incidence of chronic obstructive pulmonary disease in a cohort of young adults according to the presence of chronic cough and phlegm. American journal of respiratory and critical care medicine 2007; 175(1):32–9. - PubMed
    1. BRUMFITT W, WILLOUGHBY ML, BROMLEY LL. An evaluation of sputum examination in chronic bronchitis. Lancet (London, England) 1957; 273(7009):1306–9. - PubMed
Show all 57 references
Publication types
MeSH terms
Associated data
Grant support
This study was funded by bene-Arzneimittel GmbH, Munich, Germany, http://www.bene-arzneimittel.de. The authors declare that they have no other relevant conflicts of interest. The grant number was 100.000 €. The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig 5. Cytokine release after macrophage activation…
Fig 5. Cytokine release after macrophage activation test.
(A-D) Cytokine release, after 24-hr LPS-stimulation of whole blood cells incubated with different concentrations of Tyloxapol in vitro as measured by the CBA. * p < 0.05; ** p < 0.01; *** p < 0.001.
Fig 6. Effect of Tyloxapol on cell…
Fig 6. Effect of Tyloxapol on cell viability.
Cell viability of LPS treated whole blood cells in the presence of different concentrations of Tyloxapol (0, 0.001, 0.01, 0.1, 1.0 mg/ml) compared with hypertonic saline (3.0, 7.0%) after 24 hours was measured by 7AAD-staining using flow cytometry. (A-D) Histograms of flow cytometric analysis. (E) Quantitative analysis of four independent analysis. ** p

References

    1. Vestbo J, Hogg JC. Convergence of the epidemiology and pathology of COPD. Thorax 2006; 61(1):86–8.
    1. Fletcher C, Peto R. The natural history of chronic airflow obstruction. British medical journal 1977; 1(6077):1645–8.
    1. Vestbo J, Prescott E, Lange P. Association of chronic mucus hypersecretion with FEV1 decline and chronic obstructive pulmonary disease morbidity. Copenhagen City Heart Study Group. American journal of respiratory and critical care medicine 1996; 153(5):1530–5.
    1. de Marco R, Accordini S, Cerveri I, Corsico A, Antó JM, Künzli N et al. Incidence of chronic obstructive pulmonary disease in a cohort of young adults according to the presence of chronic cough and phlegm. American journal of respiratory and critical care medicine 2007; 175(1):32–9.
    1. BRUMFITT W, WILLOUGHBY ML, BROMLEY LL. An evaluation of sputum examination in chronic bronchitis. Lancet (London, England) 1957; 273(7009):1306–9.
    1. Kohansal R, Martinez-Camblor P, Agustí A, Buist AS, Mannino DM, Soriano JB. The natural history of chronic airflow obstruction revisited: an analysis of the Framingham offspring cohort. American journal of respiratory and critical care medicine 2009; 180(1):3–10. 10.1164/rccm.200901-0047OC
    1. Decramer M, Janssens W. Mucoactive therapy in COPD. European respiratory review: an official journal of the European Respiratory Society 2010; 19(116):134–40.
    1. Balsamo R, Lanata L, Egan CG. Mucoactive drugs. European respiratory review: an official journal of the European Respiratory Society 2010; 19(116):127–33.
    1. Poole P, Chong J, Cates CJ. Mucolytic agents versus placebo for chronic bronchitis or chronic obstructive pulmonary disease. The Cochrane database of systematic reviews 2015; 7:CD001287 10.1002/14651858.CD001287.pub5
    1. Marc Decramer MD. Global Strategy for Diagnosis, Management, and Prevention of COPD (Updated 2015) [cited 2015 Aug 21]. Available from: URL:.
    1. HUTSCHENREUTER K. Die Bedeutung moderner Netzmittel für die Chirurgie. Der Chirurg; Zeitschrift für alle Gebiete der operativen Medizen 1958; 29(6):252–8.
    1. WAGENEDER FM. [Tacholiquin in the pre- and postoperative phase of thoracic surgical interventions]. Münchener medizinische Wochenschrift (1950) 1962; 104:2296–8.
    1. PALMER KN. A new mucolytic agent by aerosol for inhalation in chronic bronchitis. Lancet (London, England) 1961; 2(7206):802–4.
    1. RAVENEL SF. New technique of humidification in pediatrics. Journal of the American Medical Association 1953; 151(9):707–11.
    1. CONLEY JJ. Diagnosis and treatment of encrustations in the trachea: their relation to radical surgery of the head and neck. Journal of the American Medical Association 1954; 154(10):829–32.
    1. TAINTER ML, NACHOD FC, BIRD JG. Alevaire as a mycolytic agent. The New England journal of medicine 1955; 253(18):764–7.
    1. Paez PN, Miller WF. Surface Active Agents in Sputum Evacuation: A Blind Comparison with Normal Saline Solution and Distilled Water. Chest 1971; 60(4).
    1. Wilde A. Oberflächenspannungs- und Viskositäts-Messungen an Sputa unter dem Einfluss von Tacholiquin. Notabene Medici 1978; (8. Heft 1.).
    1. FARBER SM, BENIOFF MA, SMITH JD. Planned care for patients with bronchiectasis. California medicine 1955; 82(2):102–6.
    1. Ghio AJ, Marshall BC, Diaz JL, Hasegawa T, Samuelson W, Povia D et al. Tyloxapol inhibits NF-kappa B and cytokine release, scavenges HOCI, and reduces viscosity of cystic fibrosis sputum. American journal of respiratory and critical care medicine 1996; 154(3 Pt 1):783–8.
    1. Criée CP, Sorichter S, Smith HJ, Kardos P, Merget R, Heise D et al. Body plethysmography—its principles and clinical use. Respiratory Medicine 2011; 105(7):959–71. 10.1016/j.rmed.2011.02.006
    1. Schulze J, Voss S, Zissler U, Rose MA, Zielen S, Schubert R. Airway responses and inflammation in subjects with asthma after four days of repeated high-single-dose allergen challenge. Respiratory research 2012; 13:78
    1. Woolhouse IS, Bayley DL, Stockley RA. Effect of sputum processing with dithiothreitol on the detection of inflammatory mediators in chronic bronchitis and bronchiectasis. Thorax 2002; 57(8):667–71.
    1. Rosewich M, Zissler UM, Kheiri T, Voss S, Eickmeier O, Schulze J et al. Airway inflammation in children and adolescents with bronchiolitis obliterans. Cytokine 2015; 73(1):156–62. 10.1016/j.cyto.2014.10.026
    1. Schmitt-Grohé S, Hippe V, Igel M, von Bergmann K, Posselt HG, Krahl A et al. Lipopolysaccharide binding protein, cytokine production in whole blood, and lipoproteins in cystic fibrosis. Pediatric research 2005; 58(5):903–7.
    1. Eickmeier O, Huebner M, Herrmann E, Zissler U, Rosewich M, Baer PC et al. Sputum biomarker profiles in cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) and association between pulmonary function. Cytokine 2010; 50(2):152–7. 10.1016/j.cyto.2010.02.004
    1. Kellett F, Robert NM. Nebulised 7% hypertonic saline improves lung function and quality of life in bronchiectasis. Respiratory Medicine 2011; 105(12):1831–5. 10.1016/j.rmed.2011.07.019
    1. Dijkstra AE, de Jong K, Boezen HM, Kromhout H, Vermeulen R, Groen HJM et al. Risk factors for chronic mucus hypersecretion in individuals with and without COPD: influence of smoking and job exposure on CMH. Occupational and environmental medicine 2014; 71(5):346–52. 10.1136/oemed-2013-101654
    1. Cerveri I, Brusasco V. Revisited role for mucus hypersecretion in the pathogenesis of COPD. European respiratory review: an official journal of the European Respiratory Society 2010; 19(116):109–12.
    1. Allegra L, Cordaro CI, Grassi C. Prevention of acute exacerbations of chronic obstructive bronchitis with carbocysteine lysine salt monohydrate: a multicenter, double-blind, placebo-controlled trial. Respiration; international review of thoracic diseases 1996; 63(3):174–80.
    1. Guyatt GH, Townsend M, Kazim F, Newhouse MT. A controlled trial of ambroxol in chronic bronchitis. Chest 1987; 92(4):618–20.
    1. Petty TL. The National Mucolytic Study. Results of a randomized, double-blind, placebo-controlled study of iodinated glycerol in chronic obstructive bronchitis. Chest 1990; 97(1):75–83.
    1. Celli BR, MacNee W. Standards for the diagnosis and treatment of patients with COPD: a summary of the ATS/ERS position paper. The European respiratory journal 2004; 23(6):932–46.
    1. Kellett F, Redfern J, McL Niven R. Evaluation of nebulised hypertonic saline (7%) as an adjunct to physiotherapy in patients with stable bronchiectasis. Respiratory Medicine 2005; 99(1):27–31.
    1. Bilton D, Daviskas E, Anderson SD, Kolbe J, King G, Stirling RG et al. Phase 3 randomized study of the efficacy and safety of inhaled dry powder mannitol for the symptomatic treatment of non-cystic fibrosis bronchiectasis. Chest 2013; 144(1):215–25. 10.1378/chest.12-1763
    1. Dueholm M, Nielsen C, Thorshauge H, Evald T, Hansen NC, Madsen HD et al. N-acetylcysteine by metered dose inhaler in the treatment of chronic bronchitis: a multi-centre study. Respiratory Medicine 1992; 86(2):89–92.
    1. Gallon AM. Evaluation of nebulised acetylcysteine and normal saline in the treatment of sputum retention following thoracotomy. Thorax 1996; 51(4):429–32.
    1. Sathe NA, Krishnaswami S, Andrews J, Ficzere C, McPheeters ML. Pharmacologic Agents That Promote Airway Clearance in Hospitalized Subjects: A Systematic Review. Respiratory care 2015; 60(7):1061–70. 10.4187/respcare.04086
    1. Faner R, Gonzalez N, Cruz T, Kalko SG, Agustí A. Systemic inflammatory response to smoking in chronic obstructive pulmonary disease: evidence of a gender effect. PloS one 2014; 9(5):e97491 10.1371/journal.pone.0097491
    1. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. The New England journal of medicine 2004; 350(26):2645–53.
    1. Donaldson SH, Bennett WD, Zeman KL, Knowles MR, Tarran R, Boucher RC. Mucus clearance and lung function in cystic fibrosis with hypertonic saline. The New England journal of medicine 2006; 354(3):241–50.
    1. Robinson M, Hemming AL, Regnis JA, Wong AG, Bailey DL, Bautovich GJ et al. Effect of increasing doses of hypertonic saline on mucociliary clearance in patients with cystic fibrosis. Thorax 1997; 52(10):900–3.
    1. Poole PJ, Black PN. Oral mucolytic drugs for exacerbations of chronic obstructive pulmonary disease: systematic review. BMJ (Clinical research ed.) 2001; 322(7297):1271–4.
    1. Decramer M, Rutten-van Mölken M, Dekhuijzen PR, Troosters T, van Herwaarden C, Pellegrino R et al. Effects of N-acetylcysteine on outcomes in chronic obstructive pulmonary disease (Bronchitis Randomized on NAC Cost-Utility Study, BRONCUS): A randomised placebo-controlled trial. The Lancet 2005; 365(9470):1552–60.
    1. Ayfer Aytemur Z, Baysak A, Ozdemir O, Köse T, Sayiner A. N-acetylcysteine in patients with COPD exacerbations associated with increased sputum. Wiener klinische Wochenschrift 2015; 127(7–8):256–61. 10.1007/s00508-014-0692-4
    1. Daviskas E, Anderson SD, Gomes K, Briffa P, Cochrane B, Chan H et al. Inhaled mannitol for the treatment of mucociliary dysfunction in patients with bronchiectasis: effect on lung function, health status and sputum. Respirology (Carlton, Vic.) 2005; 10(1):46–56.
    1. Robinson M, Daviskas E, Eberl S, Baker J, Chan HK, Anderson SD et al. The effect of inhaled mannitol on bronchial mucus clearance in cystic fibrosis patients: a pilot study. The European respiratory journal 1999; 14(3):678–85.
    1. Shah PL, Scott SF, Knight RA, Marriott C, Ranasinha C, Hodson ME. In vivo effects of recombinant human DNase I on sputum in patients with cystic fibrosis. Thorax 1996; 51(2):119–25.
    1. McCoy K, Hamilton S, Johnson C. Effects of 12-week administration of dornase alfa in patients with advanced cystic fibrosis lung disease. Pulmozyme Study Group. Chest 1996; 110(4):889–95.
    1. O'Donnell AE, Barker AF, Ilowite JS, Fick RB. Treatment of idiopathic bronchiectasis with aerosolized recombinant human DNase I. rhDNase Study Group. Chest 1998; 113(5):1329–34.
    1. Nicolson CHH, Stirling RG, Borg BM, Button BM, Wilson JW, Holland AE. The long term effect of inhaled hypertonic saline 6% in non-cystic fibrosis bronchiectasis. Respiratory Medicine 2012; 106(5):661–7. 10.1016/j.rmed.2011.12.021
    1. Tse HN, Raiteri L, Wong KY, Yee KS, Ng LY, Wai KY et al. High-dose N-acetylcysteine in stable COPD: the 1-year, double-blind, randomized, placebo-controlled HIACE study. Chest 2013; 144(1):106–18. 10.1378/chest.12-2357
    1. Sutton PP, Gemmell HG, Innes N, Davidson J, Smith FW, Legge JS et al. Use of nebulised saline and nebulised terbutaline as an adjunct to chest physiotherapy. Thorax 1988; 43(1):57–60.
    1. Poole PJ, Brodie SM, Stewart JM, Black PN. The effects of nebulised isotonic saline and terbutaline on breathlessness in severe chronic obstructive pulmonary disease (COPD). Australian and New Zealand journal of medicine 1998; 28(3):322–6.
    1. Khan SY, O'Driscoll BR. Is nebulized saline a placebo in COPD? BMC pulmonary medicine 2004; 4:9
    1. Cegla UH. Langzeittherapie über 2 Jahre mit Ambroxol (Mucosolvan) Retardkapseln bei Patienten mit chronischer Bronchitis. Ergebnisse einer Doppelblindstudie an 180 Patienten. Praxis und Klinik der Pneumologie 1988; 42(9):715–21.
    1. Olivieri D, Zavattini G, Tomasini G, Daniotti S, Bonsignore G, Ferrara G et al. Ambroxol for the prevention of chronic bronchitis exacerbations: long-term multicenter trial. Protective effect of ambroxol against winter semester exacerbations: a double-blind study versus placebo. Respiration; international review of thoracic diseases 1987; 51 Suppl 1:42–51.

Source: PubMed

Подписаться