Laryngorhinootologie 2015; 94(S 01): S32-S63
DOI: 10.1055/s-0034-1396870
Referat
© Georg Thieme Verlag KG Stuttgart · New York

Pathophysiologie der chronischen Rhinosinusitis, konservative Therapieoptionen

Pathophysiology of Chronic Rhinosinusitis, Pharmaceutical Therapy Options
C. Bachert
1   HNO-Abteilung und Upper Airways Research Laboratory, Universität Ghent
2   Division of ENT Diseases, CLINTEC, Karolinska Institute, University of Stockholm, Sweden
,
G. Holtappels
1   HNO-Abteilung und Upper Airways Research Laboratory, Universität Ghent
› Institutsangaben
Weitere Informationen

Publikationsverlauf

Publikationsdatum:
10. April 2015 (online)

Zusammenfassung

Die immunologische Forschung hat in den letzten 2 Jahrzehnten enorme Fortschritte gemacht, die auch für die oberen Atemwege von großer Bedeutung sind. Unser Verständnis zur Pathophysiologie der chronischen Rhinosinusitis hat sich damit von eher mechanistischen Betrachtungen bzgl. Engstellen und mukoziliärem Transport zu einer neuen sehr komplexen immunologischen Sichtweise entwickelt. Wir differenzieren heute mehrerer Entzündungsformen, wir beginnen komplexe Netzwerke aus immunologischen Faktoren und die Gründe einer Fehlregulation zu verstehen, und haben daraus bereits erste innovative Therapieansätze entwickelt, die v. a. den am schwersten betroffenen Patienten zugutekommen können. Durch die neuen Erkenntnisse zu Entzündungsformen und Gewebeumbau und vor allem die Identifikation der Schlüsselfaktoren ergeben sich Konsequenzen für die Diagnostik und die Therapie der chronischen Rhinosinusitis; die Einteilung in Endotypen entsprechend den die Erkrankung treibenden Entzündungsmediatoren ist unabdingbar für einen sinnvollen Einsatz der innovativen Arzneimittel, meist in Form von humanisierten monoklonalen Antikörpern. Mehrere hundert solcher Antikörper sind derzeit in verschiedenen Indikationen bereits im Einsatz oder in Entwicklung und werden unser Fach ebenso wie z. B. die Pneumologie entscheidend beeinflussen.

Abstract

Research in immunology has brought great progress in knowledge of inflammatory processes in the last 2 decades, which also has an impact on the upper airways. Our understanding of the pathophysiology of chronic rhinosinusitis developed from a rather mechanistic point of view with a focus on narrow clefts and mucociliary clearance to the appreciation of a complex network of immunological pathways forming the basis of disease. We today differentiate various forms of inflammation, we start to understand complex immune-regulatory networks and the reasons for their failure, and have already developed innovative approaches for therapy for the most severely ill subjects. Due to this new knowledge in inflammation and remodeling processes within mucosal tissue, specifically on the key driving factors, new diagnostic tools and therapeutic approaches for chronic rhinosinusitis have developed; the differentiation of endotypes based on pathophysiological principles will be crucial for the use of innovative therapies, mostly humanized monoclonal antibodies. Several hundred of those antibodies are currently developed for various indications and will impact our speciality as well as pneumology to a great extent.

 
  • Literatur

  • 1 Fokkens WJ, Lund VJ, Mullol J, Bachert C, Alobid I, Baroody F, Cohen N, Cervin A, Douglas R, Gevaert P, Georgalas C, Goossens H, Harvey R, Hellings P, Hopkins C, Jones N, Joos G, Kalogjera L, Kern B, Kowalski M, Price D, Riechelmann H, Schlosser R, Senior B, Thomas M, Toskala E, Voegels R, Wang de Y, Wormald PJ. European Position Paper on Rhinosinusitis and Nasal Polyps 2012. Rhinol Suppl. 2012; 23: 1-298
  • 2 Bachert C, Van Bruaene N, Toskala E, Zhang N, Olze H, Scadding G, Van Drunen CM, Mullol J, Cardell LO, Gevaert P, Van Zele T, Claeys S, Halldén C, Kostamo K, Foerster U, Kowalski M, Bieniek K, Olszewska-Ziaber A, Nizankowska-Mogilnicka E, Szczeklik A, Swierczynska M, Arcimowicz M, Lund V, Fokkens W, Zuberbier T, Akdis C, Canonica G, van Cauwenberge P, Burney P, Bousquet J. Important research questions in allergy and related diseases: Chronic rhinosinusitis and nasal polyposis: A GA2LEN paper. Allergy 2009; 64: 520-533
  • 3 Tomassen P, Newson RB, Hoffmans R, Fokkens W, Bachert C, Burney P, Jarvis D. on behalf of the GA2LEN Survey. Reliability of symptom criteria and nasal endoscopy in the assessment of chronic rhinosinusitis based on EP3OS-guidelines – a GA²LEN study. Allergy 2011; 66: 556-561
  • 4 Hastan D, Fokkens WJ, Bachert C, Newson RB, Bislimovska J, Bockelbrink A, Bousquet PJ, Brozek G, Bruno A, Dahlén SE, Forsberg B, Gunnbjörnsdóttir M, Kasper L, Kramer U, Kowalski ML, Lange B, Lundbäck B, Salagean E, Todo-Bom A, Tomassen P, Toskala E, Bousquet J, Zuberbier T, Jarvis D, Burney P. Chronic rhinosinusitis in Europe – an underestimated disease. A GA2LEN in study. Allergy 2011; 66: 1216-1223
  • 5 Hamilos DL. Chronic rhinosinusitis: epidemiology and medical management. J Allergy Clin Immunol 2011; 128: 693-707
  • 6 Jarvis D, Newson R, Lotvall J, Hastan D, Tomassen P, Keil T, Gjomarkaj M, Forsberg B, Gunnbjornsdottir M, Minov J, Brozek G, Dahlen SE, Toskala E, Kowalski ML, Olze H, Howarth P, Kramer U, Baelum J, Loureiro C, Kasper L, Bousquet PJ, Bousquet J, Bachert C, Fokkens W, Burney P. Asthma in adults and its association with chronic rhinosinusitis: the GA2LEN survey in Europe. Allergy 2012; 67: 91-98
  • 7 Moore WC, Meyers DA, Wenzel SE, Teague WG, Li H, Li X, D’agostino Jr R, Castro M., Curran-Everett D, Fitzpatrick AM, Gaston B, Jarjour NN, Sorkness R, Calhoun WJ, Chung KF, Comhair SA, Dweik RA, Israel E, Peters SP, Busse WW, Erzurum SC, Bleecker ER. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med 2010; 181: 315-323
  • 8 Dixon AE, Kaminsky DA, Holbrook JT, Wise RA, Shade DM, Irvin CG. Allergic rhinitis and sinusitis in asthma: differential effects on symptoms and pulmonary function. Chest 2006; 130: 429-435
  • 9 Van Zele T, Holtappels G, Gevaert P, Bachert C. Differences in initial immunoprofiles between recurrent and nonrecurrent chronic rhinosinusitis with nasal polyps. Am J Rhinol Allergy 2014; 28: 192-198
  • 10 Bosse Y, Bacot F, Montpetit A, Rung J, Qu HQ, Engert JC et al. Identification of susceptibility genes for complex diseases using pooling-based genome-wide association scans. Hum Genet 2009; 125: 305-318
  • 11 Hsu J, Avila PC, Kern RC, Hayes MG, Schleimer RP, Pinto JM. Genetics of chronic rhinosinusitis: state of the field and directions forward. J Allergy Clin Immunol 2013; 131: 977-993
  • 12 Henmyr V, Vandeplas G, Halldén C, Säll T, Olze H, Bachert C, Cardell LO. Replication study of genetic variants associated with chronic rhinosinusitis and nasal polyposis. J Allergy Clin Immunol 2014; 133: 273-253 Letter
  • 13 Kim SH, Park HS, Holloway JW, Shin HD, Park CS. Association between a TGFbeta1 promoter polymorphism and rhinosinusitis in aspirin-intolerant asthmatic patients. Respir Med 2007; 101: 490-495
  • 14 Zhang Y, Mfuna Endam L, Filali-Mouhim A, Bosse Y, Castano R, Desrosiers M. Polymorphisms in the nitric oxide synthase 1 gene are associated with severe chronic rhinosinusitis. Am J Rhinol Allergy 2011; 25: e49-e54
  • 15 Zuo L, Koozechian MS, Chen LL. Characterization of reactive nitrogen species in allergic asthma. Ann Allergy Asthma Immunol 2014; 112: 18-22
  • 16 Lee RJ, Chen B, Redding KM, Margolskee RF, Cohen NA. Mouse nasal epithelial innate immune responses to Pseudomonas aeruginosa quorum-sensing molecules require taste signaling components. Innate Immun 2013; 20: 606-617
  • 17 Zhang Y, Mfuna Endam L, Filali-Mouhim A, Zhao L, Desrosiers M, Han D et al. Polymorphisms in RYBP and AOAH genes are associated with chronic rhinosinusitis in a Chinese population: a replication study. PLoS ONE 2012; 7: e39247
  • 18 Barnes KC, Grant A, Gao P, Baltadjieva D, Berg T, Chi P et al. Polymorphisms in the novel gene acyloxyacyl hydroxylase (AOAH) are associated with asthma and associated phenotypes. J Allergy Clin Immunol 2006; 118: 70-77
  • 19 Cheong HS, Park SM, Kim MO, Park JS, Lee JY, Byun JY, Park BL, Shin HD, Park CS. Genome-wide methylation profile of nasal polyps: relation to aspirin hypersensitivity in asthmatics. Allergy 2011; 66: 637-644
  • 20 Seiberling KA, Church CA, Herring JL, Sowers LC. Epigenetics of chronic rhinosinusitis and the role of the eosinophil. International forum of allergy & rhinology 2012; 2: 80-84
  • 21 Van Bruaene N, Derycke L, Perez-Novo CA, Gevaert P, Holtappels G, De Ruyck N, Cuvelier C, Van Cauwenberge P, Bachert C. TGF-beta signaling and collagen deposition in chronic rhinosinusitis. J Allergy Clin Immunol 2009; 124: 253-259
  • 22 Cho JS, Moon YM, Park IH, Um JY, Moon JH, Park SJ, Lee SH, Kang HJ, Lee HM. Epigenetic regulation of myofibroblast differentiation and extracellular matrix production in nasal polyp-derived fibroblasts. Clin Exp Allergy 2012; 42: 872-882
  • 23 Zhang XH, Zhang YN, Li HB, Hu CY, Wang N, Cao PP, Liao B, Lu X, Cui YH, Liu Z. Overexpression of miR-125b, a novel regulator of innate immunity, in eosinophilic chronic rhinosinusitis with nasal polyps. Am J Respir Crit Care Med 2012; 185: 140-151
  • 24 Johansson ME, Larsson JM, Hansson GC. The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host-microbial interactions. Proc Natl Acad Sci USA 2011; 108: 4659-4665
  • 25 Zhang Y, Wang XD, Wang H, Jiao J, Li Y, Fan E, Zhang L, Bachert C. TMEM16A-mediated mucin secretion in IL-13-induced nasal epithelial cells from chronic rhinosinusitis patients. AAIR 2014;
  • 26 Avila P. Airway Epithelium. In: Kay AB, Kaplan AP, Bousquet J, Holt PG. (Eds.). Allergy and allergic diseases. Second ed. Wiley-Blackwell; Oxford, UK: 2008
  • 27 Laudien M, Dressel S, Harder J, Glaser R. Differential expression pattern of antimicrobial peptides in nasal mucosa and secretion. Rhinology 2001; 49: 107-111
  • 28 Meyer JE, Harder J, Sipos B, Maune S, Kloppel G, Bartels J, Schroder JM, Glaser R. Psoriasin (S100A7) is a principal antimicrobial peptide of the human tongue. Mucosal Immunol 2008; 1: 239-243
  • 29 Ramanathan Jr M, Lee WK, Spannhake EW, Lane AP. Th2 cytokines associated with chronic rhinosinusitis with polyps down-regulate the antimicrobial immune function of human sinonasal epithelial cells. Am J Rhinol 2008; 22: 115-121
  • 30 Aujla SJ, Chan YR, Zheng M, Fei M, Askew DJ, Pociask DA, Reinhart TA, Mcallister F, Edeal J, Gaus K, Husain S, Kreindler JL, Dubin PJ, Pilewski JM, Myerburg MM, Mason CA, Iwakura Y, Kolls JK. IL-22 mediates mucosal host defense against Gram-negative bacterial pneumonia. Nat Med 2008; 14: 275-281
  • 31 Pickert G, Neufert C, Leppkes M, Zheng Y, Wittkopf N, Warntjen M, Lehr HA, Hirth S, Weigmann B, Wirtz S, Ouyang W, Neurath MF, Becker C. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J Exp Med 2009; 206: 1465-1472
  • 32 Ramanathan Jr M, Spannhake EW, Lane AP. Chronic rhinosinusitis with nasal polyps is associated with decreased expression of mucosal interleukin 22 receptor. Laryngoscope 2007; 117: 1839-1843
  • 33 Peters M, Dudziak K, Stiehm M, Bufe A. T-cell polarization depends on concentration of the danger signal used to activate dendritic cells. Immunol Cell Biol 2010; 88: 537-544
  • 34 Lee RJ, Kofonow JM, Rosen PL, Siebert AP, Chen B, Doghramji L, Xiong G, Adappa ND, Palmer JN, Kennedy DW, Kreindler JL, Margolskee RF, Cohen NA. Bitter and sweet taste receptors regulate human upper respiratory innate immunity. J Clin Invest 2014; 124: 1393-1405
  • 35 Rogers GA, Den Beste K, Parkos CA, Nusrat A, Delgaudio JM, Wise SK. Epithelial tight junction alterations in nasal polyposis. International forum of allergy & rhinology 2011; 1: 50-54
  • 36 Zuckerman JD, Lee WY, Delgaudio JM, Moore CE, Nava P, Nusrat A, Parkos CA. Pathophysiology of nasal polyposis: the role of desmosomal junctions. Am J Rhinol 2008; 22: 589-597
  • 37 Tieu DD, Peters AT, Carter RG, Suh L, Conley DB, Chandra R, Norton J, Grammer LC, Harris KE, Kato A, Kern RC, Schleimer RP. Evidence for diminished levels of epithelial psoriasin and calprotectin in chronic rhinosinusitis. J Allergy Clin Immunol 2010; 125: 667-675
  • 38 Soyka MB, Wawrzyniak P, Eiwegger T, Holzmann D, Treis A, Wanke K, Kast JI, Akdis CA. Defective epithelial barrier in chronic rhinosinusitis: the regulation of tight junctions by IFN-gamma and IL-4. J Allergy Clin Immunol 2012; 130: 1087-1096
  • 39 Meng J, Zhou P, Liu Y, Liu F, Yi X, Liu S, Holtappels G, Bachert C, Zhang N. The development of nasal polyp disease involves early nasal mucosal inflammation and remodeling. PLoS One 2013; 8: e82373
  • 40 Schleimer RP, Kato A, Kern R, Kuperman D, Avila PC. Epithelium: at the interface of innate and adaptive immune responses. J Allergy Clin Immunol 2007; 120: 1279-1284
  • 41 Claeys S, De Belder T, Holtappels G, Gevaert P, Verhasselt B, Van Cauwenberge P, Bachert C. Human beta-defensins and toll-like receptors in the upper airway. Allergy 2003; 58: 748-753
  • 42 Vandermeer J, Sha Q, Lane AP, Schleimer RP. Innate immunity of the sinonasal cavity: expression of messenger RNA for complement cascade components and toll-like receptors. Arch Otolaryngol Head Neck Surg 2004; 130: 1374-1380
  • 43 Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011; 34: 637-650
  • 44 Lane AP, Truong-Tran QA, Myers A, Bickel C, Schleimer RP. Serum amyloid A, properdin, complement 3, and toll-like receptors are expressed locally in human sinonasal tissue. Am J Rhinol 2006; 20: 117-123
  • 45 Zhao CY, Wang X, Liu M, Jin DJ. Microarray gene analysis of Toll-like receptor signaling elements in chronic rhinosinusitis with nasal polyps. Int Arch Allergy Immunol 2011; 156: 297-304
  • 46 Sun Y, Zhou B, Wang C, Huang Q, Zhang Q, Han Y, Dai W, Fan E, Li Y. Biofilm formation and Toll-like receptor 2, Toll-like receptor 4, and NF-kappaB expression in sinus tissues of patients with chronic rhinosinusitis. AM J Rhino Allergy 2012; 26: 104-109
  • 47 Kato A, Schleimer RP. Beyond inflammation: airway epithelial cells are at the interface of innate and adaptive immunity. Curr Opin Immunol 2007; 19: 711-720
  • 48 Bogefors J, Rydberg C, Uddman R, Fransson M, Mansson A, Benson M, Adner M, Cardell LO. Nod1, Nod2 and Nalp3 receptors, new potential targets in treatment of allergic rhinitis?. Allergy 2010; 65: 1222-1226
  • 49 Rudack C, Steinhoff M, Mooren F, Buddenkotte J, Becker K, Von Eiff C, Sachse F. PAR-2 activation regulates IL-8 and GRO-alpha synthesis by NF-kappaB, but not RANTES, IL-6, eotaxin or TARC expression in nasal epithelium. Clin Exp Allergy 2007; 37: 1009-1022
  • 50 Ossovskaya VS, Bunnett NW. Protease-activated receptors: contribution to physiology and disease. Physiol Rev 2004; 84: 579-621
  • 51 Vroling AB, Fokkens WJ, Van Drunen CM. How epithelial cells detect danger: aiding the immune response. Allergy 2008; 63: 1110-1123
  • 52 Rudack C, Sachse F, Albert N, Becker K, Von Eiff C. Immunomodulation of nasal epithelial cells by Staphylococcus aureus-derived serine proteases. J Immunol 2009; 183: 7592-7601
  • 53 Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol 2004; 5: 987-995
  • 54 Oppenheim JJ, Yang D. Alarmins: chemotactic activators of immune responses. Curr Opin Immunol 2005; 17: 359-365
  • 55 Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol 2007; 81: 1-5
  • 56 Lotze MT, Zeh HJ, Rubartelli A, Sparvero LJ, Amoscato AA, Washburn NR, Devera ME, Liang X, Tor M, Billiar T. The grateful dead: damage-associated molecular pattern molecules and reduction/oxidation regulate immunity. Immunol Rev 2007; 220: 60-81
  • 57 Lotze MT, Zeh HJ, Rubartelli A, Sparvero LJ, Amoscato AA, Washburn NR, Devera ME, Liang X, Tor M, Billiar T. The grateful dead: damage-associated molecular pattern molecules and reduction/oxidation regulate immunity. Immunol Rev 2007; 220: 60-81
  • 58 Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediators Inflamm 2010;
  • 59 Kerkhoff C, Sorg C, Tandon NN, Nacken W. Interaction of S100A8/S100A9-arachidonic acid complexes with the scavenger receptor CD36 may facilitate fatty acid uptake by endothelial cells. Biochemistry 2001; 40: 241-248
  • 60 Vogl T, Tenbrock K, Ludwig S, Leukert N, Ehrhardt C, van Zoelen MA et al. Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock. Nat Med 2007; 13: 1042-1049
  • 61 Ghavami S, Rashedi I, Dattilo BM, Eshraghi M, Chazin WJ, Hashemi M et al. S100A8/A9 at low concentration promotes tumor cell growth via RAGE ligation and MAP kinase-dependent pathway. J Leukoc Biol 2008; 83: 1484-1492
  • 62 Boyd JH, Kan B, Roberts H, Wang Y, Walley KR. S100A8 and S100A9 mediate endotoxin-induced cardiomyocyte dysfunction via the receptor for advanced glycation end products. Circ Res 2008; 102: 1239-1246
  • 63 Bryborn M, Adner M, Cardell LO. Psoriasin, one of several new proteins identified in nasal lavage fluid from allergic and non-allergic individuals using 2-dimensional gel electrophoresis and mass spectrometry. Respir Res 2005; 6: 118
  • 64 Leclerc E, Fritz G, Vetter SW, Heizmann CW. Binding of S100 proteins to RAGE: an update. Biochim Biophys Acta 2009; 1793: 993-1007
  • 65 Fritz G. RAGE: a single receptor fits multiple ligands. Trends Biochem Sci 2011; 36: 625-632
  • 66 Lee HM, Kang HJ, Woo JS, Chae SW, Lee SH, Hwang SJ. Upregulation of surfactant protein A in chronic rhinosinusitis. Laryngoscope 2006; 116: 328-330
  • 67 Wang X, Zhao C, Liu M. Expression and significance of surfactant A in nasal polyps of chronic rhinosinusitis]. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2010; 24: 652-654
  • 68 Woodworth BA, Wood R, Baatz JE, Schlosser RJ. Sinonasal surfactant protein A1, A2, and D gene expression in cystic fibrosis: a preliminary report. Otolaryngol Head Neck Surg 2007; 137: 34-38
  • 69 Skinner ML, Schlosser RJ, Lathers D, Neal JG, Woodworth BA, Hall J et al. Innate and adaptive mediators in cystic fibrosis and allergic fungal rhinosinusitis. Am J Rhinol 2007; 21: 538-541
  • 70 Ooi EH, Wormald PJ, Carney AS, James CL, Tan LW. Surfactant protein d expression in chronic rhinosinusitis patients and immune responses in vitro to Aspergillus and alternaria in a nasal explant model. Laryngoscope 2007; 117: 51-57
  • 71 Guillot L, Balloy V, McCormack FX, Golenbock DT, Chignard M, Si-Tahar M. Cutting edge: the immunostimulatory activity of the lung surfactant protein-A involves Toll-like receptor 4. J Immunol 2002; 168: 5989-5992
  • 72 Henning LN, Azad AK, Parsa KV, Crowther JE, Tridandapani S, Schlesinger LS. Pulmonary surfactant protein A regulates TLR expression and activity in human macrophages. J Immunol 2008; 180: 7847-7858
  • 73 Murakami S, Iwaki D, Mitsuzawa H, Sano H, Takahashi H, Voelker DR et al. Surfactant protein A inhibits peptidoglycan-induced tumor necrosis factor-alpha secretion in U937 cells and alveolar macrophages by direct interaction with toll-like receptor 2. J Biol Chem 2002; 277: 6830-6837
  • 74 Ohya M, Nishitani C, Sano H, Yamada C, Mitsuzawa H, Shimizu T et al. Human pulmonary surfactant protein D binds the extracellular domains of Toll-like receptors 2 and 4 through the carbohydrate recognition domain by a mechanism different from its binding to phosphatidylinositol and lipopolysaccharide. Biochemistry 2006; 45: 8657-8664
  • 75 Yamada C, Sano H, Shimizu T, Mitsuzawa H, Nishitani C, Himi T et al. Surfactant protein A directly interacts with TLR4 and MD-2 and regulates inflammatory cellular response. Importance of supratrimeric oligomerization. J Biol Chem 2006; 281: 21771-21780
  • 76 Kalfa VC, Spector SL, Ganz T, Cole AM. Lysozyme levels in the nasal secretions of patients with perennial allergic rhinitis and recurrent sinusitis. Ann Allergy Asthma Immunol 2004; 93: 288-292
  • 77 Bascom R, Pipkorn U, Proud D, Dunnette S, Gleich GJ, Lichtenstein LM et al. Major basic protein and eosinophil-derived neurotoxin concentrations in nasal-lavage fluid after antigen challenge: effect of systemic corticosteroids and relationship to eosinophil influx. J Allergy Clin Immunol 1989; 84: 338-346
  • 78 Yang D, Chen Q, Su SB, Zhang P, Kurosaka K, Caspi RR et al. Eosinophil-derived neurotoxin acts as an alarmin to activate the TLR2-MyD88 signal pathway in dendritic cells and enhances Th2 immune responses. J Exp Med 2008; 205: 79-90
  • 79 Bilodeau L, Boulay ME, Prince P, Boisvert P, Boulet LP. Comparative clinical and airway inflammatory features of asthmatics with or without polyps. Rhinology 2010; 48: 420-425
  • 80 Nakagawa T, Yamane H, Shigeta T, Takashima T, Nakai Y. Interaction between fibronectin and eosinophils in the growth of nasal polyps. Laryngoscope 1999; 109: 557-561
  • 81 Okamura Y, Watari M, Jerud ES, Young DW, Ishizaka ST, Rose J et al. The extra domain A of fibronectin activates Toll-like receptor 4. J Biol Chem 2001; 276: 10229-10233
  • 82 Van Crombruggen K, Holtappels G, De Ruyck N, Derycke L, Tomassen P, Bachert C. RAGE processing in chronic airway conditions: involvement of Staphylococcus aureus and ECP. J Allergy Clin Immunol 2012; 129: 1515-1521 e1518
  • 83 Rubartelli A, Lotze MT. Inside, outside, upside down: damage-associated molecular-pattern molecules (DAMPs) and redox. Trends Immunol 2007; 28: 429-436
  • 84 Hudson BI, Carter AM, Harja E, Kalea AZ, Arriero M, Yang H, Grant PJ, Schmidt AM. Identification, classification, and expression of RAGE gene splice variants. FASEB J 2008; 22: 1572-1580
  • 85 Yamakawa N, Uchida T, Matthay MA, Makita K. Proteolytic release of the receptor for advanced glycation end products from in vitro and in situ alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 2011; 300: L516-L525
  • 86 Bopp C, Bierhaus A, Hofer S, Bouchon A, Nawroth PP, Martin E, Weigand MA. Bench-to-bedside review: The inflammation-perpetuating pattern-recognition receptor RAGE as a therapeutic target in sepsis. Crit Care 2008; 12: 201
  • 87 Bierhaus A, Humpert PM, Morcos M, Wendt T, Chavakis T, Arnold B, Stern DM, Nawroth PP. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med (Berl) 2005; 83: 876-886
  • 88 Pullerits R, Brisslert M, Jonsson IM, Tarkowski A. Soluble receptor for advanced glycation end products triggers a proinflammatory cytokine cascade via beta2 integrin Mac-1. Arthritis Rheum 2006; 54: 3898-3907
  • 89 Englert JM, Hanford LE, Kaminski N, Tobolewski JM, Tan RJ, Fattman CL, Ramsgaard L, Richards TJ, Loutaev I, Nawroth PP, Kasper M, Bierhaus A, Oury TD. A role for the receptor for advanced glycation end products in idiopathic pulmonary fibrosis. Am J Pathol 2008; 172: 583-591
  • 90 Demling N, Ehrhardt C, Kasper M, Laue M, Knels L, Rieber EP. Promotion of cell adherence and spreading: a novel function of RAGE, the highly selective differentiation marker of human alveolar epithelial type I cells. Cell Tissue Res 2006; 323: 475-488
  • 91 Li X, Meng J, Liu F, Zhang N, Zhang J, Holtappels G, Luo B, Zhou P, Zheng Y, Liu S, Lin P, Bachert C. Expression of transforming growth factor-β, matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases in Chinese chronic rhinosinusitis patients with and without nasal polyps. JACI 2010; 125: 1061-1068
  • 92 Queisser MA, Kouri FM, Konigshoff M, Wygrecka M, Schubert U, Eickelberg O, Preissner KT. Loss of RAGE in pulmonary fibrosis: molecular relations to functional changes in pulmonary cell types. Am J Respir Cell Mol Biol 2008; 39: 337-345
  • 93 van Zele T, Gevaert P, Claeys G, Holtappels G, van Cauwenberge P, Bachert C. Staphylococcus aureus colonization and IgE antibody formation to enterotoxins is increased in nasal polyposis. JACI 2004; 114: 981-983
  • 94 Chen Y, Akirav EM, Chen W, Henegariu O, Moser B, Desai D, Shen JM, Webster JC, Andrews RC, Mjalli AM, Rothlein R, Schmidt AM, Clynes R, Herold KC. RAGE ligation affects T cell activation and controls T cell differentiation. J Immunol 2008; 181: 4272-4278
  • 95 Loser K, Vogl T, Voskort M, Lueken A, Kupas V, Nacken W, Klenner L, Kuhn A, Foell D, Sorokin L, Luger TA, Roth J, Beissert S. The Toll-like receptor 4 ligands Mrp8 and Mrp14 are crucial in the development of autoreactive CD8+ T cells. Nat.Med 2010; 16: 713-717
  • 96 Sparvero LJ, Asafu-Adjei D, Kang R, Tang D, Amin N, Im J, Rutledge R, Lin B, Amoscato AA, Zeh HJ, Lotze MT. RAGE (Receptor for Advanced Glycation Endproducts), RAGE ligands, and their role in cancer and inflammation. J Transl Med 2009; 7: 17
  • 97 Richer SL, Truong-Tran AQ, Conley DB, Carter R, Vermylen D, Grammer LC, Peters AT, Chandra RK, Harris KE, Kern RC, Schleimer RP. Epithelial genes in chronic rhinosinusitis with and without nasal polyps. Am J Rhinol 2008; 22: 228-234
  • 98 White ES, Baralle FE, Muro AF. New insights into form and function of fibronectin splice variants. J Pathol 2008; 216: 1-14
  • 99 Van Crombruggen K, Zhang N, Gevaert P, Tomassen P, Bachert C. Pathogenesis of chronic rhinosinusitis: inflammation. J Allergy Clin Immunol 2011; 128: 728-732
  • 100 Menzies BE. The role of fibronectin binding proteins in the pathogenesis of Staphylococcus aureus infections. Curr Opin Infect Dis 2003; 16: 225-229
  • 101 Henderson B, Nair S, Pallas J, Williams MA. Fibronectin: a multidomain host adhesin targeted by bacterial fibronectin-binding proteins. FEMS Microbiol Rev 2011; 35: 147-200
  • 102 Hammad H, Lambrecht BN. Dendritic cells and airway epithelial cells at the interface between innate and adaptive immune responses. Allergy 2011; 66: 579-587
  • 103 Hazenberg MD, Spits H. Human innate lymphoid cells. Blood 2014; 124: 700-709
  • 104 Mjösberg JM, Trifari S, Crellin NK, Peters CP, van Drunen CM, Piet B, Fokkens WJ, Cupedo T, Spits H. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat Immunol 2011; 12: 1055-1062
  • 105 Shikotra A, Choy DF, Ohri CM, Doran E, Butler C, Hargadon B, Shelley M, Abbas AR, Austin CD, Jackman J, Wu LC, Heaney LG, Arron JR, Bradding P. Increased expression of immunoreactive thymic stromal lymphopoietin in patients with severe asthma. J Allergy Clin Immunol 2012; 129: 104-111
  • 106 Barnig C, Cernadas M, Dutile S et al. Lipoxin A4 regulates natural killer cell and type 2 innate lymphoid cell activation in asthma. Sci Transl Med 2013; 5: 174
  • 107 Perez- Novo CA, Claeys C, Van Cauwenberge P, Bachert C. Prostaglandin, leukotriene and lipoxin balance in chronic rhinosinusitis with and without nasal polyposis. JACI 2005; 115: 1189-1196
  • 108 Pérez- Novo CA, Holtappels G, Vinall SL, Xue L, Zhang N, Bachert C, Pettipher R. CRTH2 mediates activation of human Th2 cells in response to PGD2 released from IgE/anti-IgE treated nasal polyp tissue. Allergy 2010; 65: 304-310
  • 109 Demedts IK, Bracke KR, Maes T, Joos GF, Brusselle GG. Different roles for human lung dendritic cell subsets in pulmonary immune defense mechanisms. Am J Respir Cell Mol Biol 2006; 35: 387-393
  • 110 Reinartz SM, Van Tongeren J, Van Egmond D, De Groot EJ, Fokkens WJ, Van Drunen CM. Dendritic cells in nasal mucosa of subjects with different allergic sensitizations. J Allergy Clin Immunol 2011; 128: 887-890
  • 111 Pezato R, Pérez-Novo C, Holtappels G, De Ruyck N, Van Crombruggen K, De Vos G, Bachert C, Derycke L. The expression of dendritic cell subsets in severe chronic rhinosinusitis with nasal polyps is altered. Immunobiology 2014; 219: 729-364
  • 112 Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol 2009; 27: 451-483
  • 113 Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 2012; 122: 787-795
  • 114 Gordon S. Alternative activation of macrophages. Nature reviews 2002; 3: 23-35
  • 115 Sobol SE, Christodoulopoulos P, Manoukian JJ, Hauber HP, Frenkiel S, Desrosiers M, Fukakusa M, Schloss MD, Hamid Q. Cytokine profile of chronic sinusitis in patients with cystic fibrosis. Arch Otolaryngol Head Neck Surg 2002; 128: 1295-1298
  • 116 Krysko O, Holtappels G, Zhang N, Kubica M, Deswarte K, Derycke L, Claeys S, Hammad H, Brusselle GG, Vandenabeele P, Krysko DV, Bachert C. Alternatively activated macrophages and impaired phagocytosis of S. aureus in chronic rhinosinusitis. Allergy 2011; 66: 396-403
  • 117 Peterson S, Poposki JA, Nagarkar DR, Chustz RT, Peters AT, Suh LA, Carter R, Norton J, Harris KE, Grammer LC, Tan BK, Chandra RK, Conley DB, Kern RC, Schleimer RP, Kato A. Increased expression of CC chemokine ligand 18 in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2012; 129: 119-127
  • 118 Van Zele T, Claeys S, Gevaert P, Van Maele G, Holtappels G, Van Cauwenberge P, Bachert C. Differentiation of chronic sinus diseases by measurement of inflammatory mediators. Allergy 2006; 61: 1280-1289
  • 119 Feng L, Zhang N, Zhang J, Krysko O, Zhang Q, Xian J, Derycke L, Qi Y, Li K, Liu S, Lin P, Bachert C. Forkhead box protein 3 in human nasal polyp regulatory T cells is regulated by the protein suppressor of cytokine signaling 3. J Allergy Clin Immunol 2013; 132: 1314-1321
  • 120 Zhang N, Van Zele T, Perez-Novo C, Van Bruaene N, Holtappels G, Deruyck N, Van Cauwenberge P, Bachert C. Different types of T-effector cells orchestrate mucosal inflammation in chronic sinus disease. J Allergy Clin Immunol 2008; 122: 961-968
  • 121 Derycke L, Eyerich S, Van Crombruggen K, Perez-Novo C, Holtappels G, Deruyck N, Gevaert P, Bachert C., Mixed T. Helper Cell Signatures In Chronic Rhinosinusitis with and without Polyps. PLoS ONE 2014; 9: e97581
  • 122 Bachert C, Zhang N, Holtappels G, De Lobel L, van Steen K. Differentiation of upper airway inflammation based on T effector cell subtypes and impact of Staphylococcus aureus enterotoxins on asthma co-morbidity. JACI 2010; 126: 962-968
  • 123 Bachert C, Wagenmann M, Hauser U, Rudack C. IL-5 synthesis is upregulated in human nasal polyp tissue. J Allergy Clin Immunol 1997; 99: 837-842
  • 124 Simon HU, Yousefi S, Schranz C, Schapowal A, Bachert C, Blaser K. Direct demonstration of delayed eosinophil apoptosis as a mechanism causing tissue eosinophilia. J Immunol 1997; 158: 3902-3908
  • 125 Gevaert P, Van Bruaene N, Cattaert T, Van Steen K, Van Zele T, Acke F, De Ruyck N, Blomme K, Sousa AR, Marshall RP, Bachert C. Mepolizumab, a humanized anti-IL-5 mAb, as a treatment option for severe nasal polyposis. J Allergy Clin Immunol 2011; 128: 989-995
  • 126 Van Zele T, Gevaert P, Holtappels G, van Cauwenberge P, Bachert C. Local immunoglobulin production in nasal polyposis is modulated by superantigens. Clin Exp Allergy 2007; 37: 1840-1847
  • 127 Gevaert P, Nouri-Aria KT, Wu H, Harper CE, Takhar P, Fear DJ, Acke F, Deruyck N, Banfield G, Kariyawasam HH, Bachert C, Durham SR, Gould HJ. Local Receptor Revision and Class Switching to IgE in Chronic Rhinosinusitis with Nasal Polyps. Allergy 2013; 68: 55-63
  • 128 Mechtcheriakova D, Sobanov J, Knittelfelder R, Bachert C, Jensen-Jarolim E. Gene expression pattern associated with activation-induced cytidine deaminase, AID, in chronic paranasal disorders. PlosOne 2011; 6: e25611
  • 129 Kato A, Peters A, Suh L, Carter R, Harris KE, Chandra R, Conley D, Grammer LC, Kern R, Schleimer RP. Evidence of a role for B cell-activating factor of the TNF family in the pathogenesis of chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2008; 121: 1385-1392
  • 130 Mackay F, Schneider P. Cracking the BAFFcode. Nat Rev Immunol 2009; 9: 491-502
  • 131 Kallies A, Nutt SL. Terminal differentiationof lymphocytes depends on Blimp-1. Curr Opin Immunol 2007; 19: 156-162
  • 132 Coker HA, Durham SR, Gould HJ. Local somatic hypermutation and class switch recombination in the nasal mucosa of allergic rhinitis patients. J Immunol 2003; 171: 5602-5610
  • 133 Tan BK, Li QZ, Suh L, Kato A, Conley DB, Chandra RK et al. Evidence for intranasalantinuclear autoantibodies in patientswith chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2011; 128: 1198-1206
  • 134 Zhang N, Holtappels G, Gevaert P, Patou J, Dhaliwal B, Gould H, Bachert C. Mucosal tissue polyclonal IgE is functional in response to allergen and SEB. Allergy 2011; 66: 141-148
  • 135 Bachert C, Zhang N. Chronic rhinosinusitis and asthma: novel understanding of the role of IgE “above atopy”. J Intern Med 2012; 272: 133-143
  • 136 Perez-Novo CA, Holtappels G, Vinall SL, Xue L, Zhang N, Bachert C, Pettipher R. CRTH2 mediates the activation of human Th2 cells in response to PGD2) released from IgE/anti-IgE treated nasal polyp tissue. Allergy 2010; 65: 304-310
  • 137 Kowalski ML, Lewandowska-Polak A, Woźniak J, Ptasińska A, Jankowski A, Wagrowska-Danilewicz M, Danilewicz M, Pawliczak R. Association of stem cell factor expression in nasal polyp epithelial cells with aspirin sensitivity and asthma. Allergy 2005; 60: 631-637
  • 138 Pawankar R, Lee KH, Nonaka M, Takizawa R. Role of mast cells and basophils in chronic rhinosinusitis. Clin Allergy Immunol 2007; 20: 93-101
  • 139 Patou J, Gevaert P, Van Zele T, Holtappels G, Van Cauwenberge P, Bachert C. Staphylococcus aureus enterotoxin B, protein A, and lipoteichoic acid stimulations in nasal polyps. J Allergy Clin Immunol 2008; 121: 110-115
  • 140 Patou J, Holtappels G, Affleck K, Gevaert P, Perez-Novo C, Van Cauwenberge P, Bachert C. Enhanced release of IgE-dependent early phase mediators from nasal polyp tissue. J Inflamm (Lond) 2009; 6: 11
  • 141 Bachert C, Gevaert P, Holtappels G, Johansson SG, Van Cauwenberge P. Total and specific IgE in nasal polyps is related to local eosinophilic inflammation. J Allergy Clin Immunol 2001; 107: 607-614
  • 142 Vlaminck S, Vauterin T, Hellings PW, Jorissen M, Acke F, Van Cauwenberge P, Bachert C, Gevaert P. The importance of local eosinophilia in the surgical outcome of chronic rhino-sinusitis: A 3-year prospective observational study. Am J Rhinol Allergy 2014; 28: 260-264
  • 143 Meyer JE, Bartels J, Gorogh T, Sticherling M, Rudack C, Ross DA et al. The role of RANTES in nasal polyposis. American journal of rhinology 2005; 19: 15-20
  • 144 Bartels J, Maune S, Meyer JE, Kulke R, Schluter C, Rowert J et al. Increased eotaxin-mRNA expression in non-atopic and atopic nasal polyps: comparison to RANTES and MCP-3 expression. Rhinology 1997; 35: 171-174
  • 145 Jahnsen FL, Haye R, Gran E, Brandtzaeg P, Johansen FE. Glucocorticosteroids inhibit mRNA expression for eotaxin, eotaxin-2, and monocyte-chemotactic protein-4 in human airway inflammation with eosinophilia. J Immunol 1999; 163: 1545-1551
  • 146 Foreman A, Holtappels G, Psaltis AJ, Jervis-Bardy J, Field J, Wormald PJ, Bachert C. Adaptive immune responses in Staphylococcus aureus biofilm associated chronic rhinosinusitis. Allergy 2011; 66: 1449-1456
  • 147 Schleimer RP, Bochner BS. The effects of glucocorticoids on human eosinophils. The Journal of allergy and clinical immunology 1994; 94: 1202-1213
  • 148 Van Zele T, Gevaert P, Holtappels G, Beule A, Wormald PJ, Mayr S, Hens G, Hellings P, Ebbens FA, Fokkens W, Van Cauwenberge P, Bachert C. Oral steroids and doxycycline: two different approaches to treat nasal polyps. JACI 2010; 125: 1069-1076
  • 149 Gevaert P, Bachert C, Holtappels G, Perez Novo C, Van der Heyden J, Fransen L, Depraetere S, Walter H, van Cauwenberge P, Tavernier J. Enhanced Soluble Interleukin 5 Receptor alpha expression in nasal polyposis. Allergy 2003; 58: 371-379
  • 150 Derycke L, Zhang N, Holtappels G, Dutré T, Bachert C. IL-17A as a regulator of neutrophil survival in nasal polyp disease of patients with and without cystic fibrosis. J Cyst Fibros 2012; 11: 193-200
  • 151 Katotomichelakis M, Tantilipikorn P, Holtappels G, De Ruyck N, Feng L, Van Zele T, Muangsomboon P, Jareonchasri P, Bunnag C, Danielides V, Hellings PW, Bachert C, Zhang N. Inflammatory patterns in upper airway disease in the same geographical area may change over time. Am J Rhinol Allergy 2013; 27: 354-360
  • 152 Serhan CN, Chiang N. Resolution phase lipid mediators of inflammation: agonists of resolution. Curr Opin Pharmacol 2013; 13: 632-640
  • 153 Van Bruaene N, Caaa PN, Van Crombruggen K, De Ruyck N, Holtappels G, Van Cauwenberge P, Gevaert P, Bachert C. Inflammation and remodelling patterns in early stage chronic rhinosinusitis. Clin Exp Allergy 2012; 42: 883-890
  • 154 Abreu NA, Nagalingam NA, Song Y, Roediger FC, Pletcher SD, Goldberg AN, Lynch SV. Sinus microbiome diversity depletion and Corynebacterium tuberculostearicum enrichment mediates rhinosinusitis. Sci Transl Med 2012; 4: 151
  • 155 Ba L, Zhang N, Lin P, Liu S, Bachert C. Bacterial colonization is associated with different inflammatory pattern in rhinosinusitis with nasal polyps. Allergy 2011; 66: 1296-1303
  • 156 Heikkinen T, Jarvinen A. The common cold. Lancet 2003; 361: 51-59
  • 157 Johnston SL, Pattemore PK, Sanderson G, Smith S, Lampe F, Josephs L, Symington P, O’toole S, Myint SH, Tyrrell DA et al. Community study of role of viral infections in exacerbations of asthma in 9-11 year old children. BMJ 1995; 310: 1225-1229
  • 158 Nicholson KG, Kent J, Ireland DC. Respiratory viruses and exacerbations of asthma in adults. BMJ 1993; 307: 982-986
  • 159 Corne JM, Marshall C, Smith S, Schreiber J, Sanderson G, Holgate ST, Johnston SL. Frequency, severity, and duration of rhinovirus infections in asthmatic and non-asthmatic individuals: a longitudinal cohort study. Lancet 2002; 359: 831-834
  • 160 Message SD, Laza-Stanca V, Mallia P, Parker HL, Zhu J, Kebadze T, Contoli M, Sanderson G, Kon OM, Papi A, Jeffery PK, Stanciu LA, Johnston SL. Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and Th1/2 cytokine and IL-10 production. Proc Natl Acad Sci USA 2008; 105: 13562-13567
  • 161 Edwards MR, Regamey N, Vareille M, Kieninger E, Gupta A, Shoemark A, Saglani S, Sykes A, Macintyre J, Davies J, Bossley C, Bush A, Johnston SL. Impaired innate interferon induction in severe therapy resistant atopic asthmatic children. Mucosal Immunol 2013; 6: 797-806
  • 162 Busse WW, Morgan WJ, Gergen PJ, Mitchell HE, Gern JE, Liu AH, Gruchalla RS, Kattan M, Teach SJ, Pongracic JA, Chmiel JF, Steinbach SF, Calatroni A, Togias A, Thompson KM, Szefler SJ, Sorkness CA. Randomized trial of omalizumab (anti-IgE) for asthma in inner-city children. N Engl J Med 2011; 364: 1005-1015
  • 163 Wang XD, Zhang N, Glorieux S, Holtappels G, Vaneechoutte M, Krysko O, Zhang L, Han DM, Nauwynck HJ, Bachert C. Herpes simplex virus type 1 infection facilitates invasion of Staphylococcus aureus into the nasal mucosa and nasal polyp tissues. PLoS One 2012; 7: e39875
  • 164 Corriveau MN, Zhang N, Holtappels G, Van Roy N, Bachert C. Detection of Staphylococcus aureus in nasal tissue with peptide nucleic acid-fluorescence in situ hybridization. American journal of rhinology & allergy 2009; 23: 461-465
  • 165 Sachse F, Becker K, Von Eiff C, Metze D, Rudack C. Staphylococcus aureus invades the epithelium in nasal polyposis and induces IL-6 in nasal epithelial cells in vitro. Allergy 2010; 65: 1430-1437
  • 166 Van Zele T, Vaneechoutte M, Holtappels G, Gevaert P, Van Cauwenberge P, Bachert C. Detection of enterotoxin DNA in Staphylococcus aureus strains obtained from the middle meatus in controls and nasal polyp patients. Am J Rhinol 2008; 22: 223-227
  • 167 Marone Superantigens and Superallergens. Basel: Karger; 2007
  • 168 Ziegler C, Goldmann O, Hobeika E, Geffers R, Peters G, Medina E. The dynamics of T cells during persistent Staphylococcus aureus infection: from antigen-reactivity to in vivo anergy. EMBO Mol Med 2011; 3: 652-666
  • 169 Herz U, Ruckert R, Wollenhaupt K, Tschernig T, Neuhaus-Steinmetz U, Pabst R, Renz H. Airway exposure to bacterial superantigen (SEB) induces lymphocyte-dependent airway inflammation associated with increased airway responsiveness–a model for non-allergic asthma. Eur J Immunol 1999; 29: 1021-1031
  • 170 Hellings PW, Hens G, Meyts I, Bullens D, Vanoirbeek J, Gevaert P, Jorissen M, Ceuppens JL, Bachert C. Aggravation of bronchial eosinophilia in mice by nasal and bronchial exposure to Staphylococcus aureus enterotoxin B. Clin Exp Allergy 2006; 36: 1063-1071
  • 171 Huvenne W, Callebaut I, Plantinga M, Vanoirbeek JA, Krysko O, Bullens DM, Gevaert P, Van Cauwenberge P, Lambrecht BN, Ceuppens JL, Bachert C, Hellings PW. Staphylococcus aureus enterotoxin B facilitates allergic sensitization in experimental asthma. Clin Exp Allergy 2010; 40: 1079-1090
  • 172 Huvenne W, Callebaut I, Reekmans K, Hens G, Bobic S, Jorissen M, Bullens D, Ceuppens J, Bachert C, Hellings P. Staphylococcus aureus enterotoxin B augments granulocyte migration and survival via airway epithelial cell activation. Allergy 2010; 65: 1013-1020 epub Feb 4
  • 173 Hofer MF, Harbeck RJ, Schlievert PM, Leung DY. Staphylococcal toxins augment specific IgE responses by atopic patients exposed to allergen. J Invest Dermatol 1999; 112: 171-176
  • 174 Hofer MF, Lester MR, Schlievert PM, Leung DY. Upregulation of IgE synthesis by staphylococcal toxic shock syndrome toxin-1 in peripheral blood mononuclear cells from patients with atopic dermatitis. Clin Exp Allergy 1995; 25: 1218-1227
  • 175 Gevaert P, Holtappels G, Johansson SG, Cuvelier C, Cauwenberge P, Bachert C. Organization of secondary lymphoid tissueand local IgE formation to Staphylococcusaureus enterotoxins in nasal polyp tissue. Allergy 2005; 60: 71-79
  • 176 Bachert C, Van Steen K, Zhang N, Holtappels G, Cattaert T, Maus B, Buhl R, Taube C, Korn S, Kowalski M, Bousquet J, Howarth P. Specific IgE against Staphylococcus aureus enterotoxins: an independent risk factor for asthma. J Allergy Clin Immunol 2012; 130: 376-381
  • 177 Tomassen P, Jarvis D, Newson R, Van Ree R, Forsberg R, Howarth P, Janson C, Kowalski ML, Krämer U, Matricardi PM, Middelveld RJM, Todo-Bom A, Toskala E, Thilsing T, Brożek G, Van Drunen C, Burney P, Bachert C. Staphylococcus aureus enterotoxin specific IgE and its association with asthma in the general population: a GA²LEN. Allergy 2013; 68: 1289-1297
  • 178 Song WJ, Chang YS, Lim MK, Yun EH, Kim SH, Kang HR, Park HW, Tomassen P, Choi MH, Min KU, Cho SH, Bachert C. Staphylococcal enterotoxin sensitization in a community-based population: a potential role in adult-onset asthma. CEA 2014; 44: 553-562
  • 179 Semic-Jusufagic A, Bachert C, Gevaert P, Holtappels G, Lowe L, Woodcock A, Simpson A, Custovic A. S. aureus sensitization and allergic disease in early childhood: population-based birth cohort study. J Allergy Clin Immunol 2007; 119: 930-936
  • 180 Hollams E, Hales B, Bachert C, Huvenne W, Parsons F, De Klerk N, Serralha M, Holt B, Ahlstedt S, Thomas W, Sly P, Holt P. Th2-associated immunity to bacteria in asthma in teenagers and susceptibility to asthma. Eur Respir J 2010; 36: 509-516 epub Jan 28
  • 181 Moore WC, Meyers DA, Wenzel SE, Teague WG, Li H, Li X, D’agostino Jr R, Castro M, Curran-Everett D, Fitzpatrick AM, Gaston B, Jarjour NN, Sorkness R, Calhoun WJ, Chung KF, Comhair SA, Dweik RA, Israel E, Peters SP, Busse WW, Erzurum SC, Bleecker ER. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med 2010; 181: 315-323
  • 182 Schubert MS. Allergic fungal sinusitis. Clin Rev Allergy Immunol 2006; 30: 205-216
  • 183 Schubert MS. Allergic fungal sinusitis: pathophysiology, diagnosis and management. Med Mycol 2009; 47: S324-S330
  • 184 Dutre T, Al Dousary S, Zhang N, Bachert C. Allergic fungal rhinosinusitis (AFRS)- a bacterial presence coexisting with fungal disease?. J Allergy Clin Immunol 2013; 132: 487-489
  • 185 Scadding GK, Durham SR, Mirakian R, Jones NS, Drake-Lee AB, Ryan D, Dixon TA, Huber PA, Nasser SM. BSACI guidelines for the management of rhinosinusitis and nasal polyposis. Clin Exp Allergy 2008; 38: 260-275
  • 186 Accomazzo MR, Rovati GE, Vigano T, Hernandez A, Bonazzi A, Bolla M, Fumagalli F, Viappiani S, Galbiati E, Ravasi S, Albertoni C, Di Luca M, Caputi A, Zannini P, Chiesa G, Villa AM, Doglia SM, Folco G, Nicosia S. Leukotriene D4-induced activation of smooth-muscle cells from human bronchi is partly Ca2+-independent. Am J Respir Crit Care Med 2001; 163: 266-272
  • 187 Bandeira-Melo C, Weller PF. Eosinophils and cysteinyl leukotrienes. Prostaglandins Leukot Essent Fatty Acids 2003; 69: 135-143
  • 188 Roca-Ferrer J, Garcia-Garcia FJ, Pereda J, Perez-Gonzalez M, Pujols L, Alobid I, Mullol J, Picado C. Reduced expression of COXs and production of prostaglandin E(2) in patients with nasal polyps with or without aspirin-intolerant asthma. J Allergy Clin Immunol 2011; 128: 66-72
  • 189 Perez-Novo CA, Claeys C, Van Cauwenberge P, Bachert C. Expression of eicosanoid receptors subtypes and eosinophilic inflammation: implication on chronic rhinosinusitis. Respir Res 2006; 7: 75
  • 190 Liu CM, Hong CY, Shun CT, Hsiao TY, Wang CC, Wang JS, Hsiao M, Lin SK. Inducible cyclooxygenase and interleukin 6 gene expressions in nasal polyp fibroblasts: possible implication in the pathogenesis of nasal polyposis. Arch Otolaryngol Head Neck Surg 2002; 128: 945-951
  • 191 Okano M, Fujiwara T, Yamamoto M, Sugata Y, Matsumoto R, Fukushima K, Yoshino T, Shimizu K, Eguchi N, Kiniwa M, Urade Y, Nishizaki K. Role of prostaglandin D2 and E2 terminal synthases in chronic rhinosinusitis. Clin Exp Allergy 2006; 36: 1028-1038
  • 192 Yamamoto M, Okano M, Fujiwara T, Kariya S, Higaki T, Nagatsuka H, Tsujigiwa H, Yamada M, Yoshino T, Urade Y, Nishizaki K. Expression and characterization of PGD2 receptors in chronic rhinosinusitis: modulation of DP and CRTH2 by PGD2. Int Arch Allergy Immunol 2009; 148: 127-136
  • 193 Sanak M, Levy BD, Clish CB, Chiang N, Gronert K, Mastalerz L, Serhan CN, Szczeklik A. Aspirin-tolerant asthmatics generate more lipoxins than aspirin-intolerant asthmatics. Eur Respir J 2000; 16: 44-49
  • 194 Videler WJ, Badia L, Harvey RJ, Gane S, Georgalas C, van der Meulen FW, Menger DJ, Lehtonen MT, Toppila-Salmi SK, Vento SI, Hytönen M, Hellings PW, Kalogjera L, Lund VJ, Scadding G, Mullol J, Fokkens WJ. Lack of efficacy of long-term, low-dose azithromycin in chronic rhinosinusitis: a randomized controlled trial. Allergy 2011; 66: 1457-1468
  • 195 Cervin A, Wallwork B. Efficacy and safety of long-term antibiotics (macrolides) for the treatment of chronic rhinosinusitis. Curr Allergy Asthma Rep 2014; 14: 416
  • 196 Coles CL, Mabula K, Seidman JC, Levens J, Mkocha H, Munoz B, Mfinanga SG, West S. Mass distribution of azithromycin for trachoma control is associated with increased risk of azithromycin-resistant Streptococcus pneumoniae carriage in young children 6 months after treatment. Clin Infect Dis 2013; 56: 1519-1526
  • 197 Schäfer T, Schnoor M, Wagenmann M, Klimek L, Bachert C. Therapeutic Index (TIX) for intranasal corticosteroids in the treatment of allergic rhinitis. Rhinology 2011; 49: 272-280
  • 198 Perez Novo C, Kowalski M, Pattinska A, Kuna P, Holtappels G, Van Cauwenberge P, Gevaert P, SGO Johannson, Bachert C. Aspirin sensitivity and IgE antibodies to Staphylococcus aureus enterotoxins in nasal polyposis: Studies on the relationship. Int Arch Allergy Immunol. 2004; 133: 255-260
  • 199 Klimek L, Dollner R, Pfaar O, Mullol J. Aspirin desensitization: useful treatment for chronic rhinosinusitis with nasal polyps (CRSwNP) in aspirin-exacerbated respiratory disease (AERD)?. Curr Allergy Asthma Rep 2014; 14: 441
  • 200 Casale TB, Condemi J, LaForce C, Nayak A, Rowe M, Watrous M, McAlary M, Fowler-Taylor A, Racine A, Gupta N, Fick R, Della Cioppa G. Omalizumab Seasonal Allergic Rhinitis Trail Group . Effect of omalizumab on symptoms of seasonal allergic rhinitis: a randomized controlled trial. JAMA 2001; 286: 2956-2967
  • 201 Liu Y, Zhang S, Li DW, Jiang SJ. Efficacy of anti-interleukin-5 therapy with mepolizumab in patients with asthma: a meta-analysis of randomized placebo-controlled trials. PLoS One 2013; 8: e59872
  • 202 Castro M, Mathur S, Hargreave F, Boulet LP, Xie F, Young J, Wilkins HJ, Henkel T, Nair P. Reslizumab for poorly controlled, eosinophilic asthma: a randomized, placebo-controlled study. Am J Respir Crit Care Med 2011; 184: 1125-1132
  • 203 Laviolette M, Gossage DL, Gauvreau G, Leigh R, Olivenstein R, Katial R, Busse WW, Wenzel S, Wu Y, Datta V. and all . Effects of benralizumab on airway eosinophils in asthmatic patients with sputum eosinophilia. J Allergy Clin Immunol 2013; 132: 1086-1096 e1085
  • 204 “Clinical Trials Register.,” Online]. Available https://www.clinicaltrialsregister.eu
  • 205 Pelaia G, Vatrella A, Maselli R. The potential of biologics for the treatment of asthma. Nat Rev Drug Discov 2012; 11: 958-972
  • 206 Steinke JW. Anti-interleukin-4 therapy. Immunol Allergy Clin North Am 2004; 24: 599-614 vi
  • 207 Slager RE, Otulana BA, Hawkins GA, Yen YP, Peters SP, Wenzel SE, Meyers DA, Bleecker ER. IL-4 receptor polymorphisms predict reduction in asthma exacerbations during response to an anti-IL-4 receptor alpha antagonist. J Allergy Clin Immunol 2012; 130: 516-522 e514
  • 208 Wenzel S, Ford L, Pearlman D, Spector S, Sher L, Skobieranda F, Wang L, Kirkesseli S, Rocklin R, Bock B. and all . Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med 2013; 368: 2455-2466
  • 209 Oh CK, Leigh R, McLaurin KK, Kim K, Hultquist M, Molfino NA. A randomized, controlled trial to evaluate the effect of an anti-interleukin-9 monoclonal antibody in adults with uncontrolled asthma. Respir Res 2013; 14: 93
  • 210 Busse WW, Holgate S, Kerwin E, Chon Y, Feng J, Lin J, Lin SL. Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am J Respir Crit Care Med 2013; 188: 1294-1313
  • 211 Piper E, Brightling C, Niven R, Oh C, Faggioni R, Poon K, She D, Kell C, May RD, Geba GP. and all . A phase II placebo-controlled study of tralokinumab in moderate-to-severe asthma. Eur Respir J 2013; 41: 330-338
  • 212 Gauvreau GM, Boulet LP, Cockcroft DW, Fitzgerald JM, Carlsten C, Davis BE, Deschesnes F, Duong M, Durn BL, Howie KJ. and all . Effects of interleukin-13 blockade on allergen-induced airway responses in mild atopic asthma. Am J Respir Crit Care Med 2011; 183: 1007-1014
  • 213 Noonan M, Korenblat P, Mosesova S, Scheerens H, Arron JR, Zheng Y, Putnam WS, Parsey MV, Bohen SP, Matthews JG. Dose-ranging study of lebrikizumab in asthmatic patients not receiving inhaled steroids. J Allergy Clin Immunol 132: 567-574
  • 214 Scheerens H, Arron JR, Zheng Y, Putnam WS, Erickson RW, Choy DF, Harris JM, Lee J, Jarjour NN, Matthews JG. The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge. Clin Exp Allergy 2014; 44: 38-46
  • 215 Barnes PJ. New therapies for asthma: is there any progress?. Trends Pharmacol Sci 2010; 31: 335-343
  • 216 Taille C, Poulet C, Marchand-Adam S, Borie R, Dombret MC, Crestani B, Aubier M. Monoclonal Anti-TNF-alpha Antibodies for Severe Steroid-Dependent Asthma: A Case Series. Open Respir Med J 2013; 7: 21-25
  • 217 Gauvreau GM, O’Byrne PM, Boulet LP, Wang Y, Cockcroft D, Bigler J, FitzGerald JM, Boedigheimer M, Davis BE, Dias C. and all . Effects of an anti-TSLP antibody on allergen-induced asthmatic responses. N Engl J Med 2014; 370: 2102-2110
  • 218 Neighbour H, Boulet LP, Lemiere C, Sehmi R, Leigh R, Sousa AR, Martin J, Dallow N, Gilbert J, Allen A. and all . Safety and efficacy of an oral CCR3 antagonist in patients with asthma and eosinophilic bronchitis: a randomized, placebo-controlled clinical trial. Clin Exp Allergy 2014; 44: 508-516
  • 219 Busse WW, Wenzel SE, Meltzer EO, Kerwin EM, Liu MC, Zhang N, Chon Y, Budelsky AL, Lin J, Lin SL. Safety and efficacy of the prostaglandin D2 receptor antagonist AMG 853 in asthmatic patients. J Allergy Clin Immunol 2013; 131: 339-345
  • 220 Nair P, Gaga M, Zervas E, Alagha K, Hargreave FE, O’Byrne PM, Stryszak P, Gann L, Sadeh J, Chanez P. 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
  • 221 Beeh KM, Kanniess F, Wagner F, Schilder C, Naudts I, Hammann-Haenni A, Willers J, Stocker H, Mueller P, Bachmann MF. and all . The novel TLR-9 agonist QbG10 shows clinical efficacy in persistent allergic asthma. J Allergy Clin Immunol 2013; 131: 866-874
  • 222 “ClinicalTrials.gov.,” [Online]. Available https://clinicaltrials.gov/
  • 223 Franciosi LG, Diamant Z, Banner KH, Zuiker R, Morelli N, Kamerling IM, de Kam ML, Burggraaf J, Cohen AF, Cazzola M. and all . Efficacy and safety of RPL554, a dual PDE3 and PDE4 inhibitor, in healthy volunteers and in patients with asthma or chronic obstructive pulmonary disease: findings from four clinical trials. Lancet Respir Med 2013; 1: 714-727
  • 224 Gevaert P, Calus L, Van Zele T, Blomme K, De Ruyck N, Bauters W, Hellings P, Brusselle G, De Bacquer D, Van Cauwenberge P, Bachert C. Omalizumab is effective in allergic and nonallergic patients with nasal polyps and asthma. J Allergy Clin Immunol 2013 131: 110-116
  • 225 Gevaert P, Lang-Loidolt D, Stammberger H, Van Zele T, Holtappels G, Tavernier J, van Cauwenberge P, Bachert C. Nasal Interleukin-5 levels determine the response to anti-interleukin-5 treatment in nasal polyp patients. J Allergy Clin Immunol 2006; 118: 1133-1141
  • 226 Donovan R, Johansson SG, Bennich H, Soothill JF. Immunoglobulins in nasal polyp fluid. Int Arch Allergy Appl Immunol 1970; 37: 154-166
  • 227 Verbruggen K, Van CP, Bachert C. Anti-Ige for the treatment of allergic rhinitis–and eventually nasal polyps?. Int Arch Allergy Immunol 2009; 148: 87-98
  • 228 Pinto JM, Mehta N, DiTineo M, Wang J, Baroody FM, Naclerio RM. A randomized, double-blind, placebo-controlled trial of anti-IgE for chronic rhinosinusitis. Rhinology 2010; 48: 318-324
  • 229 Holgate S, Casale T, Wenzel S, Bousquet J, Deniz Y, Reisner C. The anti-inflammatory effects of omalizumab confirm the central role of IgE in allergic inflammation. J Allergy Clin Immunol 2005; 115: 459-465
  • 230 Garcia G, Magnan A, Chiron R, Contin-Bordes C, Berger P, Taillé C, Devouassoux G, de Blay F, Couderc LJ, Didier A, O’Callaghan DS, Girodet PO, Bourdeix I, Le Gros V, Humbert M. A proof-of-concept, randomized, controlled trial of omalizumab in patients with severe, difficult-to-control, nonatopic asthma. Chest 2013; 144: 411-419
  • 231 Vennera Mdel C, Picado C, Mullol J, Alobid I, Bernal-Sprekelsen M. Efficacy of omalizumab in the treatment of nasal polyps. Thorax 2011; 66: 824-825
  • 232 Gevaert P, Bachert C, Holtappels G, Perez Novo C, Van der Heyden J, Fransen L, Depraetere S, Walter H, van Cauwenberge P, Tavernier J. Enhanced Soluble Interleukin 5 Receptor alpha expression in nasal polyposis. Allergy 2003; 58: 371-379
  • 233 Long A, Rahmaoui A, Rothman KJ, Guinan E, Eisner M, Bradley MS, Iribarren C, Chen H, Carrigan G, Rosén K, Szefler SJ. Incidence of malignancy in patients with moderate-to-severe asthma treated with or without omalizumab. J Allergy Clin Immunol 2014; 134: 560-567
  • 234 Sel S, Wegmann M, Dicke T, Sel S, Henke W, Yildirim AO, Renz H, Garn H. Effective prevention and therapy of experimental allergic asthma using a GATA-3-specific DNAzyme. J Allergy Clin Immunol 2008; 121: 910-916
  • 235 Bermúdez-Humarán LG, Aubry C, Motta JP, Deraison C, Steidler L, Vergnolle N, Chatel JM, Langella P. Engineering lactococci and lactobacilli for human health. Curr Opin Microbiol 2013; 16: 278-283
  • 236 Krug N, Hohlfeld JM, Kirsten AM, Kornmann O, Beeh KM, Kappeler D, Korn S, Ignatenko S, Timmer W, Rogon C, Zeitvogel J, Nan Z, Bille J, Homburg U, Turowska A, Bachert C, Werfel T, Buhl R, Renz J, Garn H, Renz H. Allergen-Induced Asthmatic Responses Modified by a GATA-3 Specific DNAzyme. New England Journal of Medicine 2015; [in Press]