An update on emerging drugs for asthma. Garry M. Walsh PhD School of Medicine, University of Aberdeen, Scotland, U.K. Corresponding Author: Dr Garry M. Walsh Section of Immunology & Infection Division of Applied Medicine School of Medicine & Dentistry Institute of Medical Sciences University of Aberdeen Foresterhill, Aberdeen AB25 2ZD, U.K. Tel +44 (0)1224-437354 Fax +44 (0)1224-437348 e mail g.m.walsh@abdn.ac.uk 1 Abstract Introduction Biopharmaceutical approaches have identified new biologic-based therapies that target key cells and mediators that drive inflammatory responses in the asthmatic lung. This review based on English-language original articles in Pub Med or MedLine published in the last 5 years will update the current status, therapeutic potential and potential problems of recent drug developments in asthma therapy. It is recognised that airway inflammation is key to asthma pathogenesis. Biopharmaceutical approaches have identified new therapies that target key cells and mediators that drive the inflammatory responses in the asthmatic lung. Such an approach resulted in the development of biologics including IL-4, IL-5 and IL-13. However, clinical trials with these biologics in patients with asthma were for the most part disappointing even though they proved to be highly effective in animal models of asthma. Expert opinion It is becoming apparent that significant clinical effects with anti-cytokine-based therapies are more likely in carefully selected patient populations that take asthma phenotypes into account. It might also be more clinically effective if more than one cytokine and/or chemokine were to be targeted rather than a single mediator. . 2 1.0 Background Asthma is a major cause of ill-health that has markedly increased in prevalence in the UK and the rest of the developed world in recent decades [1]. In addition to reversible airway obstruction and airway hyperresponsiveness (AHR), asthma pathology results in fundamental structural changes to the airway including goblet cell hyperplasia, airway smooth muscle hypertrophy and subepithelial fibrosis [2]. Inhaled glucocorticoids (GCs) are first-line therapy for asthma because of their potent anti-inflammatory properties that primarily result in reduced numbers of airway inflammatory cells and their associated mediators [3]. Clinically, GCs reduce AHR, disease exacerbations and hospitalizations while improving lung function and quality of life and in their inhaled form are considered the most effective medications for asthma. However, they are symptomatic medications usually requiring lifetime therapy for the patient and asthma symptoms usually return on GC withdrawal. Moreover, variations in the clinical response of asthmatics to inhaled GC therapy are common while a significant subgroup of asthmatic patients responds poorly or not at all to high-dose inhaled or systemic GC treatment [4] These considerations, together with concerns over the adverse effects of GCs, which include adrenal suppression, reduced growth and reduced bone mineral density; indicate a clear need for more effective asthma therapy [5]. We have considerable knowledge regarding the cells, mediators and other factors controlling the pathogenic changes in asthma [6]. This information has informed the identification of molecules that target aspects of the complex inflammatory cascade in asthma. This article will update the current status of these compounds. 3 2. Anti-cytokine therapy Interleukin-5 Eosinophils play an important protective role in the immune response to parasitic infections and, detrimentally, in inflammatory conditions affecting the upper and lower airways, skin and gastrointestinal tract. Eosinophils are not normally present in healthy lungs but their accumulation is a well defined feature of the inflammatory processes in the lungs of patients with allergic asthma where release of their arsenal of proinflammatory mediators makes a major contribution to asthma pathogenesis. It has long been known that interleukin (IL)-5 is crucial to the development and release of eosinophils from the bone marrow, their enhanced adhesion to endothelial cells lining the post-capillary venules and their persistence, activation and secretion in the tissues. IL-5 was therefore selected as a potential target to prevent or blunt eosinophil-mediated inflammation in patients with asthma, leading to the development of humanized anti-IL-5 mAb, such as mepolizumab, reslizumab and benralizumab [7]. Early clinical trials with mepolizumab in patients with mild to severe asthma reported significant reductions in blood and sputum eosinophil numbers but clinical outcomes were disappointing, most likely because subjects were recruited on the basis of clinical and physiological characteristics rather than the presence of eosinophilic airway inflammation [8]. Asthma management strategies aimed at the control of eosinophilic airway inflammation in addition to clinical manifestations of the condition are associated with a reduction in the frequency of exacerbations [9]. Importantly therefore, two studies demonstrated that 4 mepolizumab treatment of asthmatics not only reduced eosinophil numbers in the blood and sputum but also resulted in a significant reduction in asthma exacerbations. Both studies were randomized, double-blind, placebo-controlled, parallel-group studies that investigated the effects of monthly infusions of 750 mg mepolizumab or placebo. The first study [10] used patients with a sputum eosinophilia whose asthma symptoms were refractory to high-dose corticosteroids and who had greater than two exacerbations requiring oral corticosteroids during the previous 12 months. The primary end point was the number of severe exacerbations per patient; secondary outcomes included asthma symptoms, health-related quality of life (HRQoL) measured with the Asthma Quality of Life Questionnaire (AQLQ), lung function, AHR and blood and sputum, eosinophil count. Compared with placebo (32 patients), mepolizumab treatment (29 patients) resulted in a significant reduction in blood sputum eosinophil numbers, with fewer severe exacerbations per patient as compared to placebo together with a significant improvement in quality of life. No significant improvements were reported for the other outcome measures and mepolizumab appeared to be well tolerated. The second study [11] used fewer patients (9 active, 11 placebo). These subjects exhibited persistent sputum eosinophilia despite requiring oral prednisone for control of asthma symptoms together with a history of exacerbations. In the mepolizumab treated group, one patient had an exacerbation compared with twelve exacerbations in the placebo group. Of those subjects in the placebo group, 9/10 of those analyzed exhibited sputum eosinophilia at the time of exacerbation while in the treatment group mepolizumab significantly reduced sputum and blood eosinophil counts to normal values. Mepolizumab treatment gave a significant and sustained reduction in the oral prednisone dose, a modest improvement in lung function, 5 significant improvement in scores on the Juniper Asthma Control Questionnaire and quality of life indices with no significant side effects reported. These studies in highly selected asthma patient populations demonstrate that mepolizumab attenuates aspects of eosinophil-induced airway inflammation refractive to GC therapy and may therefore potentially interfere with eosinophil-induced airway remodelling in addition to reducing asthma exacerbations. Mepolizumab may also prove useful in other settings such as GC resistant asthma or asthma in smokers with a sputum eosinophilia. However, as mepolizumab had no effect on AHR it is unlikely to be an effective mono therapy for asthma and it is only likely to be effective in tightly defined patient populations who exhibit eosinophil-induced airway inflammation. In addition, a recent open-label trial of seven patients [12] and a case report of one patient [13] suggested that mepolizumab may also be effective in the treatment of the eosinophilic disease Churg-Strauss syndrome, in which IL-5 plays a pivotal role. Ception Therapeutics Inc (presumed to be under license from Schering-Plough Corp and UCB Celltech) are developing the humanized anti-IL-5 mAb reslizumab. One recent randomized, placebo-controlled trial evaluated intravenous infusions of reslizumab (3.0 mg/kg) or placebo at weeks 0, 4, 8 and 12 in 106 patients with poorly controlled asthma who were taking high-dose inhaled GCs and exhibited a sputum eosinophilia greater than 3%. When all patients were included for analysis, there was a non-significant trend toward improvement in asthma control associated with a significant improvement in lung function as assessed by the Asthma Control Questionnaire score together with a significant decrease in sputum eosinophilia. In those patients with concomitant nasal 6 polyposis, reslizumab treatment was associated with a significant improvement in asthma symptom control at baseline compared with week 15. There was a non-significant reduction in asthma exacerbations in the reslizumab group while the adverse-event profile for reslizumab and placebo were similar [14]. Other data from limited pilot trials suggested that reslizumab was a potentially efficacious and well-tolerated treatment for eosinophilic esophagitis, hypereosinophilic syndrome and eosinophilic nasal polyposis [15]. Benralizumab is a novel humanised afucosylated IgG1 mAb indicated for the potential treatment of asthma and COPD that binds to a distinct epitope within the extracellular domain of recombinant human IL-5R. At the time of publication, benralizumab was undergoing phase II clinical trials in both the specified indications (ClinicalTrials.gov identifiers: NCT01238861 and NCT01227278). Afucosylation is associated with enhanced antibody-dependent cell cytotoxicity and benralizumab was found to induce apoptosis in eosinophils and basophils [16]. Other anti-IL-5 mAb act by neutralising the effects of IL-5 whereas benralizumab targets the effector cells, mainly eosinophils and basophils, many of whose functions are driven by IL-5. Tissue eosinophils resident in bronchial biopsies of patients with mild atopic asthma exhibited intense immune positivity for benralizumab in contrast to resident mast cells, which were negative [16]. These findings indicate that benralizumab binds human lung tissue-resident eosinophils expressing IL-5Ralpha and could delete these cells, thereby acting as a potential asthma therapeutic. Indeed a phase-1 study in subjects with mild asthma demonstrated that intravenous benralizumab (0.3-3.0 mg/kg) rapidly induced near total depletion of 7 peripheral blood eosinophils while exhibiting an adequate safety profile and doseproportional pharmacokinetics [17]. IL-4 and IL-13 Eosinophil involvement as a major pro-inflammatory effector cell in asthma is wellestablished [18] with T-helper-2 (TH-2) cells that express IL-4, IL-5 and IL-13 representing essential and central co-ordinators of asthmatic inflammation [19]. Both IL4 and IL-13 are important in eosinophil accumulation and are key factors in IgE synthesis by B cells [20]. There is also evidence that in asthma IL-13 may act directly on airway smooth muscle cell contractility [21,22, 23]; this together with direct effects by IL-13 on epithelial cells and mucus production [24] may contribute to AHR and airway narrowing. The genes encoding IL-4 and IL-13 are located on the cytokine cluster on chromosome 5q31; both cytokines share some structural similarities. Each exerts their actions through the IL-4R/IL-13R1 receptor complex which then activates the transcription factor STAT-6 [25]. They therefore have multiple overlapping functions although IL-4 has independent effects via the IL-4R receptor. Receptors for IL-13 are expressed by a multitude of cell types important in the pathogenesis of asthma including eosinophils, mast cells together with structural airway epithelial cells, fibroblasts and smooth muscle cells. In asthma, the most important cellular sources of IL-13 are T cells, mast cells and eosinophils. Increased levels of IL-13 have been reported in the sputum and bronchial biopsies from patients with asthma which correlated with increased eosinophil numbers [26, 27]. Importantly, mast cells expressing IL-13 are present within the airway smooth muscle in asthmatic subjects suggesting a 8 pivotal role for IL-13 in interactions between these cell types [28]. IL-13 may also have direct effects on airway smooth muscle function by enhancing the contraction effect of acetylcholine [29]. It is now widely appreciated that the asthmatic airway epithelium is intrinsically abnormal and critically contributes to the airway inflammatory response [30]. An important aspect of IL-13 therefore is the effect it has on the functions of airway epithelial cells including induction of a hyper-secretory state [31] and reduced barrier function through down-regulation of proteins associated with maintaining epithelial tight junctions [32]. IL-13 is a potent promoter of epithelial cell TGF production; that in turn activates myofibroblasts and is also involved in ECM deposition leading to sub-epithelial membrane thickening; both thought to contribute to airway remodelling [33, 34]. IL-13 therefore appears to represent a potentially effective target for asthma therapy [35]. The anti-IL-13 mAb lebrikizumab (Genentech/Chugai Pharmaceutical) has recently been demonstrated to significantly improve lung function in patients with inadequately controlled asthma, but only in a subgroup defined on the basis of high serum levels of periostin [36]. The latter is a cellular matrix protein that is released by airway epithelial cells stimulated with IL-13. Periostin exhibits effects on epithelial cells and fibroblasts that may contribute to airway remodelling in asthma [37, 38]. A recombinant human IL-4 variant, pitrakinra (Aerovant), was developed that competitively inhibits the IL-4Rreceptor complex to interfere with the actions of both IL-4 and IL-13 and initial clinical trials indicated that such an approach may prove beneficial in patients with atopic asthma [reviewed in 39]. A more recent double-blind, 9 randomized, placebo-controlled trial of inhaled pitrakinra in 534 patients with uncontrolled, moderate-to-severe asthma reported significant effects on exacerbations rates and symptom scores in patients with an elevated blood eosinophilia [40]. However, another recent 12-week clinical trial with an mAb directed at the IL-4rα (AMG-317, Amgen) reported significant reductions in blood IgE levels and eosinophils but found no significant change in measured asthma outcomes [41]. Dual inhibition of IL-4 and IL-13 can affect the course of the late asthmatic response after experimental allergen challenge but further large scale clinical trials on patients with day to day asthma are required to fully validate such an approach. Tumour necrosis factor-α Tumour necrosis factor (TNF)-α is an important cytokine in innate immune responses that has been implicated in several chronic inflammatory diseases including rheumatoid arthritis and Crohn’s disease with anti-TNFα therapy proving useful in these conditions. It is produced principally by macrophages while other pro-inflammatory cells including monocytes, dendritic cells, B lymphocytes, T cells, neutrophils, mast cells and eosinophils, together with the structural cells fibroblasts, epithelial cells and smooth muscle cells represent significant sources. TNF- has pro-inflammatory effects on eosinophils, neutrophils, T cells, epithelial cells and endothelial cells and may play a key role in amplifying airway inflammation through activation of transcription factors such as NF-B and AP-1. TNF- is expressed in biopsies and lavage fluid from asthmatic airways, particularly in patients with severe asthma compared with those with wellcontrolled disease. TNF- is thought to contribute to AHR, airway remodelling and GC 10 resistance in asthma and therefore represents a potential target for therapy [7]. Humanized murine anti-TNF mAb (infliximab) and soluble TNF receptor linked to human IgG1 (etanercept) have been developed and preliminary clinical studies in asthma did show significant improvements in lung function, airway hypereactivity and exacerbation rate, particularly in patients with severe asthma refractory to GC treatment [42]. However, a more recent clinical trial with the anti-TNF biologic golimumab in patients with severe uncontrolled persistent asthma reported negative clinical findings. Importantly, this study was terminated early due to unacceptable adverse events including frequent serious infections, eight malignancies and one death in the active-treatment group compared with the placebo group [43]. Overall it appears that TNF- inhibitors are effective in a sub-group of patients with asthma and that identification of the correct patient population may improve clinical outcomes [44]. However, the unfavorable risk/benefit ratio exhibited by golimumab does cast doubt on the future of anti-TNF therapy in severe asthma. 3.0 Conclusion The development of novel asthma anti-inflammatory therapy based on targeting cytokines has proven to be for the most part disappointing; in particular results from animal-based studies have been very misleading. The majority of biologics have proven inadequate in the clinical setting in asthma even though they were highly effective in animal models of asthma. This issue has been emphasized recently by Holgate who points out that at the time of publication of his review articles, out of around 3,000 peer-reviewed publications implicating IL-13 as a central mediator in asthma, only four relate to direct evidence in human asthma [45]. Two of 11 the latter studies demonstrated that elevated levels of IL-13 were present in the sputum of around half of asthmatic patients tested with no significant association with disease severity [26, 27]. It is not surprising therefore that anti-IL-13 therapy proved to be disappointing in asthma therapy given that only half of the patients recruited to a given clinical trial have any likelihood of responding to this biologic. Indeed the development of discriminatory biomarkers and genetic profiling may identify patients with particular sub-phenotypes of asthma. This point is illustrated by the recent studies that suggest that both mepolizumab and lebrikizumab therapy have beneficial effects in carefully selected patient populations. Expert Opinion Inhaled glucocorticoids remain the gold standard treatment for asthma because they suppress multiple inflammatory mechanisms in parallel. It might be more clinically effective, therefore, if novel biologics were to be targeted at more than one cytokine and/or chemokine, an approach whose potential is illustrated by pitrakinra. Another major issue is that of the potential or actual side effects of novel compounds that are designed to interfere with important aspects of both the innate and acquired immune systems as exemplified by the unacceptable adverse effects associated with the golimumab treatment in patients with severe uncontrolled asthma. What is becoming more widely accepted is that future clinical trials of novel biologic therapy for asthma should include very careful patient selection to ensure a significant clinical outcome. One other important consideration is that the placebo group in trials of biologics in asthma often exhibit a marked improvement in symptoms which may reduce the likelihood of significant findings being observed for the test compound. This phenomenon is most 12 likely due to the close monitoring of trial subjects that ensures greater compliance in adhering to their normal anti-inflammatory therapy. The ultimate goal would be to develop asthma treatments that are truly disease modifying rather than the symptomatic treatments currently available and we do seem to be starting to make some progress towards this goal. 13 4.0 References 1 Anandan C, Nurmatov U, van Schayck OC, Sheikh A. Is the prevalence of asthma declining? Systematic review of epidemiological studies.Allergy. 2010;65(2):152-67. 2 Barnes PJ. Pathophysiology of allergic inflammation Immunol Rev. 2011;242(1):31-50. 3 Barnes PJ, Adcock IM, Ito K (2005) Histone acetylation and deacetylation: importance in inflammatory lung diseases. Eur Respir J, 25: 552-563 4 Berger WE. New approaches to managing asthma: A US perspective. Therap Clin Risk Management 2008;4:363-379 5 Holtzman, MJ (2003) Drug development for asthma. Am J Resp Cell Mol Biol 29: 163-171 6 Walsh GM. Targeting airway inflammation: Novel therapies for the treatment of asthma. Curr Med Chem 2006;13:3105-3111. 7 Walsh GM. Novel cytokine-directed therapies for asthma. Discov Med. 2011;11(59):283-91. 14 8 O’Byrne, P.M. The demise of anti IL-5 for asthma, or not. Am. J. Respir. Crit Care Med. 2007, 176, 1059-60. 9 Walsh GM. Mepolizumab and eosinophil-mediated disease. Curr Med Chem. 2009;16:4774-8. 10 Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, Sousa A, Marshall RP, Bradding P, Green RH, Wardlaw AJ, Pavord ID. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med 360:973-984, 2009. ••This trial used patients with sputum eosinophilia whose asthma symptoms were refractory to high-dose GCs and who had greater than two exacerbations requiring oral GCs during the previous 12 months. The primary endpoint was the number of severe exacerbations per patient; secondary outcomes included asthma symptoms, HRQoL measured with the Asthma Quality of Life Questionnaire, lung function, AHR and blood or sputum eosinophil count. Compared with placebo (n = 32), mepolizumab treatment (n = 29) resulted in a significant reduction in blood and sputum eosinophil numbers, with fewer severe exacerbations per patient compared with placebo, together with a significant improvement in HRQoL. 11 Nair P, Pizzichini MM, Kjarsgaard M, Inman MD, Efthimiadis A, Pizzichini E, Hargreave FE, O'Byrne PM. Mepolizumab for prednisone-dependent asthma with sputum eosinophilia. N Engl J Med 360:985-993, 2009. 15 • Patients (n = 9 active, 11 placebo) with a history of exacerbations exhibited persistent sputum eosinophilia despite requiring oral prednisone for control of asthma symptoms. In the mepolizumab-treated group, 1 patient had an exacerbation compared with 12 exacerbations in the placebo group. Mepolizumab treatment significantly reduced sputum and blood eosinophil counts to normal values, yielded a significant reduction in the oral prednisone dose, a modest improvement in FEV1 (mean = 300 ml), significant improvement in scores on the Juniper Asthma Control Questionnaire (p = 0.01) and significant improvement in health-related quality of life (HRQoL) with no significant side effects reported. 12 Kim S, Marigowda G, Oren E, Isreal E and Wechsler M. Mepolizumab as a steroidsparing treatment option in patients with Churg-Strauss syndrome, J Allergy Clin Immunol 2010;125: 1336–1343. 13 Kahn JE, Grandpeix-Guyodo C, Marroun I, Catherinot E, Mellot E and Roufosse F et al. Sustained response to mepolizumab in refractory Churg-Strauss syndrome, J Allergy Clin Immunol 2010;125 : 267–270. 14 Castro M, Mathur S, Hargreave F, Boulet LP, Xie F, Young J, Wilkins HJ, Henkel T, Nair P; for the Res-5-0010 Study Group. Reslizumab for Poorly Controlled, Eosinophilic Asthma: A Randomized, Placebo-Controlled Study. Am J Respir Crit Care Med. 2011 Aug 18. [Epub ahead of print] 16 15 Walsh GM. Reslizumab, a humanized monoclonal anti-IL-5 antibody for the treatment of eosinophil-mediated inflammatory conditions. Curr Opin Mol Ther 2009; 11:329-336. 16 Kolbeck R, Kozhich A, Koike M et al. MEDI-563, a humanized anti-IL-5 receptor alpha mAb with enhanced antibody-dependent cell-mediated cytotoxicity function. J Allergy Clin Immunol 2010; 125:1344–53.e2 17 Busse WW, Katial R, Gossage D, Sari S, Wang B, Kolbeck R, Coyle AJ, Koike M, Spitalny GL, Kiener PA, Geba GP, Molfino NA. Safety profile, pharmacokinetics, and biologic activity of MEDI-563, an anti–IL-5 receptor antibody, in a phase I study of subjects with mild asthma. J Allergy Clinl Immunol 2010; 125: 1237-1244. •• An open-label clinical trial demonstrating that single, escalating doses of benralizumab had a reasonable safety profile and resulted in marked reduction of peripheral blood eosinophil counts in patients with mild allergic asthma within 24 h of administration. 18 Blanchard, C.; Rothenberg ME. Biology of the eosinophil. Adv. Immunol. 2009; 101: 81-121. 19 Barrett NA, Austen KF. Innate cells and T helper 2 cell immunity in airway inflammation. Immunity. 2009;31(3):425-37. 17 20 Wills-Karp M,; Luyimbazi J, Xu X et al Interleukin-13: central mediator of allergic asthma. Science 1998; 282: 2258-2260 21 Tliba O, Deshpande D, Chen H et al. IL-13 enhances agonist evoked calcium signals and contractile responses in airway smooth muscle. Br J Pharmacol 2003; 140:1159–62. 22 Deshpande DA, Dogan S, Walseth TF, Miller SM, Amrani Y, Panettieri RA, Kannan MS. Modulation of calcium signaling by interleukin-13 in human airway smooth muscle: role of CD38/cyclic adenosine diphosphate ribose pathway. Am J Respir Cell Mol Biol. 2004;31:36-42. 23 Chiba Y, Nakazawa S, Todoroki M, Shinozaki K, Sakai H, Misawa M. Interleukin13 Augments Bronchial Smooth Muscle Contractility with an Upregulation of RhoA Protein. Am J Respir Cell Mol Biol. 2009;40(2):159-67. 24 Kuperman DA, Huang X, Koth LL et al. Direct effects of interleukin-13 on epithelial cells cause airway hyperreactivity and mucus overproduction in asthma. Nat Med 2002; 8:885–9. 25 Jiang H, Harris MB, Rothman P. IL-4/IL-13 signalling beyond JAK/STAT. J Allergy Clin Immunol 2000; 105: 1063-1070 18 26 Berry MA, Parker D, Neale N, Woodman L, Morgan A, Monk P, Bradding P, Wardlaw AJ, Pavord ID, Brightling CE: Sputum and bronchial submucosal IL-13 expression in asthma and eosinophilic bronchitis. J Allergy Clin Immunol 2004, 114:1106-1109. 27 Saha SK, Berry MA, Parker D, Siddiqui S, Morgan A, May R, Monk P, Bradding P, Wardlaw AJ, Pavord ID, Brightling CE: Increased sputum and bronchial biopsy IL-13 expression in severe asthma. J Allergy Clin Immunol 2008, 121:685-691 28 Brightling CE, Symon FA, Holgate ST, Wardlaw AJ, Pavord ID, Bradding P: Interleukin-4 and -13 expression is co-localized to mast cells within the airway smooth muscle in asthma. Clin Exp Allergy 2003, 33:1711-1716. 29 Grunstein MM, Hakonarson H, Leiter J et al. IL-13-dependent autocrine signaling mediates altered responsiveness of IgE sensitized airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2002; 282:L520–8. 30 Holgate ST. Has the time come to rethink the pathogenesis of asthma? Curr Opin Allergy Clin Immunol. 2010;10(1):48-53 31 Danahay H, Atherton H, Jones G, Bridges RJ, Poll CT. Interleukin-13 induces a hypersecretory ion transport phenotype in human bronchial epithelial cells. Am J Physiol Lung Cell MolPhysiol 2002; 282:L226–36. 19 32 Ahdieh M, Vandenbos T, Youakim A. Lung epithelial barrier function and wound healing are decreased by IL-4 and IL-13 and enhanced by IFN-gamma. Am J Physiol Cell Physiol 2001;281:C2029–38. 33 Wenzel SE, Schwartz LB, Langmac EL, Halliday JL, Trudeau JB, Gibbs RL, Chu HW Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am J Respir Crit Care Med 1999; 160: 1001-8. 34 Hashimoto S, Gon Y, Takeshita I, Maruoka S, Horie T. IL-4 and IL-13 induce myofibroblastic phenotype of human lung fibroblasts through c-Jun NH2-terminal kinase-dependent pathway. J Allergy Clin Immunol 2001; 107:1001–8. 35 Blease K. Therapeutics targeting IL-13 for the treatment of pulmonary inflammation and airway remodeling. Curr OpinInvestig Drugs 2008; 9: 1180–4. 36 Corren J, Lemanske RF, Hanania NA, Korenblat PE, Parsey MV, Arron JR, Harris JM, Scheerens H, Wu LC, Su Z, Mosesova S, Eisner MD, Bohen SP, Matthews JG. Lebrikizumab Treatment in Adults with Asthma. N Engl J Med. 2011 Aug 3. [Epub ahead of print] • This clinical trial with the anti-IL-13 biologic lebrikizumab in patients with uncontrolled asthma despite inhaled GC therapy reported that patients with high 20 pretreatment levels of serum periostin, a cellular matrix protein that is released by airway epithelial cells stimulated with IL-13, had greater and significant improvement in lung function following lebrikizumab treatment than did patients with low periostin levels. 37 Sidhu SS, Yuan S, Innes AL, et al. Roles of epithelial cell-derived periostin in TGF-β activation, collagen production and collagen gel elasticity in asthma. Proc Natl Acad Sci USA 2010;107:14170-5. 38 Takayama G, Arima K, Kanaji T, et al. Periostin: A novel component of subepithelial fibrosis of bronchial asthma downstream of IL-4 and IL-13 signals. J Allergy Clin Immunol 2006;118:98-104. 39 Walsh GM. Emerging drugs for asthma. Expert Opin Emerg Drugs. 2008;13:643-53. 40 Wenzel SE, Ind PW, Otulana BA, Bleecker ER, Kuna P, Yen YP. Inhaled pitrakinra, an IL-4/IL-13 antagonist, reduced exacerbations in patients with eosinophilic asthma. Ann Cong Eur Resp Soc (September 18-22, 2010). Abstr #P3980. 41 Corren J, Busse W, Meltzer EO, Mansfield L, Bensch G, Fahrenholz J, Wenzel SE, Chon Y, Dunn M, Weng HH, Lin SL. A randomized, controlled, phase 2 study of AMG 317, an IL-4Ralpha antagonist, in patients with asthma. Am J Respir Crit Care Med 181(8):788-796, 2010. 21 42 Brightling C, Berry M, Amrani Y. Targeting TNF-alpha: a novel therapeutic approach for asthma. J Allergy Clin Immunol 121:5-10, 2008. 43 Wenzel SE, Barnes PJ, Bleecker ER, Bousquet J, Busse W, Dahlén SE, Holgate ST, Meyers DA, Rabe KF, Antczak A, Baker J, Horvath I, Mark Z, Bernstein D, Kerwin E, Schlenker-Herceg R, Lo KH, Watt R, Barnathan ES, Chanez P; A randomized, doubleblind, placebo-controlled study of tumor necrosis factor-alpha blockade in severe persistent asthma. Am J Respir Crit Care Med 179(7):549-558, 2009. • This clinical trial with the anti-TNFalpha biologic golimumab in patients with severe uncontrolled persistent asthma reported negative clinical findings. Importantly, this trial was terminated early due to unacceptable adverse events, including frequent serious infections and eight malignancies in the active-treatment group compared with the placebo group. 44 Matera MG, Calzetta L, Cazzola M TNF-alpha inhibitors in asthma and COPD: we must not throw the baby out with the bath water. Pulm Pharmacol Ther. 2010;23(2):1218. 45 Holgate ST. A look at the pathogenesis of asthma: the need for a change in direction. Discov Med 9(48):439-447, 2010 22