Kinases as Novel Therapeutic Targets in Asthma...

28
1521-0081/68/3/788815$25.00 http://dx.doi.org/10.1124/pr.116.012518 PHARMACOLOGICAL REVIEWS Pharmacol Rev 68:788815, July 2016 Copyright © 2016 by The American Society for Pharmacology and Experimental Therapeutics ASSOCIATE EDITOR: CLIVE PAGE Kinases as Novel Therapeutic Targets in Asthma and Chronic Obstructive Pulmonary Disease Peter J. Barnes National Heart and Lung Institute, Imperial College, London, United Kingdom Abstract ................................................................................... 789 I. Introduction ............................................................................... 789 II. Role of Kinases in Asthma and COPD ...................................................... 790 III. Kinases.................................................................................... 790 A. Kinase Inhibitors ...................................................................... 791 IV. Nuclear Factor-kB inhibition ............................................................... 791 A. Role in Asthma and Chronic Obstructive Pulmonary Disease............................ 791 B. Inhibitors.............................................................................. 793 V. Mitogen-Activated Protein Kinase Inhibition ............................................... 793 A. p38 Mitogen-Activated Protein Kinase .................................................. 794 1. Role in Asthma and Chronic Obstructive Pulmonary Disease. ....................... 794 B. p38 Inhibitors .......................................................................... 795 C. Jun NH2-terminal Kinase Inhibitors ................................................... 796 D. Extracellular Regulating Kinase/MAP Kinase Kinase Inhibitors ......................... 796 E. Apoptosis Signal-Regulating Kinase-1 Inhibition ........................................ 797 F. TGFb-Activated Kinase-1 Inhibition .................................................... 797 G. MAPKAPK2 Inhibition ................................................................. 797 VI. Phosphoinositide-3-kinase Inhibition ....................................................... 797 A. Role in Asthma and Chronic Obstructive Pulmonary Disease............................ 798 B. Inhibiting Phosphoinositide-3-kinase-Akt-Mammalian Target of Rapamycin Signaling . . . 799 1. Phosphoinositide-3-kinase Inhibitors. ............................................... 799 2. Akt-Mammalian Target of Rapamycin Inhibitors. ................................... 800 3. Activating Endogenous Inhibitors................................................... 800 VII. Janus-Activated Kinase Inhibition ......................................................... 801 A. Role in Asthma and Chronic Obstructive Pulmonary Disease. ....................... 801 B. Jakinibs............................................................................ 802 VIII. Spleen Tyrosine Kinase Inhibition.......................................................... 803 A. Role in Asthma and Chronic Obstructive Pulmonary Disease. ....................... 803 B. Spleen Tyrosine Kinase Inhibitors................................................... 804 IX. Receptor Tyrosine Kinases ................................................................. 804 A. Role in Asthma and Chronic Obstructive Pulmonary Disease............................ 804 B. Receptor Tyrosine Kinase Inhibitors .................................................... 805 C. c-Kit Inhibitors ........................................................................ 805 X. Transforming Growth Factor-b Receptor Kinase ............................................ 805 A. Role in Asthma and Chronic Obstructive Pulmonary Disease............................ 805 B. T-bRI Kinase Inhibitors ................................................................ 806 XI. Protein Kinase C Inhibition ................................................................ 806 A. Role in Asthma and Chronic Obstructive Pulmonary Disease............................ 806 B. Protein Kinase C Inhibitors ............................................................ 806 XII. Other Kinase Targets ...................................................................... 807 Address correspondence to: Prof. P. J. Barnes, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse St, London SW3 6LY, UK. E-mail: [email protected] dx.doi.org/10.1124/pr.116.012518. 788 by guest on June 30, 2020 Downloaded from

Transcript of Kinases as Novel Therapeutic Targets in Asthma...

Page 1: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

1521-0081/68/3/788–815$25.00 http://dx.doi.org/10.1124/pr.116.012518PHARMACOLOGICAL REVIEWS Pharmacol Rev 68:788–815, July 2016Copyright © 2016 by The American Society for Pharmacology and Experimental Therapeutics

ASSOCIATE EDITOR: CLIVE PAGE

Kinases as Novel Therapeutic Targets in Asthma andChronic Obstructive Pulmonary Disease

Peter J. Barnes

National Heart and Lung Institute, Imperial College, London, United Kingdom

Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 789I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 789II. Role of Kinases in Asthma and COPD. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 790III. Kinases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 790

A. Kinase Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 791IV. Nuclear Factor-kB inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 791

A. Role in Asthma and Chronic Obstructive Pulmonary Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . 791B. Inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 793

V. Mitogen-Activated Protein Kinase Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 793A. p38 Mitogen-Activated Protein Kinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 794

1. Role in Asthma and Chronic Obstructive Pulmonary Disease. . . . . . . . . . . . . . . . . . . . . . . . 794B. p38 Inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795C. Jun NH2-terminal Kinase Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 796D. Extracellular Regulating Kinase/MAP Kinase Kinase Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . 796E. Apoptosis Signal-Regulating Kinase-1 Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 797F. TGFb-Activated Kinase-1 Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 797G. MAPKAPK2 Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 797

VI. Phosphoinositide-3-kinase Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 797A. Role in Asthma and Chronic Obstructive Pulmonary Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . 798B. Inhibiting Phosphoinositide-3-kinase-Akt-Mammalian Target of Rapamycin Signaling . . . 799

1. Phosphoinositide-3-kinase Inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7992. Akt-Mammalian Target of Rapamycin Inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8003. Activating Endogenous Inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 800

VII. Janus-Activated Kinase Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 801A. Role in Asthma and Chronic Obstructive Pulmonary Disease. . . . . . . . . . . . . . . . . . . . . . . . 801B. Jakinibs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 802

VIII. Spleen Tyrosine Kinase Inhibition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 803A. Role in Asthma and Chronic Obstructive Pulmonary Disease. . . . . . . . . . . . . . . . . . . . . . . . 803B. Spleen Tyrosine Kinase Inhibitors.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 804

IX. Receptor Tyrosine Kinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 804A. Role in Asthma and Chronic Obstructive Pulmonary Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . 804B. Receptor Tyrosine Kinase Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805C. c-Kit Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805

X. Transforming Growth Factor-b Receptor Kinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805A. Role in Asthma and Chronic Obstructive Pulmonary Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . 805B. T-bRI Kinase Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 806

XI. Protein Kinase C Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 806A. Role in Asthma and Chronic Obstructive Pulmonary Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . 806B. Protein Kinase C Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 806

XII. Other Kinase Targets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 807

Address correspondence to: Prof. P. J. Barnes, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse St,London SW3 6LY, UK. E-mail: [email protected]

dx.doi.org/10.1124/pr.116.012518.

788

by guest on June 30, 2020D

ownloaded from

Page 2: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

A. Rho-Associated Protein Kinase Inhibition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 807B. Interleukin-1 Receptor-Associated Kinase Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 807C. Src Family Kinase Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 807D. Hematopoietic Cell Kinase Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 807E. Bruton’s Tyrosine Kinase. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 808

XIII. Future Developments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 808A. Improving Selectivity and Safety . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 808B. Targeting Several Kinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 809C. Overcoming Kinase Inhibitor Resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 809D. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 809References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 809

Abstract——Multiple kinases play a critical role inorchestrating the chronic inflammation and structuralchanges in the respiratory tract of patients with asthmaand chronic obstructive pulmonary disease (COPD). Ki-nases activate signaling pathways that lead to contractionof airway smooth muscle and release of inflammatorymediators (suchas cytokines, chemokines, growth factors)as well as cell migration, activation, and proliferation. Forthis reason there has been great interest in the develop-ment of kinase inhibitors as anti-inflammatory therapies,particular where corticosteroids are less effective, as insevere asthma andCOPD.However, it has proven difficultto develop selective kinase inhibitors that are botheffective and safe after oral administration and this hasled to a search for inhaled kinase inhibitors, which would

reduce systemic exposure. Although many kinases havebeen implicated in inflammation and remodeling ofairway disease, very few classes of drug have reachedthe stage of clinical studies in these diseases. The mostpromisingdrugsarep38MAPkinases, isoenzyme-selectivePI3-kinases, Janus-activated kinases, and Syk-kinases,and inhaled formulations of these drugs are now indevelopment. There has also been interest in developinginhibitors that block more than one kinase, because thesedrugs may be more effective and with less risk of losingefficacy with time. No kinase inhibitors are yet on themarket for the treatment of airway diseases, but as kinaseinhibitors are improved from other therapeutic areasthere is hope that these drugs may eventually proveuseful in treating refractory asthma and COPD.

I. Introduction

Asthma and chronic obstructive pulmonary disease(COPD) both cause obstruction of the airways and involvechronic inflammation of the respiratory tract. Both are veryprevalent diseases andare increasing throughout theworld.Asthma is now the most common chronic disease in

developed countries, affecting up to 20% of the popula-tion, and is increasing, particularly in developingcountries (Beasley et al., 2000). It still causes deathand leads to hospital admissions because of acute

exacerbations. Although asthma is usually well con-trolled with inhaled corticosteroids, many patients withasthma are poorly controlled in the real world asadherence to this treatment is poor and inhaler useoften inadequate (Partridge et al., 2006; Braido et al.,2013). Approximately 20% of asthma patients areparticularly difficult to manage, and 3–5% of patientshave severe disease that is not controlled even bymaximal inhaled treatment with an inhaled corticoste-roid and a long-acting b2-agonist (Chung and Wenzel,

ABBREVIATIONS: AHR, airway hyperresponsiveness; AMPK, adenosine monophosphate kinase; AP-1, activator protein-1; ASK, apoptosissignal-regulatory kinase; ASM, airway smooth muscle; ATP, adenosine triphosphate; AZD2014, 3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin-7-yl)-N-methylbenzamide; Bay 65-1942, (7E)-7-[2-(cyclopropylmethoxy)-6-oxocyclohexa-2,4-dien-1-ylidene]-5-[(3S)-piperidin-3-yl]-4,8-dihydro-1H-pyrido[2,3-d][1,3]oxazin-2-one; BTK, Bruton’s tyrosine kinase; COPD, chronic obstructive pulmonary disease; CT99021,6-[[2-[[4-(2,4-Dichlorophenyl)-5-(5-methyl-1H-imidazol-2-yl)-2-pyrimidinyl]amino]ethyl]amino]-3-pyridinecarbonitrile; CXCL8, interleukin-8;EGFR, epidermal growth factor receptor; ERK, extracellular regulating kinase; FEV1, forced expiratory volume in 1 second; FGF(R), fibroblastgrowth factor (receptor); GM-CSF, granulocyte-macrophage colony stimulating factor; GR, glucocorticoid receptor; GSK2269557, 2-(6-(1H-indol-4-yl)-1H-indazol-4-yl)-5-((4-isopropylpiperazin-1-yl)methyl)oxazole hydrochloride; GSK3b, glycogen synthase-3b; Hck, hematopoietic cell kinase; HDAC2,histone deacetylase-2; hsp, heat shock protein; IFN, interferon; IgE, immunoglobulin E; IKK, inhibitor of kB kinase; IL, interleukin; IRAK,interleukin-1 receptor-associated kinase; ITAM, immunoreceptor-tyrosine-based activation motif; JAK, Janus-activated kinase; JNK, Jun NH2-terminal kinase; LAS 189386, 1-{2-[(1S,4S)-2,5-diazabicyclo[2.2.1]hept-2-yl]pyridin-4-yl}-N-pyrazin-2-yl-1H-indazol-3-amine; LPS, lipopolysaccharide;LY294002, 2-Morpholin-4-yl-8-phenylchromen-4-one; MAPK, mitogen-activated protein kinase; MAPKAPK or MK2, MAP-activated protein kinase;MKP, MAP kinase phosphatase; mTOR, mammalian target of rapamycin; NF-kB, nuclear factor-kappa B; NIK, NF-kB-inducing kinase; PBMC,peripheral blood mononuclear cells; PD-98059, 29-Amino-39-methoxyflavone; PDGF(R), platelet-derived growth factor (receptor); PF-03715455, 1-[5-tertbutyl-2-(3-chloro-4-hydroxyphenyl)pyrazol-3-yl]-3-[[2-[[3-[2-(2-hydroxyethylsulfanyl)phenyl]-[1,2,4]triazolo[4,3-a]pyridin-6-yl]sulfanyl]phenyl]methyl]urea;PH-797804, (aS)-3-{3-bromo-4-[(2,4-difluorobenzyl)oxy]-6-methyl-2-oxopyridin-1(2H)-yl}-N,4-dimethylbenzamde; PI3K, phosphoinositide-3-kinase; PKC, protein kinase C; PTEN, phosphatase and tensin homolog deleted from chromosome 10; ROCK, Rho-associated protein kinase;RTK, receptor tyrosine kinase; S6K, p70 ribosomal S6-kinase; SB-505154, 2-(5-benzo[1,3]dioxol-5-yl-2-tert-butyl-3H-imidazol-4-yl)-6-methylpyridine hydrochloride; SD-282, indole-5-carboxamide; SHIP, Src homology 2 domain-containing inositol 59-phosphatase; STAT, signaltransducer of activators of transcription; Syk, spleen tyrosine kinase; TAK, transforming growth factor-b-activated kinase; TG100-115, 3-[2,4-diamino-6-(3-hydroxyphenyl)pteridin-7-yl]phenol (6,7-Bis(3-hydroxyphenyl)pteridine-2,4-diamine); TGF, transforming growth factor; TLR, Toll-likereceptors; TNF, tumor necrosis factor; VEGF(R), vascular-endothelial growth factor (receptor).

Kinase Inhibitors in Airway Disease 789

Page 3: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

2014; Hekking et al., 2015). There is a need to developmore effective therapies for these patients who consumea disproportionate amount of medical resources.COPD is also very prevalent, affecting about 10% of

people over 45 years in developed countries and in-creasing especially in developing countries (Barneset al., 2015). The increasing world-wide prevalence ofCOPD reflects aging of the population because it is adisease of elderly people. COPD has a high morbidityand is one of the leading causes of hospitalization andloss of time from work. It has a rising mortality and isnow the third most common cause of death in developedcountries (Lozano et al., 2012). No current treatments,including corticosteroids, reduce disease progression ormortality and have relatively little effect (;20% re-duction) in preventing exacerbations, reflecting theirlack of anti-inflammatory effects in this disease. Thereis therefore an enormous unmet need for the develop-ment of new treatments for COPD that target theunderlying inflammatory process and aberrant repairmechanisms (Barnes, 2013b).Although asthmaandCOPDare distinct inflammatory

diseases, some patients with COPDmay have features ofasthma such as eosinophilic inflammation and greaterairway reversibility, whereas some patients with asthmamay have features of COPD, such as neutrophilic in-flammation, reduced reversibility, and reduced respon-siveness to corticosteroids. These patients have beendescribed as having asthma-COPD overlap syndrome,although several phenotypes are included in this label(Bateman et al., 2015; Postma and Rabe, 2015; Barnes,2016). Improved treatment of patients with asthma-COPD overlap is needed (Barnes, 2015b).Thus, asthma and COPD are very common chronic

inflammatory diseases of the respiratory tract withseveral unmet needs for more effective medication.Because chronic inflammation involves the activationof multiple kinase signaling pathways (Cohen, 2002),this has provided a rationale for the development ofselective kinase inhibitors for the treatment of asthmaand COPD, particularly in patients with severe diseasethat cannot be controlled by existing therapies. In viewof the role of protein kinases in chronic inflammationand tissue remodeling, the use of kinase inhibitors is alogical approach. Furthermore, there is increasingevidence that kinases are also involved in corticosteroidresistance, which is a feature of severe asthma andCOPD (Barnes and Adcock, 2009). Development ofkinase inhibitors for inflammatory diseases has provento be problematic because of the adverse effects whenthese drugs are given systemically. Theoretically, thiscould be overcome by the development of inhaledinhibitors, but it has proven difficult to develop kinaseinhibitors that have local activity in the airways. Thisreview summarizes the current evidence for the effectsof protein kinase inhibitors in inflammatory airwaydiseases.

II. Role of Kinases in Asthma and COPD

Both asthma and COPD are characterized by achronic inflammatory response in the respiratory tractthat involves the recruitment and activation of severalinflammatory and immune cells and the secretion ofmultiple inflammatory mediators, including lipid me-diators, cytokines, chemokines, and growth factors, andwell as enzymes that generate inflammatory productsor are involved in tissue remodeling (Barnes, 2008a,b).Many of these inflammatory proteins are regulated byproinflammatory transcription factors, such as nuclearfactor-kB (NF-kB) and activator protein-1 (AP-1) thatare regulated by kinase signaling pathways. Further-more, thesemediators act on surface receptors on targetcells in the airways that also activate kinase pathways.In the case of asthma, allergen may be an importantdriver of the allergic inflammatory response and thisinvolves the crosslinking of allergens with immunoglob-ulin E (IgE) bound to mast cells, with the activation ofkinase s that result in release of preformed mediators,such as histamine and the synthesis and release of lipidmediators, such as cysteinyl-leukotrienes and prosta-glandins (Barnes, 2011; Galli and Tsai, 2012). Allergenis also taken up by dendritic cells and this activateskinase pathways, leading to the recruitment of T-helper2 (Th2) lymphocytes to the airways that drive eosino-philic inflammation and airway hyperresponsiveness,which are the defining features of asthma (Barnes,2011). Innate immunity also plays a key role in asthma,particularly severe and nonallergic asthma that is drivenin innate lymphoid-2 cells (Scanlon and McKenzie,2012). Innate immune mechanisms and the activa-tion of pattern recognition receptors, such as Toll-likereceptors (TLR) that are activated by cigarette smokeand wood smoke (which are key risk factors for thedevelopment of COPD), and by pathogens, such ascolonizing bacteria. Mitogen-activated protein kinases(MAPK) play a key role in the activation of both innateand adaptive immune cells, resulting in the secretionof multiple cytokines, chemokines, and growth factorsthat then activate further waves of inflammation(Arthur and Ley, 2013). COPD represents an acceler-ation of the normal aging process in lungs and thesepathways of cellular senescence are also regulated bymultiple interacting kinase pathways (Mercado et al.,2015).

III. Kinases

Kinases are enzymes that transfer the g-phosphate ofadenosine-triphosphate (ATP) to hydroxyl-bearing sub-strates and thus phosphorylate proteins, lipids, andsugars, which results in activation of enzymes, cellulartranslocation, or interaction with other proteins andplay a critical role in signal transduction. Over 500kinase genes have now been recognized in humans,

790 Barnes

Page 4: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

making up ;2% of the genome (the “kinome”), and over30% of proteins are regulated by phosphorylation(Manning et al., 2002). These are organized into distinctfamilies, such as mitogen-activated protein kinases(MAPK). The majority of kinases phosphorylate serine(Ser) or threonine (Thr) residues (serine/threoninekinases); some act on tyrosine (Tyr) residues (tyrosinekinases), whereas other act on both types (dual-specificity kinases). Tyrosine-kinases may act as recep-tors for ligands such as growth factors, such as epidermalgrowth factor receptor (EGFR). Because kinases playsuch a critical role in signaling, they are carefullyregulated by phosphatases, which remove phosphategroups and thus reverse the effects of a specific kinase.Kinases are comprised of several domains, including awell conserved catalytic domain that binds ATP. Al-though the majority of kinases target proteins, sometarget lipids (such as phosphatidylinositol and sphin-gosine), carbohydrates (such as hexokinase), or nucleo-tides, such as thymidine kinase.Protein kinases are selective for Ser/Thr or Tyr res-

idues in their target proteins and recognize a con-sensus sequence around these residues rather than allof the residues on a particular target protein (Fabbroet al., 2015). However, a particular kinase is usually notspecific for a particular protein but may act on severalproteins sharing this consensus sequence. Pseudosub-strates bind to the kinase but lack the amino acid thatis phosphorylated, so that they effectively act as aninhibitor of the kinase. Kinases are organized intosignaling pathways, whereby one kinase phosphory-lates a downstream kinase, which then phosphorylatesthe next downstream kinase in the sequence. Eachkinase may phosphorylate different kinases so thatseveral signaling cascades may be activated and theremay be crosstalk between different kinase signalingpathways, resulting in amplification or inhibition ofsignaling. This makes signaling pathways highly com-plex, and it is often difficult to predict the effects ofinhibiting a single kinase. Furthermore, there arecomplex feedback inhibitory loops that may result inunexpected effects of interrupting kinase pathways.

A. Kinase Inhibitors

The recognition that kinases are key regulators of cellproliferation, apoptosis, and inflammation and thatmutation of kinase genes is an important mechanismin many immune and inflammatory diseases hasprompted a search for selective small molecule inhibi-tors in many human diseases (Cohen, 2002). Manysmall molecule kinase inhibitors have now been ap-proved for use in cancer (Wu et al., 2015c), and althoughmost of the drug discovery efforts have focused onmalignancy, kinase inhibitors are now seen as impor-tant potential therapies for many chronic inflammatoryand immune disorders (Cohen, 2014). Indeed, kinaseinhibitors are now the second largest group of drugs in

development after G-protein-coupled receptor ligandsand account for ;30% of drug discovery projects inmajor pharmaceutical companies (Wu et al., 2015c).Because both asthma and COPD involve inflammatoryand immune mechanisms, there have been great effortsto develop kinase inhibitors as new therapies, particu-larly targeting areas of unmet need (Adcock et al., 2006).Development of specific kinase inhibitors has proven tobe very difficult because the catalytic site is wellconserved between different kinases and most inhibi-tors target the ATP-binding site. This means that manykinase inhibitors are poorly selective and often haveseveral off-target effects on structurally related kinases,which may increase the risks of adverse effects. Afurther problem with kinase inhibitors in cancer ther-apy is the development of resistance due to sitemutations at the ATP-binding pocket. With chronictreatment of inflammatory diseases there may also bea gradual wearing off of efficacy of kinase inhibitors dueto the upregulation of compensatory pathways. Forexample, p38MAPK inhibitors for rheumatoid arthritismay lose efficacy because of upregulation of otherproinflammatory kinase pathways (Firestein et al.,2008). This might be less of a problem if more upstreamkinases are targeted, although the risks of side effectsmay be even greater.

Most kinase inhibitors so far have been given orally,so that systemic side effects are common and often dose-limiting. As a means of reducing adverse effects, therehas been great interest in the development of allostericinhibitors that target less conserved sites in the kinasedomain or remote sites (Wu et al., 2015b). These drugsmay not need to be of such high potency that is needed tocompete with the high concentrations of ATP at thecatalytic site and may also be more resistant tomutations. Allosteric kinase inhibitors, such as trame-tinib, are now in clinical development, at least for thetreatment of cancer. For example, allosteric inhibitorsthat bind to a distal site have now been discovered forp38 MAPK (Comess et al., 2011). Inhaled delivery mayalso reduce the risk of adverse effects by targeting thedrug to the airways. So far it has proven difficult todevelop inhaled kinase inhibitors that are effectivebecause they do not have topical activity. This is relatedto the fact that kinases are intracellular targets, so thedrug properties that target cell penetration also in-crease oral bioavailability and rapid transport acrossthe lung into the systemic circulation.

IV. Nuclear Factor-kB inhibition

A. Role in Asthma and Chronic ObstructivePulmonary Disease.

The proinflammatory transcription factor NF-kB isactivated in asthma and COPD and switches on multi-ple inflammatory genes, including cytokines, chemo-kines, proteases, and inhibitors of apoptosis, resulting

Kinase Inhibitors in Airway Disease 791

Page 5: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

in amplification of the inflammatory response (Barnesand Karin, 1997; Hart et al., 1998; Di Stefano et al.,2002; Caramori et al., 2003). NF-kB activation, mea-sured by nuclear localization of p65 (RelA), is seenpredominantly in airway epithelial cells and macro-phages andmay be activated by inflammatory cytokines(such as TNF-a and IL-1b), oxidative stress, allergens,and bacteria. NF-kB may play an important amplifyingrole in acute exacerbations of asthma and COPD, whichare commonly triggered by upper respiratory tractviruses, such as rhinovirus (Caramori et al., 2003).NF-kB is activated by rhinovirus in airway epithelialcells (Footitt et al., 2016; Papi et al., 2013) and there ismarked activation of NF-kB in the lungs of patients whodied from asthma (Caramori et al., 2009).The molecular pathways involved in NF-kB activa-

tion include several kinases, which are targets forinhibition. The classic (canonical) pathway for inflam-matory stimuli and infections to activate NF-kB signal-ing involve the IKK (inhibitor of kB kinase) complex,which is composed of two catalytic subunits, IKK-a andIKK-b, and a regulatory subunit, IKK-g (or NF-kBessential modulator) (Hayden and Ghosh, 2012). TheIKK complex phosphorylates NF-kB-bound IkBs, tar-geting them for degradation by the proteasome andthereby releasing NF-kB dimers that are composed ofp65 and p50 subunits, which translocate to the nucleus

where they bind to kB recognition sites in the promoterregions of inflammatory and immune genes, resultingin their transcriptional activation (Fig. 1). This re-sponse depends mainly on the catalytic subunit IKK-b(also known as IKK2), which carries out IkB phosphor-ylation. Although the canonical pathway has beenstudied most extensively in asthma and COPD, thereis a noncanonical (alternative) pathway, which involvesthe upstream kinase NF-kB-inducing kinase (NIK) thatphosphorylates IKK-a homodimers and releases RelBand processes p100 to p52 in response to certainmembers of the TNF family, such as lymphotoxin-b(Sun, 2012). This pathway switches on different genesets and may mediate different immune functionsfrom the canonical pathway. Dominant-negative IKK-binhibitsmost of the proinflammatory functions ofNF-kB,whereas inhibiting IKK-a has a role only in re-sponse to limited stimuli and in certain cells suchas B-lymphocytes. The noncanonical pathway is in-volved in development of the immune system and inadaptive immune responses, but its role in asthma andCOPD has not been explored in detail. The coactivatormolecule CD40 is expressed on antigen-presenting cells,such as dendritic cells and macrophages, activates thenoncanonical pathway when it interacts with CD40Lexpressed on lymphocytes, and may amplify allergicresponses in asthma (Lombardi et al., 2010). CD40

Fig. 1. Nuclear factor-kB signaling pathway. The canonical (classic) pathway is activated by inflammatory cytokines, such as tumor necrosis factor(TNF)-a, interleukin(IL)-1b, and lipopolysaccharide (LPS), leading to phosphorylation of inhibitor of kB kinases [IKKa, IKKb, which are in a complexwith NF-kB essential modulator (NEMO)], resulting in phosphorylation of inhibitor of kB (IkB-b), which is ubiquitinated and degraded by theproteasome releasing p65 and p50 to translocate to the nucleus, where they bind to a kB DNA recognition sequence, leading to the activation ofcytokines, chemokine, and protease genes. The noncanonical pathway is activated by CD40 and lymphotoxin(LT)-b, which activate NF-kB-inducingkinase (NIK), resulting in activation IKKa homodimers, which phosphorylate RelB/p100, generating RelB/p50 complexes that translocate to thenucleus and switches on immune genes. IKK-b may be inhibited by several compounds (shown in the green box), but no inhibitors of the non-canonicalpathway have been identified.

792 Barnes

Page 6: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

stimulation induces emphysema in mice, and solubleCD40L is increased in plasma of COPD patients andcorrelates with the degree of emphysema, suggesting apossible pathogenic role (Shigeta et al., 2012). Indirectevidence for the involvement of the noncanonical path-way is the activation of NIK and phosphorylation ofIKK-a in airway epithelial cells after exposure tocigarette smoke and oxidative stress (Chung et al.,2011). On the other hand, RelB overexpression in miceinhibits cigarette smoke neutrophilic inflammationthrough stimulation of cyclooxygenase-2 (McMillanet al., 2011).

B. Inhibitors.

Inhibition of NF-kB is a logical approach to treatinginflammation in asthma and COPD because there isevidence for its activation and increased activation ofmultiple proinflammatory genes. However, deletion ofNF-kB related genes inmice is lethal or animals developsevere septicemia, indicating that there are risks ofNF-kB inhibition (Hayden and Ghosh, 2012). Cortico-steroids inhibit NF-kB activation in patients withasthma (Hart et al., 2000), but there is resistance tothis effect in patients with severe asthma and COPD(Barnes and Adcock, 2009). Theophylline is also report-ed to inhibit NF-kB in human airway epithelial cellsthrough IKK inhibition, although high concentrationsare needed for this effect (Ichiyama et al., 2001). Severalnaturally occurring compounds, such as plant polyphe-nols, have also been reported to have inhibitory effectson NF-kB. For example, resveratrol inhibits NF-kB andinflammatory cytokine release from COPD epithelialcells in vitro (Donnelly et al., 2004). The green teaextract epigallocatechin-3-gallate inhibits IKK activity(IKK-a as well as IKK-b) in epithelial cells in vitro(Yang et al., 2001).Several IKK-b inhibitors of differing chemical classes

have been developed and in vitro have demonstratedinhibition of p65 nuclear translocation and the activa-tion of inflammatory genes after various stimuli as wellas anti-inflammatory effects in in vivo animal models(Suzuki et al., 2011). Both direct and allosteric inhibitorshave been developed. A decoy oligodeoxyribonucleotideagainst NF-kB markedly inhibits allergic lung inflam-mation in an ovalbumin-sensitized mice with markedinhibition of Th2 cytokine release (Desmet et al., 2004).A selective IKK-b inhibitor IMD-0354 [N-[3,5-bis(trifluoromethyl)phenyl]-5-chloro-2-hydroxybenzamide]also reduced allergic inflammation and airway hyper-responsiveness in this murine model, predominantlythrough inhibition of T cell trafficking (Sugita et al.,2009; Maslanka et al., 2016). Another IKK-b inhibitor[Bay 65-1942 ((7E)-7-[2-(cyclopropylmethoxy)-6-oxocy-clohexa-2,4-dien-1-ylidene]-5-[(3S)-piperidin-3-yl]-4,8-dihydro-1H-pyrido[2,3-d][1,3]oxazin-2-one)] is also ef-fective at inhibiting pulmonary allergic inflammationin a rat model (Ziegelbauer et al., 2005). Yet another

IKK-b inhibitor TPCA-1 is effective in a mouse modelof neutrophilic inflammation induced by inhaled endo-toxin, although it was not effective in a neutrophilelastase-induced inflammation model (Birrell et al.,2006). IKK-b inhibitors have also been ineffective incigarette smoke-induced lung inflammation in mice(Rastrick et al., 2013). Several IKK-b inhibitors sup-pressed the release of cytokines and chemokines fromhuman airway smooth muscle and epithelial cells andwere mimicked by adenovirus mediated delivery of adominant-negative IKK-b inhibitor (Catley et al.,2006; Newton et al., 2007). To avoid potential systemicexposure, an inhaled IKK-b inhibitor (PF-104) wasdeveloped to inhibit neutrophilic lung inflammationinduced by inhaled endotoxin in mice (Sommers et al.,2009). Ainsliadimer-A is found in a Chinese herbalproduct that has been described as a potent andselective inhibitor of IKKa/b, thus inhibiting theclassic and alternative pathways of NF-kB (Donget al., 2015). It inhibits the release of inflammatorycytokines after LPS injection in mice, so has potentialas an anti-inflammatory drug.

Because NF-kB plays a critical role in host defenseagainst pathogens, there are concerns that inhibitorsmay increase the risk of infection. There is also anuncertain relationship to cancer, because NF-kB maydrive tumorigenisis, but its inhibitionmay also increasecell proliferation. Limited numbers of clinical trialshave been conducted. IMD-1041, a prodrug of theIKK-b inhibitor IMD-0354, was tested in patients withCOPD for anti-inflammatory effects, but the trial wasterminated because of side effects of the medication(ClinicalTrials.gov: NCT00883584). Other IKK-b inhib-itors are in clinical trials for other inflammatorydiseases, but no results have been published and noproducts have been approved. It is likely that an inhaledformulation will be necessary to reduce adverse effects.

V. Mitogen-Activated Protein Kinase Inhibition

MAPK are a large family of highly conserved serine/threonine protein kinases that are involved in signaltransduction (Johnson and Lapadat, 2002). Althoughoriginally shown to be activated by mitogens, they areactivated by many stimuli, including cytokines, growthfactors, and various stresses (osmotic, oxidative, heatshock) and result in diverse cellular responses, such asproliferation, differentiation, mitosis, cell survival, ap-optosis, and inflammatory gene expression. MAPK arenormally inactive in the resting cell and become acti-vated by phosphorylation, usually of a threonine and atyrosine residue. Several scaffold proteins are associ-ated with MAPK pathways and appear to direct signal-ing within the cell by directing proteins to particularcellular compartments and may also modulate signal-ing. Typically MAPKs operate in a three-tiered cascaderegulated by successive phosphorylation starting with

Kinase Inhibitors in Airway Disease 793

Page 7: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

activation of MAP kinase kinase kinases (MAP3K), thenMAP kinase kinases (MAP2K), and finally MAPK (Fig.2). ThemajorMAPK pathways involved in inflammatorydiseases are ERK (extracellular regulating kinase), p38MAPK, and JNK (c-Jun NH2-terminal kinase), but mostattention has focused on inhibition of the p38 MAPKpathway, because several small molecule inhibitors werediscovered and there has been particular interest in p38inhibitors for the treatment of asthma and COPD.

A. p38 Mitogen-Activated Protein Kinase

The p38 MAPK family comprises a-, b, g-, and d-isoforms, which are highly conserved proline-directedprotein kinases that are activated by several extracel-lular stresses, including oxidative and osmotic stressand inflammatory cytokines. P38 MAPK is activated byphosphorylation of Thr180 and Tyr182 in the Thr-Gly-Tyrmotif of the activation loop, which allows p38 tobind ATP at its catalytic binding site to transfer theg-phosphate group from ATP to the protein targeted forphosphorylation. P38 MAPK is activated by upstreamkinases MKK3, MKK4, and MKK6, which are them-selves activated by MAP3Ks, such as ASK1(apoptosissignal-regulatory kinase 1), TAK1 (transforminggrowth factor-b-activated kinase-1), MLK3 (mixed-lineage kinase 3), and MAP kinase kinase kinase(MEKK3), which are activated by many stimuli,

including inflammatory cytokines, growth factors, andoxidative stress. P38 MAPK target many proteins,including other kinases, transcription factors, andcytoskeletal proteins, resulting in multiple inflamma-tory effects. P38 MAPK activates over 60 downstreamtargets, so may be a key point for inhibition (Trempolecet al., 2013). P38a and p38b isoforms are widelydistributed and target a downstream target MAP-activated protein kinase 2 (MAPKAPK2 orMK2), whichmediates many of these effects. P38a, via activation ofMK2, increases inflammation by stabilizing mRNAtranscripts of some cytokines, such as TNFa, by phos-phorylating adenosine-uracil–binding proteins, such astristeraprolin, that bind to the adenosine-uracil–rich 39-untranslated regions of mRNAs to regulate their sta-bility (Dean et al., 2004; Brook et al., 2006). MK2 alsophosphorylates heat shock protein(hsp)-27, which hasbeen used as a biomarker of p38a activation. P38apotentiates the proinflammatory effects of NF-kB byphosphorylation of histone H3 at the promoter sites onproinflammatory cytokine and chemokine genes to in-crease binding of NF-kB binding to their promoterregions (Saccani et al., 2002). P38g and p38d isoformshave a more restricted distribution, particularly mac-rophages, and target mainly transcription factors.

1. Role in Asthma and Chronic Obstructive Pulmo-nary Disease. There is considerable evidence that p38

Fig. 2. Mitogen-activated protein kinase signaling pathways. MAPK signaling is activated by external stimuli, such as growth factors and cellularstress, and activates a three-tiered cascade with MAPK kinase kinases (MAP3K), activating MAPK kinases (MAP2K) and finally MAPK. The majorMAPK pathways involved in inflammatory diseases are ERK (extracellular regulating kinase), p38 MAPK, and JNK (c-Jun NH2-terminal kinase).Upstream kinases include TGFb-activated kinase-1 (TAK1) and apoptosis signal-regulating kinase-1 (ASK1). Downstream pf p38 MAPK is MAPKactivated protein kinase 2 (MAPKAPK2 or MK2). Several classes of inhibitor are now developed and shown in green boxes.

794 Barnes

Page 8: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

MAPK is activated in asthma and COPD and playsan important role in driving chronic inflammation aswell as corticosteroid insensitivity (Chung, 2011). P38ais activated in airway epithelial cells and macrophagesby lipopolysaccharide (LPS) and cigarette smoke andmediates the release of inflammatory proteins, suchas CXCL8 (IL-8), IL-6, TNF-a, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Se-lective inhibitors of p38a inhibit the release of thesecytokines from human macrophages activated by ciga-rette smoke and LPS (Koch et al., 2004; Smith et al.,2006). However, p38a inhibitors did not cause maximalinhibition in macrophages and it is possible that otherisoforms, such as p38d are important in macrophages(Smith et al., 2006). Alveolar macrophages from pa-tients with severe asthma show increased activation ofp38, which may reflect a reduction in the endogenousinhibitor of p38, MAPK phosphatase-1 (MKP1, alsoknown as dual specificity phosphatase-1) (Bhavsaret al., 2008). In mice exposed to cigarette smoke and toozone, a p38a inhibitor SD-282 significantly suppressedthe neutrophilic inflammatory response, whereas corti-costeroids were ineffective (Medicherla et al., 2007;Williams et al., 2008). This suggests that p38a mayplay an important role in pulmonary neutrophilic in-flammation in COPD. Indeed, phosphorylated p38ashows increased expression in alveolar macrophages,epithelial cells, and CD4+ and CD8+ lymphocytes inlungs from COPD patients (Renda et al., 2008; Gaffeyet al., 2013).P38a is also an important mediator of inflammation

in response to upper respiratory tract virus infectionsand so may play a role in acute exacerbations of asthmaand COPD. Rhinovirus-induced release of chemokinesfrom alveolar macrophages is associated with activa-tion of p38a (Hall et al., 2005) and a p38a inhibitorsuppresses release of inflammatory cytokines fromhuman airway epithelial cells infected with rhinovirus(Griego et al., 2000). Haemophilus influenzae, bacteriacommonly colonizing the lower airways and causingacute exacerbations of COPD patients, also activatesp38 in airway epithelia cells andmediates the release ofCXCL8 (Wang et al., 2003). H. influenzae also stimu-lates the secretion of the mucin MUC5AC from epithe-lial cells via p38a activation, suggesting that inhibitionof this pathway may reduce mucus hypersecretion inCOPD patients (Shen et al., 2008).P38 activation is also important in mediating allergic

inflammation. An inhaled p38a antisense oligonucleo-tide was effective in inhibiting eosinophilic inflamma-tion and airway hyperresponsiveness after allergenchallenge in an ovalbumin-sensitized mouse model(Duan et al., 2005b). A selective p38a inhibitor, indole-5-carboxamide (SD-282), similarly reduced allergic in-flammation, airway hyperresponsiveness and airwayremodeling in a chronic allergen exposure mouse model(Nath et al., 2006). P38a may play an important role in

allergic inflammation through the activation of thetranscription factor GATA3, which regulates the Th2cytokines IL-4, IL-5, and IL-13, because p38 phosphory-latesGATA3 in the cytoplasmof Th2 cells so that it is ableto translocate to the nucleus to activate Th2 cytokinegenes (Maneechotesuwan et al., 2007). A p38 inhibitor, bypreventing GATA3 phosphorylation, inhibits the expres-sion of Th2 cytokines (Maneechotesuwan et al., 2009).P38 also prolongs eosinophil survival by enhancing theeffects of IL-5 and a p38 inhibitor promotes apoptosis(Kankaanranta et al., 1999).

P38a appears to play an important role in the corti-costeroid insensitivity that occurs in severe asthmaand COPD (Barnes, 2013a). The corticosteroid insensi-tivity induced by IL-2 and IL-4 or by IL-13 in peripheralblood mononuclear cells (PBMC) is reversed by a p38ainhibitor (Matthews et al., 1999; Irusen et al., 2002).Corticosteroids increase the expression of MKP-1 andthus inhibit p38 activation. In mice with deletion ofMKP-1 corticosteroid lose their anti-inflammatory ef-fects (Abraham et al., 2006). In addition, TNF-a andIL-1b result in phosphorylation of the glucocorticoidreceptor (GRa) at Ser211 via p38a activation, preventingtranslocation to the nucleus (Szatmary et al., 2004).P38a is activated in PBMC from patients with severeand steroid-resistant asthma and is correlated withGRa phosphorylation and reduced responsiveness tocorticosteroids (Mercado et al., 2012; Li et al., 2015).Furthermore, a p38a inhibitor is able to restore cortico-steroid responsiveness in these cells. Similarly, in PBMCfrom COPD patients the reduced responsiveness tocorticosteroids was reversed by treatment with a p38ainhibitor (Khorasani et al., 2015). P38g also phosphory-lates GRa and reduces nuclear translocation, resulting incorticosteroid insensitivity (Mercado et al., 2011b). P38gis increased in PBMC of patients with severe asthma andmay be reduced by treatment with the long-acting b2-agonist formoterol, which thereby reduces corticosteroidresistance in these cells (Mercado et al., 2011b).

B. p38 Inhibitors

There is a strong rationale for the development of p38inhibitors to reduce chronic inflammation (both neutro-philic and eosinophilic), to reduce corticosteroid resis-tance, and to reduce structural changes, particularly inpatients with severe asthma and COPD. Selective p38ainhibitors have been rather effective in asthmatic andCOPD cells in vitro and in in vivo animal models of thesediseases. Several p38 inhibitors have been developed forclinical use, and these drugs aremainly selective for p38aand p38b isoforms (Norman, 2015). These drugs havebeen studied largely in patients with rheumatoid arthri-tis, but have been disappointing because they have beendose-limited by side effects and have lost effusivenessafter repeated administration due to the development ofcompensatory signaling pathways. Most of the clinicalstudies have been inCOPD,where there is a great need to

Kinase Inhibitors in Airway Disease 795

Page 9: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

develop effective anti-inflammatory treatments becausecorticosteroids are largely ineffective, and eight p38ainhibitors are known to have entered clinical trials inCOPD (Norman, 2015). An oral p38a inhibitor dilmapi-mod (SB-681323) significantly reduced phosphorylatedhsp-27 (demonstrating target engagement) and TNF-arelease from circulating leukocytes of COPD patients,demonstrating proof of mechanism (Singh et al., 2010)Losmapimod is an oral dual p38a/b inhibitor that had noeffect on sputum neutrophils in a phase II study of about300 COPD patients over 12 weeks but reduced phosphor-ylation of hsp-27 and reduced plasma fibrinogen byapproximately 11% (Lomas et al., 2012). In the largestclinical trial of about 600 COPD patients studied over24 weeks, three doses of losmapimod were ineffective onexercise tolerance (6 minute walking distance), althoughthere was a trend for reduction in exacerbations;however, there were side effects at the highest dose,particularly dermatological effects (Watz et al., 2014).In a post hoc analysis in patients with low bloodeosinophils (,2%), there was a dose-related reductionin exacerbations with a 55% reduction at the highestdose and a small improvement in FEV1 (15mg)(Marks-Konczalik et al., 2015). PH-797804 [(aS)-3-{3-bromo-4-[(2,4-difluorobenzyl)oxy]-6-methyl-2-oxopyridin-1(2H)-yl}-N,4-dimethylbenzamdePF-03715455=1-[5-tertbutyl-2-(3-chloro-4-hydroxyphenyl)pyrazol-3-yl]-3-[[2-[[3-[2-(2-hydroxyethylsulfanyl)phenyl]-[1,2,4]triazolo[4,3-a]pyridin-6-yl]sulfanyl]phenyl]methyl]urea] is an oralp38a-selective inhibitor, which increased trough FEV1

values and dyspnea at some doses in a 6-week study of230 patients with COPD (MacNee et al., 2013). Because ofthe problems of side effects at high oral doses of p38inhibitors, several inhaled drugs have been developed, butno results in airway disease have been reported (Millanet al., 2011). An inhaled p38 inhibitor, PF-03715455[1-[5-tertbutyl-2-(3-chloro-4-hydroxyphenyl)pyrazol-3-yl]-3-[[2-[[3-[2-(2-hydroxyethylsulfanyl)phenyl]-[1,2,4]tri-azolo[4,3-a]pyridin-6-yl]sulfanyl]phenyl]methyl]urea],had no significant effect on sputum inflammatory bio-markers although an oral p38 inhibitor had some sup-pressive effect in the same population (Singh et al., 2015).RV-568 is a potent inhaled p38 inhibitor that inhibits allfour isoforms of p38 and also the Src family kinase Hckand is referred to as a narrow spectrum kinase inhibitor,which significantly increased FEV1 and reduced sputummalondealdehyde after 2weeks of administration (Russellet al., 2013).No clinical studies of p38 inhibitors in asthmahave been reported, andmost of the drugs in developmentfor COPD have now been discontinued.

C. Jun NH2-terminal Kinase Inhibitors

JNK is an important activator of the proinflammatorytranscription factor activator protein-1 (AP-1), whichregulates many inflammatory genes in asthma andCOPD. Sensitized mice exposed to inhaled allergenshow increased activation of JNK in airway epithelial

cells and lung. Furthermore, a JNK inhibitor SP600125(1,9-pyrazoloanthrone) suppressed the inflammatoryresponse to allergen and reduced mucus hypersecretionand airway hyperresponsiveness, suggesting a role forJNK in allergic inflammation (Nath et al., 2005; Wuet al., 2015a). TNF-a prolongs the survival of humaneosinophils via activation of JNK, which thereby sus-tains eosinophilic inflammation (Kankaanranta et al.,2014). JNK may also play a role in airway remodeling,because SP600125 also reduces airway smooth muscleproliferation after chronic inhaled allergen exposure inmice (Eynott et al., 2003). In JNK-deficient mice, thefibrosis after chronic exposure to allergen (house dustmite) was significantly reduced (van der Velden et al.,2014). JNK regulates airway smooth muscle prolifera-tion and increased collagen synthesis through theactivation of transforming growth factor (TGF)-b (Xieet al., 2007) and this is mediated through the activationof AP-1 (Lee et al., 2006b). JNK also plays a role inCD40-mediated class switching in B cells, andSP600125 inhibits the formation of IgE transcripts inB cells (Jabara and Geha, 2005). JNK activation is alsoinvolved in corticosteroid resistance in severe asthmapatient, because AP-1 competes with GRa and there isevidence for JNK activation in PBMC of patients withsteroid-resistant asthma (Adcock et al., 1995; Lane et al.,1998). The corticosteroid resistance induced by IL-2 andIL-4 in PBMC ismediated by activation of JNK, resultingin phosphorylation of Ser226 in GR-a to prevent itsnuclear localization (Kobayashi et al., 2012). JNK activa-tion is also involved in the corticosteroid resistanceinduced by rhinovirus infect ion of airway epithelial cells,which is reduced by a JNK inhibitor and completelyreversed by a combination of JNK and IKK-b inhibitors(Papi et al., 2013). JNK activation may also play a role inCOPD. Cigarette smoke cytotoxicity in airway epithelialcells is mediated via activation of JNK (Lee et al., 2008).

The activation of JNK and AP-1 in models of asthmaandCOPDand its role in stimulating cytokine release andcorticosteroid resistance suggests that JNK inhibitorsmay have therapeutic value in the treatment of asthmaandCOPD,which is supported by thebeneficial effects of aselective JNK inhibitor SP600125 in these models.SP600125 is a competitive inhibitor of ATP for bothJNK1 and JNK2. No clinical trials of JNK inhibitors havebeen undertaken, presumably because of toxicity issues.

Curcumin is a plant polyphenol found in turmeric,which inhibits JNK, and is currently in clinical trials forchronic inflammatory diseases, such as psoriasis; how-ever, it is nonselective and targets several other signal-ing pathways, such as NF-kappaB and nuclear factor(erythroid-derived 2)-like 2 (Hatcher et al., 2008).

D. Extracellular Regulating Kinase/MAP KinaseKinase Inhibitors

Several studies demonstrated activation of ERK1 andERK2 in asthma and COPD models. Activation of mast

796 Barnes

Page 10: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

cells may release IL-1 and IL-6 through activation ofERK1/2, which is inhibited by genistein, a flavonoid thathas additional pharmacological effects (Kim et al., 2014).IL-13 causes mucus hypersecretion as a result of in-creasedMUC5AC via ERK activation (Kono et al., 2010).Epithelial growth factor receptor (EGFR)-inducedmucussecretion is alsomediated via ERK and inhibited by theERK kinase (MEK1) inhibitor PD-98059 [29-Amino-39-methoxyflavone] (Li et al., 2012). Although p38 MAPKappears to be the dominant regulator of cytokine re-lease from monocytes, in lung and alveolar macro-phage ERK appears to be predominant and ERKinhibitors are more effective than p38 inhibitors insuppressing cytokine secretion (Koch et al., 2004;Tudhope et al., 2008). The ERK pathway is activated bygrowth factors and may be involved in proliferation ofairway smooth muscle cells and other structural changesin the airways.The proinflammatory effect of ERK suggest that an

ERK inhibitor may be of potential benefit, particularlyin reducing mucus secretion. The MEK1/ERK inhibitorPD-98059 is effective as an inhibitor of this pathwayand acts as a noncompetitive allosteric inhibitor with ahigh degree of specificity. However, this compound isnot suitable for in vivo or clinical studies because of itsvery low solubility. Several other MEK-1 inhibitors,such as trametinib and solumetinib, have been tested inclinical trials in cancer and have shown some efficacy insolid tumors and are relatively well-tolerated, but so farno studies in COPD or asthma have been reported(Wang et al., 2007; Fremin and Meloche, 2010). In-terestingly, macrolide antibiotics inhibit cigarettesmoke-induced CXCL8 secretion by epithelial cells,which is mediated via ERK phosphorylation, althoughmacrolides have other effects on signaling such asinhibition of NF-kB (Shinkai et al., 2007).

E. Apoptosis Signal-Regulating Kinase-1 Inhibition

Apoptosis signal-regulating kinase-1 (ASK1) is anupstream kinase (MAP3K5) that activates both p38MAPK and JNK and is activated by oxidative andendoplasmic reticulum stress (Hattori et al., 2009).Thioredoxin is normally bound to ASK1 to suppressits activity, and ASK becomes activated when thiore-doxin is reduced by oxidative stress. ASK1mediates thecytotoxicity of cigarette smoke in airway epithelial cells,which is inhibited by the closely associated antioxidantthioredoxin (Lee et al., 2008). ASK1 is a good potentialtarget for COPD, and several small molecule inhibitorshave been identified (Starosyla et al., 2015). Interest-ingly, ASK1 knockout mice are viable, whereas p38aand JNK knockout mice are embryonically lethal.Clinical trials are underway with ASK1 inhibitors, suchas GS-4997, in diabetic renal disease (Lin et al., 2015),but so far these drugs have not been explored in COPDor asthma. Thioredoxin is the endogenous inhibitor ofASK1 but is difficult to deliver intracellularly.

F. TGFb-Activated Kinase-1 Inhibition

TGFb-activated kinase-1 (TAK1) is another up-streamMAP kinase (MAP3K7) that is tightly regulatedand activates both ERK and p38a MAPK pathways aswell as NF-kB, thus playing a pivotal role in innateimmunity in response to TGFb, TNFa, IL-1b, and TLRvia the adapter protein MyD88 (Sato et al., 2005;Ajibade et al., 2013). Proliferation of human airwaysmooth muscle in response to PDGF is significantlyreduced by the selective TAK1 inhibition, with adominant-negative inhibitor and small molecule inhib-itor, 5Z-7-oxozeaenol (Pera et al., 2011). TAK1 inhibi-tion also inhibits cigarette smoke-induced release ofCXCL8 from airway smooth muscle cells (Pera et al.,2012). H. influenzae activates NF-kB and p38 MAPK inhuman airway epithelial cells via the activation of TAK1and NIK (Shuto et al., 2001). TAK1may also activate theadaptive immune system through the activation ofNF-kB in T-lymphocytes (Sun et al., 2004). TAK1 istherefore an attractive target for inhibition, because bothMAPK and NF-kB pathways would be suppressed.Several small molecule TAK1 inhibitors are in develop-ment (mainly for cancer therapy), but there are limiteddata on kinase selectivity and they have not yet advancedto clinical studies (Kilty and Jones, 2015).

G. MAPKAPK2 Inhibition

MAPKAPK2 (MK2) is activated directly via p38aMAPK, and inhibition of this downstream kinase mightavoid some of the toxicity problems seen with p38ainhibitors (Gaestel, 2013). MK2 appears to play a role inprolonging NF-kB activation in Th2 cells and thereforeamplifies allergic inflammation (Gorska et al., 2007). MK2is also important in mediating the increased release ofCXCL8 in response to cigarette smoke in human airwaysmooth muscle cells (Moretto et al., 2012). However, ATP-competitive MK2 inhibitors have poor selectivity and lowsolubility (Schlapbach and Huppertz, 2009). By contrast,noncompetitive inhibitors lookmore attractive and severalcompounds are in development but have not yet enteredclinical trials (Anderson et al., 2009; Fiore et al., 2016).

VI. Phosphoinositide-3-kinase Inhibition

PI3Ks are a family of lipid kinases that generate lipidsecond messengers that regulate a number of cellularevents, including innate and adaptive immune responses,growth, differentiation, cell motility and survival, andintracellular trafficking (Vanhaesebroeck et al., 2012;Hawkins and Stephens, 2015). PI3Ks catalyze the phos-phorylation of the inositol ring of cell membrane phos-phatidylinositols to generate lipid second messengers.They are classified into three classes based on thephosphorylated lipids generated. Class I PI3Ks catalyzethe production of phosphatidylinositol-(3,4,5)-trisphos-phate (PIP3) from PIP2 and are heterodimers comprising

Kinase Inhibitors in Airway Disease 797

Page 11: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

a p110 catalytic subunit with four isoforms (designatedP110a, b, g, and d) together with an adaptor subunitof either p85 (class IA) that are triggered by recep-tor tyrosine kinases (RTKs) and include PI3Ka, b, andd, or of p110/p84 (class IB) that are mainly triggeredby G-protein-coupled receptors that include PI3Kg(Vanhaesebroeck et al., 2016). PIP3 accumulates in theplasma membrane and recruits pleckstrin homologydomain-containing proteins, including phosphoinositide-dependent kinase-1 (PDK1) and Akt (also called proteinkinase B). PIP3-activated PDK1 then phosphorylates Aktat Thr308 site and, together with Ser473 phosphorylationbymammalian target of rapamycin complex 2 (mTORC2),this ensures full activation of Akt. P-Akt then triggersmultiple downstream signaling pathways (Fig. 3). Down-stream signaling fromp-Akt is complex but amajor targetin mTORC1, which activates cellular senescence and isinvolved in accelerated aging (Johnson et al., 2013).PI3Ka and PI3Kb are expressed in most cell types,whereas PI3Kg and PI3Kd are localized predominantlyin leukocytes. By contrast, there is relatively little in-formation about the function of Class II and III PI3Ks.Class II PI3Ks include PI3K-C2a, PI3K-C2b, and PI3K-C2g. PI3K-C2a and PI3K-C2g are the best characterizedisoforms, and they appear to contribute to vesiculartrafficking by regulating the production of PtdIns(3)P,

but because there are no selective inhibitors they aredifficult to investigate. Class III includes only one mem-ber, Vps34, which is involved in vesicular trafficking, TLRsignaling, and autophagy via PtdIns(3)P generation.

The PI3K pathway is tightly regulated by phos-phatases, particularly PTEN (phosphatase and ten-sin homolog deleted from chromosome 10), the majorendogenous PI3K inhibitor, which is expressed inall cell types and dephosphorylates PIP3 to PIP2,thus turning off PI3K activation (Carracedo andPandolfi, 2008; Worby and Dixon, 2014). PTEN actsas a tumor suppressor and deletions are commonlyfound in cancers such as lung cancer, but it is nowrecognized as an important regulator of inflamma-tion and cell proliferation. The Src homology 2domain-containing inositol 59-phosphatase (SHIP) alsoinhibits PI3K signaling and is found mainly in hema-topoietic cells, including macrophages (Backers et al.,2003).

A. Role in Asthma and Chronic ObstructivePulmonary Disease

There is increasing evidence that PI3K plays a keyrole in asthma and COPD through activation of in-flammation, corticosteroid resistance, and cellularsenescence, resulting in accelerated aging (Fig. 3)

Fig. 3. Phosphoinositide-3-kinase signaling pathways. PI3K is activated by extracellular signals, Including growth factors and chemokines, resultingin activation of Akt (protein kinase B), then tuberous sclerosis complex (TSC)1/2, then mammalian target of rapamycin complex-1 (mTORC1), which inturn activates ribosomal S6-kinase (S6K), which reduces sirtuin-1 (SIRT1) and histone deacetylase(HDAC)2 and impairs autophagy. There is crosstalkto other signaling pathways because Akt1 activates IKKb and mTORC1 activates JNK. Oxidative stress activates PI3K signaling though inhibition ofPTEN (phosphatase and tensin homolog deleted from chromosome 10) and SHIP1 (Src homology 2 domain-containing inositol 59-phosphatase-1). AMPkinase (AMPK) inhibits mTORC1 via an effect on TSC1/2. Several classes of inhibitor (shown in green boxes) may reduce PI3K signaling.

798 Barnes

Page 12: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

(Marwick et al., 2010b). These effects are mediated viaClass I PI3Ks, and the potential role of Class II and IIPI3Ks is unknown. A nonselective PI3K inhibitorLY294002 [2-Morpholin-4-yl-8-phenylchromen-4-one]given by inhalation inhibits the allergic inflammatoryresponse to allergen in sensitized mice, with reduction ineosinophils, Th2 cytokines, and airway hyperresponsive-ness (AHR) (Duan et al., 2005a). Similar results wereobtained with a dominant-negative inhibitor of the regu-latory subunit p85, indicating that Class IA PI3Ks areinvolved and may determine the switch from Th1 to Th2cytokines (Myou et al., 2003). More specifically Pi3Kdplays a key role in mast cell degranulation after cross-linking of IgE receptors, an effect blocked by inactivationof PI3Kd andby the selectivePI3Kd inhibitor IC87114 (Aliet al., 2008; Kim et al., 2008). In the murine model ofasthma, intratracheal administration of IC87114 sup-pressed the allergic response, Th2 cytokines, and AHR(Lee et al., 2006a), and these effects are mirrored inPI3Kd-inactivated mice (Nashed et al., 2007). In anAspergillus fumigatus model of allergic inflammation inmice, PI3Kd inhibition suppressed the allergic response,and this effect was mediated through inhibition ofendoplasmic reticulum stress, which is increased inasthma and results in NF-kB activation (Lee et al.,2016). PI3Kmayalso play a role in increasing contractilityof canine airway smoothmuscle (Halayko et al., 2004) andthe increased proliferation of airway smooth muscle inresponse to growth factors such as TGF-b (Goldsmithet al., 2006).PI3K activation is also important in COPD. Total

PI3K activity measured as p-Akt is markedly in-creased in peripheral lungs and macrophages of pa-tients with COPD, and there is increased expression ofthe PI3Kd isoform (Marwick et al., 2009; To et al.,2010). In vitro oxidative stress increases p-Akt inperipheral bloodmonocytes and alveolarmacrophages,which is prevented by a PI3Kd but not PI3Kg inhibitor(Marwick et al., 2010a). PI3Kg knockout mice showreduced neutrophil migration and activation, as wellas impaired T-lymphocyte and macrophage function(Medina-Tato et al., 2007), and PI3Kg is the pivotalsignaling pathway mediating neutrophil recruitment tothe lung after instillation of chemokines (Thomas et al.,2005). PI3Kg plays an important role in chronic in-flammatory and immune diseases (Costa et al., 2011).PI3Kd is also involved in the inflammatory responseand may cooperate with PI3Kg in the activation ofneutrophils, for example (Condliffe et al., 2005). BothPI3Kg and PI3Kd inhibitors improve the accuracy ofmigration in COPD neutrophils (Sapey et al., 2014). Anaerosolized PI3Kg/d inhibitor [TG100-115 (3-[2,4-diamino-6-(3-hydroxyphenyl)pteridin-7-yl]phenol (6,7-Bis(3-hydroxyphenyl)pteridine-2,4-diamine))] waseffective in cigarette smoke-induced pulmonary inflam-mation in mice, which is corticosteroid resistant, andalso in the allergic inflammation model (Doukas et al.,

2009). PI3Kd is also involved in corticosteroid resis-tance after oxidative stress, which is mediated byreduced histone deacetylase-2 (HDAC2), and PI3Kdinhibitors may reverse corticosteroid resistance inpatients with COPD by increasing HDAC2 expres-sion. Inhibition of PI3Kd with IC87114 reversescorticosteroid-resistance to cigarette smoke exposurein mice (To et al., 2010), and PI3Kd-inactivated mice donot develop corticosteroid-resistance after cigarettesmoke exposure, whereas PI3Kg-inactivated animalsdevelop corticosteroid-resistant inflammation as nor-mal (Marwick et al., 2009). PI3Ka is involved in cellproliferation and survival and is activated in early lungcancer cells and so may contribute to the increased riskof developing lung cancer in patients with COPD. PI3Kactivation in COPD also activates mTOR, measured byphosphorylation of p70 ribosomal S6-kinase (S6K),which is increased in COPD lungs and PBMC (Mitaniet al., 2015). S6Knot only reducesHDAC2 but also leadsto activation of JNK, increased c-Jun, and activation ofAP-1, which also contributes to corticosteroid resis-tance. PI3K activation also drives cellular senescenceand accelerated aging in COPD lungs through theactivation of mTOR, which plays a pivotal role inaccelerated aging (Johnson et al., 2013; Mercadoet al., 2015). This leads to a reduction in the keyantiaging molecule sirtuin-1, which is reduced in thelungs of patients with COPD (Rajendrasozhan et al.,2008; Nakamaru et al., 2009). The same PI3K-mTORpathway may also lead to the comorbidities associatedwith COPD, such as ischemic heart disease, type2 diabetes, osteoporosis, chronic renal disease, anddementia (Barnes, 2015a). For example, endothelialprogenitor cells, which are important for repairingdamage to endothelial cells to maintain normal vascu-lar function, show accelerated aging and decreasedsirtuin-1 due to activation of PI3K (Paschalaki et al.,2013).

B. Inhibiting Phosphoinositide-3-kinase-Akt-Mammalian Target of Rapamycin Signaling

In view of the critical role of PI3K-Akt-mTORsignaling it is not surprising that there has been anintense effort to discover drugs that inhibit thispathway. This has been mainly in the field of cancerin view of the key role of PI3K in cell proliferation, andthe PI3K-Akt-mTOR pathways are the most fre-quently dysregulated in human solid tumors. Thepathways are also relevant to chronic degenerativediseases that involve accelerated aging (Johnson et al.,2013) and in the treatment of inflammatory andimmune diseases (Hawkins and Stephens, 2015).There has been great interest in the potential forinhibitors of the PI3K pathways in the treatment ofsevere asthma and COPD (Ito et al., 2007).

1. Phosphoinositide-3-kinase Inhibitors. Nonselectiveinhibitors of Class I PI3K include wortmannin and

Kinase Inhibitors in Airway Disease 799

Page 13: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

LY294002, which are useful in in vitro and in vivoanimal studies but are not suitable for clinical usebecause of their poor pharmacokinetics and high tox-icity. Several pan-class I PI3K inhibitors have beendeveloped for use in malignancy, including hematologicmalignancies and solid tumors, but so far none havebeen approved for clinical use, mainly because oftoxicity issues (Dienstmann et al., 2014). This has ledto a search for isoform-selective inhibitors. However,PI3K isoforms may compensate for each other, at leastin cancer, through feedback mechanisms, where block-ing one isoform leads to a compensatory increased in thenoninhibited isoforms. For example a PI3Kb-selectiveinhibitor leads to an increase in PI3Ka (Schwartz et al.,2015). PI3Ka inhibitors have been developed for treat-ing solid tumors but may result in hyperglycemia,because they interfere with insulin signaling. PI3Kginhibitors have been considered, but so far no drugshave been developed for clinical use (Ruckle et al., 2006)as potential anti-inflammatory treatments (Ameriksand Venable, 2009). PI3Kd inhibitors have been de-veloped for the treatment of B cell malignancies and thePI3Kd-selective inhibitor idelalisib (CAL-101) has beenapproved for the treatment of some B cell malignancies(Vanhaesebroeck and Khwaja, 2014). However, in thetreatment of chronic lymphocytic leukemia, side effectssuch as leukopenia, colitis, and skin rashes may developwith idelalisib (Brown, 2015; Louie et al., 2015). There isa good rationale for combining inhibition of PI3Kg andd, and several combined PI3Kg/d inhibitors are indevelopment, such as IPI-145 (duvelisib), which iseffective in several inflammation and autoimmuneanimalmodels and is now in early clinical development(Winkler et al., 2013). To overcome dose-limiting sideeffects for the treatment of asthma and COPD, theseinhibitors have been developed for inhaled delivery. Acombined PI3Kg/d inhibitor TG100-115 is effective inmodels of allergic asthma and cigarette smoke-inducedinflammation in mice and is not in clinical development(Doukas et al., 2009). There is a good rationale for thiscombination because PI3Kg inhibition should be anti-inflammatory and inhibit cell trafficking into the lungs,whereas PI3Kd inhibition should be effective in reversingcorticosteroids resistance in COPD and severe asthma.InhaledPI3Kd inhibitors, suchasGSK2269557 [2-(6-(1H-indol-4-yl)-1H-indazol-4-yl)-5-((4-isopropylpiperazin-1-yl)methyl)oxazole hydrochloride], are also in developmentfor the treatment of COPD and severe asthma (Wilsonet al., 2013). Another approach to inhibition of PI3Kd isthe use of existing therapies. Theophylline was shown toincreaseHDAC2 and reverse corticosteroids resistance inmacrophages form COPD patients (Ito et al., 2002; Cosioet al., 2004) and is effective through the selective in-hibition of oxidant-activated PI3Kd in relatively lowconcentrations that are lower than those used forits bronchodilator effect (To et al., 2010). A clinicaltrial of low-dose theophylline combined with inhaled

corticosteroids is now underway in COPD patients(Barnes, 2013c; Devereux et al., 2015). Nortriptyline, atricyclic antidepressant, and solithromycin, a novelmacrolide antibiotic, are also effective in reversingcorticosteroid-resistance in COPD cells through inhibit-ing PI3Kd (Mercado et al., 2011a; Kobayashi et al., 2013).

2. Akt-Mammalian Target of Rapamycin Inhibitors.Akt is a downstream target of PI3K activation, andseveral oral inhibitors have been developed for use incancer therapy. Perfosine blocks p-Akt and is in severalclinical trials for malignancy, but this drug appears toalso target several other pathways, leading to thedevelopment of more selective inhibitors (Alexander,2011). Rapamycin (sirolimus) is a macrolide that bindsto FK506-binding protein-12, which functions as anallosteric inhibitor of mTORC1. It is already in clinicaluse as an immunosuppressant in transplantation andautoimmunity, although it has frequent and severeadverse effects that limit its chronic use. There isparticular interest in finding less toxic analogs ofrapamycin with improved pharmacokinetics (rapalogs),because rapamycin extends life span in mammalsby targeting cellular senescence and autophagy(Wilkinson et al., 2012; Lamming et al., 2013). Indeed,a recent clinical study suggests that a rapalog ever-olimus is able to reduce immunosenescence in elderlypeople (Mannick et al., 2014). Results in cancer therapyhave been disappointing (Chiarini et al., 2015), and thismay reflect the fact that blocking mTOR, which reducesS6K activation, results in blocking of a feedback in-hibitory loop that results in PI3K-Akt activation inmalignant cells. The reduction in autophagy by rapa-mycin may also work against the killing of malignantcells. Dual inhibitors of mTORC1 and 2, such asAZD2014 [3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin-7-yl)-N-methylbenzamide], may bemore effective because this blocks the feedback loopwhere mTORC2 activates Akt (Dienstmann et al.,2014). This has suggested that dual inhibitors of PI3Kand mTOR may be more beneficial, because this avoidsthe PI3K-Akt feedback loop. There is close structuralhomology between these kinases, and several dualinhibitors have been developed and are now in clinicaltrials (Chiarini et al., 2015). None of these treatmentshas so far been tested in airway disease. However,rapamycin is very effective in inhibiting mTOR activa-tion in COPD lung and PBMC, which results in reducedcorticosteroids resistance (Mitani et al., 2015). In addi-tion, rapamycin is effective in suppressing allergicinflammation in sensitized mice and inhibits eosinophildifferentiation (Mushaben et al., 2013; Hua et al., 2015).Interestingly, rapamycin switches the phenotypeof lymphocytes from Th2 to regulatory T cells (Foxp3+)(Kim et al., 2010).

3. Activating Endogenous Inhibitors. Adenosinemonophosphate-activated kinase (AMPK) is an impor-tant regulator of PI3K signaling through the inhibition

800 Barnes

Page 14: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

of mTORC1 via phosphorylation of the tumor suppres-sor complex TSC1/2 and plays an important role in theregulation of metabolism, cell energetics, andmitochon-drial function, but may also have antiaging effectsthrough increasing sirtuin-1 and anti-inflammatoryeffects (Steinberg and Kemp, 2009). This has prompteda search for AMPK activators, which should have valuein asthma and COPD (Scarpulla, 2011; Salt andPalmer, 2012). Metformin, a biguanide, is commonlyused to treat type 2 diabetes and works partly throughactivation of AMPK via changes in nucleotide levels.Experimentally it has been shown to extend life span inmice (Martin-Montalvo et al., 2013). It also has poten-tial as an anti-inflammatory drug (Salt and Palmer,2012). Metformin and the more specific AMPK activator59-aminoimidazole-4-carboxamide-1-b-4-ribofuranosidereduce allergic inflammation in a chronic allergic mousemodel, and this is reproduced in AMPK-deficient(AMPKa12/2) mice (Park et al., 2012). 59-Aminoimida-zole-4-carboxamide-1-b-4-ribofuranoside inhibits ciga-rette smoke-induced release of CXCL8 from A549 cells,and AMPKa12/2mice have reduced emphysema aftercigarette smoke exposure, indicating that activation ofthis pathway has therapeutic potential in asthma andCOPD (Lee et al., 2015). Flavones, such as quercitin,and resveratrol also activate AMPK. Resveratrol hasanti-inflammatory effects in COPD airway epithelialcells, although this may be mediated through severalmechanisms (Donnelly et al., 2004). More direct activa-tors of AMPK are now in development, such as thecompound A-769662, which is entering clinical trials fordiabetes (Goransson et al., 2007).Another approach is through activation of the phos-

phatases PTEN and SHIP, which inhibit PI3K signal-ing. It has been difficult to identify selective activatorsof PTEN, which may reflect the complexity of its regu-lation and may interactions (Song et al., 2012). SHIP ismore amenable to activation and a small moleculeactivator of SHIP-1, AQX-1125, and is effective inreducing p-Akt and reducing mediator release, mastcell activation, and leukocyte chemotaxis in vitro(Stenton et al., 2013b) and inflammation in allergen-induced and cigarette smoke-induced pulmonary in-flammation in mice in vivo (Stenton et al., 2013a). Arecent clinical trial of oral AQX-1125 in patients withmild asthma showed a significant reduction in laterresponse after allergen challenge and a (nonsignificant)reduction in inflammatory biomarkers (Leaker et al.,2014). The drug is well tolerated, suggesting that itmight be a potential treatment of asthma and COPD.Another downstream inhibitor of PI3K signaling

pathways is glycogen synthase-3b (GSK3b), which isconstitutively active but inhibited by phosphorylationby Akt and ERK1/2 as a result of oxidative stress(Takahashi-Yanaga, 2013). Inactivated GSK3b(p-GSKb-Ser9) is increased in PBMC, macrophages,and airway epithelial cells of patients with COPD,

especially those with severe disease (Ngkelo et al.,2015). This is associated with corticosteroid resistanceas a result of reduced HDAC2 in these cells, which ismimicked by GSK3b knockdown and a GSK3b inhibitorCT99021 [6-[[2-[[4-(2,4-Dichlorophenyl)-5-(5-methyl-1H-imidazol-2-yl)-2-pyrimidinyl]amino]ethyl]amino]-3-pyridinecarbonitrile]. This suggests that GSK3bactivators may be beneficial in reversing corticosteroidresistance, but it has been difficult to identify suchcompounds. In addition, GSK3b regulates many differ-ent signaling pathways, including NF-kB, and itsactivation might amplify inflammation.

VII. Janus-Activated Kinase Inhibition

The Janus kinase (JAK)-signal transducer of activa-tors of transcription (STAT) signaling pathway regu-lates the expression of multiple inflammatory andimmune proteins and has been implicated in manyinflammatory diseases (O’Shea et al., 2015). This hasled to the development of JAK inhibitors (Jakinibs),which are a new class of anti-inflammatory drug nowused for the treatment of inflammatory diseases, suchas rheumatoid arthritis, autoimmune diseases, hema-topoietic diseases, and cancer, with several inhibitors inclinical development and some already in clinical use(Kontzias et al., 2012; Menet et al., 2013). Afteroccupation of the cytokine receptor, the associated JAKsbecome activated and both phosphorylate each otherand the intracellular tail of their receptors, thuscreating docking sites for cytoplasmic STATs, which inturn directly bind as dimers to DNA and regulate geneexpression. There are four JAKs (JAK1-3) and Tyk2,which selectively bind different receptor chains (Fig. 4).The selective usage of JAKs by different receptorsexplains their distinct in vivo roles. There are sevenSTATs (STAT1, 2, 3, 4, 5a, 5b, 6) that are activated bydifferent combination of JAKs, but JAKs may alsodirectly affect genes through histone phosphorylation,which changes chromatin structure to influence geneexpression. Various polymorphisms of STAT-encodinggenes have been described in cancer and inflammatoryand immune diseases.

A. Role in Asthma and Chronic Obstructive Pulmo-nary Disease. There is recent evidence that JAKs areinvolved in asthma and COPD. Many of the cytokinesinvolved in asthma, including the Th2 cytokines (IL-4,IL-5, IL-9, and IL-13), GM-CSF, and thermic stromallymphopoietin, activate JAK-STAT signaling (Pernisand Rothman, 2002; Ghoreschi et al., 2009). Thereceptors for these cytokines involve activation of allof the JAKs, suggesting that a broad spectrum inhibitoris more likely to be useful. JAK3 expression is restrictedto hematopoietic cells and colocalizes with JAK1, trans-mitting signals through the common g-chain of thecytokine receptors for IL-2, IL-4, IL-7, IL-9, IL-15, andIL-21. Tofacitinib and pyridone-6, which are pan-JAK

Kinase Inhibitors in Airway Disease 801

Page 15: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

inhibitors, are effective and reduce AHR in a murinemodel of allergic inflammation (Kudlacz et al., 2008;Matsunaga et al., 2011). There has been concern thatblocking JAK2 may have inhibitory effects on hemato-poiesis, so there has been a search for more selectiveinhibitors. JAKs play a key role in differentiation of Th2cells and a JAK1/3 inhibitor (R256) blocked the differ-entiation of Th2 cells and the phosphorylation of STAT5and STAT6, but not of Th1 or Th17 cells in vitro (Ashinoet al., 2014). In vivo R256 inhibited allergic inflamma-tion, AHR, and mucus hypersecretion in a mouse modelwithout any changes in Th2 cytokine concentrations.JAKs are also involved in the mechanisms of COPD

and several cytokines implicated in COPD activateJAK-STAT signaling, including IL-6, IL-12, andinterferon(IFN)-g, which activate JAK1, JAK2, andTyk2. In mice with hyperreactivity of the receptor forIL-6 (gp130), there was an increase in STAT3 andpulmonary inflammation that were prevented inSTAT3 knockout mice (Ruwanpura et al., 2012). Fur-thermore STAT3 was increased in the lungs of COPDpatients and correlated with inflammation but not withairway obstruction. STAT1 and STAT3 are phosphory-lated in the lungs of severe COPD patients to a greaterextent than in smokers and nonsmokers, indicatingactivation of several JAKs (Yew-Booth et al., 2015).STAT4 is also increased in the airway of COPDpatients, presumably in response to IL-12 or IL-23 (DiStefano et al., 2004). The chemokines CXCL9, CXCL10,and CXCL11 are all increased in the airways of COPDpatients and target a common receptor, CXCR3, whichis involved in the recruitment of T-cells (particularlyCD8+ cells) into the lung (Costa et al., 2008). These

chemokines are induced by IFNg through the activa-tion of JAKs and phosphorylation of predominantlySTAT1 (Tudhope et al., 2007). Pan-JAK inhibitorsare effective in reducing IFNg-induced secretion ofCXCR3-activating chemokines from human airwayepithelial cells, demonstrating their potential as anti-inflammatory treatments in COPD (Fenwick et al.,2015).

B. Jakinibs. There is a good rationale for the use ofJAK inhibitors in both asthma and COPD, because theymediate the effects of several mediators involved inthese diseases. Several oral JAK inhibitors (jakinibs)are now approved and in clinical trial for the treatmentof other inflammatory diseases, including rheumatoidarthritis, inflammatory bowel disease, and psoriasis(Kontzias et al., 2012; Menet et al., 2013; Schwartzet al., 2016). Over 20 pan-JAK inhibitors and isoform-selective inhibitors have now been developed. Tofaciti-nib is JAK1/3 selective and indicated for rheumatoidarthritis and psoriasis, whereas JAK2-selective inhibi-tors are indicated for hematologic malignancies andmyeloproliferative disease. Ruxolitinib is JAK1/2 selec-tive and indicated for rheumatoid arthritis and myelo-proliferative disease. However, the selective JAKinhibitors are not specific so tend to affect other JAKs,increasing the risk of side effects. Thus pan-JAKinhibitors may cause anemia, neutropenia, and thom-bocytopenia through inhibition of JAK2, which medi-ates the effects of erythropoietin, thrombopoietin, andGM-CSF. Side effects of jakinibs after systemic admin-istration are dose limiting and include increased infec-tions, diarrhea, increased cholesterol levels, andlymphomas, so a strategy to reduce these adverse

Fig. 4. JAK-STAT signaling pathways. Janus-activated kinases (JAK1-3, Tyk2) are activated by several extracellular stimuli and phosphorylateseveral signal transducer of activators of transcription (STAT) proteins, which translocate to the nucleus and activate target genes resulting in cellsurvival, activation of antiviral mechanisms, inflammation, and Th1 and Th2 immunity.

802 Barnes

Page 16: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

effects is to deliver the drugs topically. Thus, topicaltofacitinib and ruxolitinib have been developed for skinapplication for the treatment of psoriasis. Inhaled pan-JAK inhibitors, such as LAS194046 and R256, are nowin development for asthma and COPD and have shownefficacy in models of allergic asthma (Ramis et al.,2014). An inhaled pan-JAK inhibitor (VR560) markedlyreduced the allergic inflammatory response in micesensitized and exposed to chronic house dust mites,inhibited Th2 cytokines and IL-17, and inhibitedSTAT3 phosphorylation (Wiegman et al., 2015). Thissuggests that inhaled pan-JAK inhibitors may havegood potential in asthma and COPD and clinical trialsare now in progress.

VIII. Spleen Tyrosine Kinase Inhibition

Spleen tyrosine kinase (Syk) is a nonreceptor cyto-plasmic protein tyrosine kinase that is primarilyexpressed in hematopoietic cells (particularlyB lymphocytes) and links immune cell receptors tomultiple intracellular signaling pathways that regulatemany cellular responses, including cytokine release andcell survival (Mocsai et al., 2010; Riccaboni et al., 2010).Syk appears to have many diverse functions, includinginnate and adaptive immunity, cell adhesion, and organdevelopment and has been implicated in many autoim-mune, inflammatory, and malignant diseases. Immu-noreceptors include B-cell and T-cell receptors andFc-receptors, which associate with transmembraneproteins that have immunoreceptor-tyrosine-based ac-tivation motifs (ITAMs), which are short peptide

sequences containing two tyrosine residues 6–12 aminoacids apart. ITAM tyrosines are phosphorylated afterligand binding, resulting in the recruitment and acti-vation of Syk or the related enzyme z-activated proteinkinase of 70 kDa, which results in recruitment of otherkinases and the activation of downstream signalingpathways, includingNF-kBand PI3K pathways (Fig. 5).

A. Role in Asthma and Chronic Obstructive PulmonaryDisease. In a chronic allergic murine model of asthmathere was an increase in Syk activation in epithelial cells,and a Syk inhibitor [N-[4-[6-(cyclobutylamino)-9H-purin-2-ylamino]phenyl]-N-methylacetamide (NVP-QAB-205)]significantly reduceAHRafter allergen and the enhancedAHR after additional exposure to diesel particulates andozone, although there was no reduction in inflammatorycells (Penton et al., 2013). A Syk inhibitor [6-(5-fluoro-2-(3,4,5-trimethoxyphenylamino) pyrimidin-4-ylamino)-2,2-dimethyl-2H-pyrido[3,2-b][1,4]oxazin-3(4H)-one (R406)]also prevents activation of sensitized mast cells anddendritic cells from mice, with a reduction in AHR andallergic inflammation in vivo (Matsubara et al., 2006a,b).Similar effects are seen with an inhaled Syk antisense,indicating the specificity of the small molecule Sykinhibitors (Stenton et al., 2000, 2002). An oral inhibitor[2-(7-(3,4-dimethoxyphenyl)-imidazo[1,2-c]pyrimidin-5-ylamino)-nicotinamide HCl (Bay 61-3606)] is also effec-tive in reducing the allergic reaction to inhaled allergensin rats and also in preventing passive cutaneous anaphy-laxis and mast cell activation (Yamamoto et al., 2003).This Syk inhibitor also inhibits the activation of humanbasophils. The reduction in AHR seen after a Sykinhibitor may be due to an effect on airway smooth

Fig. 5. Spleen tyrosine kinase signaling. Syk is activated by immune receptors via phosphorylation by Lyn kinase and may activate PI3K orphospholipase Cg, resulting in activation of immune cells, such as mast cells, B-lymphocytes, and T-lymphocytes. Syk may also be activated byintegrins. Several Syk inhibitors have been identified (shown in green box).

Kinase Inhibitors in Airway Disease 803

Page 17: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

muscle contractility, because Syk-deficient mice do notdevelop AHRafter allergens and the contractility of smallairways to spasmogens is reduced (Wang et al., 2015).This is mimicked by treatment with a Syk inhibitor (Moyet al., 2013). Syk is also activated by infection of humanairway epithelial cells after rhinovirus binding to ICAM-1and results in the activation of p38 MAPK and PI3Kactivation, with release of mediators such as CXCL8 andVEGF and so may play a role in acute exacerbations ofboth asthma and COPD (Lau et al., 2008). Syk alsoregulates internalization of rhinovirus via clathrin-mediated endocytosis, which involves ITAMs previouslyonly associated with immune cells (Lau et al., 2011).B. Spleen Tyrosine Kinase Inhibitors. Because of

the key role of Syk in mast cell and immune cellsignaling, the effects of Syk inhibition on the allergicinflammatory response, AHR, and the inhibition ofrhinovirus signaling there is a persuasive case fordevelopment of Syk inhibitors (Geahlen, 2014). Severalorally administered Syk inhibitors have been developedfor the treatment of rheumatoid arthritis and B cellleukemias (Norman, 2014b; Thorarensen and Kaila,2014) For example, fostamatinib (R788) has shownefficacy in rheumatoid arthritis patients (but not thoserefractory to biologics), and the most frequent adverseeffects are gastrointestinal symptoms, hypertension,and neutropenia, but no serious infections (Nijjaret al., 2013). To avoid side effects, topical formulationshave been developed. A nasal spray formulation of R112was weakly effective in patients with seasonal allergicrhinitis exposed to outdoor allergens (Meltzer et al.,2005) and transiently decreased the concentration ofPGD2 in nasal lavage after nasal allergen challenge buthad no effect on either histamine or mast cell tryptase(Guyer et al., 2006). A novel Syk inhibitor suitable forinhalation, LAS189386 [1-{2-[(1S,4S)-2,5-diazabicyclo[2.2.1]hept-2-yl]pyridin-4-yl}-N-pyrazin-2-yl-1H-indazol-3-amine], delivered to ovalbumin-sensitized BrownNorway rats showed good inhibition of the early re-sponse to allergen and of mast cell degranulation,without any effects on systemic mast cells (Ramiset al., 2015), and so is in clinical development forasthma.

IX. Receptor Tyrosine Kinases

Growth factors are cytokines that induce structuralchanges, such as fibrosis and tissue remodeling, and acton high-affinity surface receptors that have tyrosinekinase activity. Of the 90 human protein tyrosinekinase genes, over half encode receptor tyrosine kinases(RTK) (Hubbard and Till, 2000). RTKs are important innormal tissue growth, development, and repair afterinjury and have been closely linked to cancers. MostRTKs function as single unit receptors, although some(e.g., insulin receptor) are multimeric. An N-terminalextracellular domain interacts with the protein ligand

(growth factor or hormone), and the C-terminalintracellular domain contains a relatively well-conserveddomain that catalyzes receptor autophosphorylation withreceptor dimerization and phosphorylation of RTK sub-strates, which include proteins with Src homology-2domains and phosphotyrosine domains (Lemmon andSchlessinger, 2010). There are several distinct familieswith RTKs, but those most relevant to airway disease arethe epidermal growth factor receptor (EGFR, ErbB)family, fibroblast growth factor receptor (FGFR) family,vascular-endothelial growth factor receptor (VEGFR)family, and platelet-derived growth factor receptor(PDGFR).

A. Role in Asthma and Chronic ObstructivePulmonary Disease

EGFR play an important role in mediating mucushypersecretion and mucus metaplasia, with goblet cellproliferation in both asthma and COPD (Nadel andBurgel, 2001). EGFR shows increased expression inairway epithelial cells of asthmatic airways and isassociated with CXCL8 release and neutrophilic in-flammation (Hamilton et al., 2003). EGFR plays apivotal role in mucus hypersecretion (chronic bronchi-tis) in COPD through increased expression of MUC5ACandmay be activated by ligand such as TGF-a that maybe generated by neutrophilic inflammation and throughnonligand mechanism such as oxidative stress(Takeyama et al., 2000; Takeyama et al., 2001; Shaoand Nadel, 2005). There is increased expression ofEGFR in airway epithelia cells of COPD airways(de Boer et al., 2006).

Basic fibroblast growth factor-2 (bFGF) also stimu-lates airway smooth muscle proliferation and acts inconcert with other growth factors (Bosse and Rola-Pleszczynski, 2008). Chronic allergen exposure in sensitizedmice induces bFGF expression in airway macrophages andis associated with airway remodeling, both of whichwere reduced after treatment with a corticosteroid(Yum et al., 2011).

Increased angiogenesis contributes to the structuralremodeling in asthma, with increased numbers ofbronchial vessels, which show increased leakiness(Paredi and Barnes, 2009). These effects are largelymediated by VEGF secreted from airway epithelial cellsand airway smooth muscle cells, T cells and eosinophils(Hoshino et al., 2001). There is increased expression ofVEGF, VEGF receptors, and angiogenin-1 in airways ofasthmatic patients that correlates with increased vas-cularity (Feltis et al., 2006). By contrast, VEGF levelsare reduced in COPD sputum and lung parenchyma(Taraseviciene-Stewart et al., 2006; Kanazawa et al.,2003) and may be linked to the development of emphy-sema through apoptosis of endothelial cells. VEGFlevels are reduced in the epithelia lining fluid ofperipheral airways of patients with COPD and normalsmokers (Kanazawa et al., 2014).

804 Barnes

Page 18: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

The platelet-derived growth factor (PDGF) familyincludes the isoforms A-D, which may exist as homo- orheterodimers and activate the receptors PDGFR-a and -b,which dimerize. (PDGF)-BB is a potent stimulator ofairway smooth muscle proliferation (Hirst et al., 1996)and may act synergistically with other growth factorssuch as endothelin-1 (Yahiaoui et al., 2006). The pro-liferative effect of PDGF is mediated via activation ofSTAT-3 and the small GTPase Rac-1 (Simeone-Penneyet al., 2008). Overexpression of PDGF-BB in airwayepithelial cells induces airway smooth muscle (ASM)proliferation and airway hyperresponsiveness in amousemodel (Hirota et al., 2011). Chronic allergen exposureinduces secretion of PDGF into BAL fluid in this model.

B. Receptor Tyrosine Kinase Inhibitors

Several small molecule EGFR inhibitors, such asgefitinib and erlotinib, have been developed for thetreatment of solid cancers, including lung cancers,where mutations of EGFR are commonly seen. Exper-imental studies with EGFR reduces mucus secretion.An EGFR inhibitor AG1478 [N-(3-chlorophenyl)-6,7-dimethoxy-4-quinazolinamine] is effective at inhibitingMUC5AC secretion induced by LPS, TNF-a, or cigarettesmoke from human airway epithelial cells in vitro andin cigarette smoke-induced MUC5AC secretion in ratsin vivo (Hegab et al., 2007; Takezawa et al., 2016).However, a potent and selective EGFR inhibitor BIBW2948 given by inhalation for 4 weeks in patients withCOPD failed to show any reduction in epithelial mucus,despite evidence for target engagement measured byEGFR internalization (Woodruff et al., 2010). In addi-tion, there was a reduction in lung function, and liverenzyme elevation was seen in some patients. VEGFRinhibitors, such as cediranib, have been developed forcertain solid cancers but have not been tested in asthmaand would be contraindicated in COPD because experi-mental VEGFR inhibitors have induced emphysema.Several drugs inhibit PDGFR and usually inhibit othertyrosine kinases. For example, masitinib targets PDGFRand c-Kit and tyrosine kinases, improves asthma controlin severe asthma patients, and reduces the requirementfor oral corticosteroids, although it was associated withskin rashes and edema (Humbert et al., 2009). Ninteda-nib inhibits PDGFR, but also VEGFRandFGFR, and hasbeen approved for pulmonary fibrosis but has not beenstudied in airway disease (Keating, 2015). Similarly,imatinib inhibits PDGFR and c-Kit, as well as ABL-kinase, the key target in chronic myeloid leukemia, buthas not been studied in asthma or COPD.

C. c-Kit Inhibitors

c-Kit (also known as CD117 and SCFR) is a proto-oncogene, a tyrosine kinase receptor for stem cell factor,which is an essentialmast cell growth factor. It is encodedby the KIT gene and expressed on hematopoietic pro-genitor cells and mature mast cells (Lennartsson and

Ronnstrand, 2012). Activating mutations of KIT areassociated with mastocytosis, with an accumulation ofmast cells in tissues. Silencing of c-Kit with a smallinterference RNA reduces the inflammatory response toallergen in amurinemodel of asthma, with a reduction inTh2 cytokines and eosinophils (Wu et al., 2012). Imatinib,an ABL-kinase inhibitor for chronic myeloid leukemia, isalso an inhibitor of c-Kit tyrosine kinase activity and iseffective in a mouse model of asthma with hypersensitiv-ity to cockroach allergen (Berlin and Lukacs, 2005). Thissuggests that more selective c-Kit inhibitors may beuseful in the treatment of asthma (Reber et al., 2006).As discussed above, masitinib inhibits c-Kit and hasproduced benefits in asthma. Other receptor tyrosinekinases, such as sunitinib and nilotinib, have beendeveloped for cancer therapy and may also be effectivebut have dose-limiting side effects (von Mehren, 2006).

X. Transforming Growth Factor-bReceptor Kinase

Transforming growth factor(TGF)-b is a member of alarge superfamily, which includes bone morphogenicproteins, activins, and inhibins. It plays an importantrole in tissue repair, proliferation, and cell differentia-tion (Massague, 2012). It is implicated in several chroniclung diseases, including asthma and COPD, where it isinvolved in tissue remodeling (Aschner and Downey,2016). It is often associated with tissue fibrosis throughthe induction of the growth factor connective tissuegrowth factor (also known as CCN2) (Ihn, 2002). TGF-balso plays an important role in immunoregulation and issecreted by subtypes of regulatory T cells (Levings et al.,2002). There are three isoforms of TGF-b (TGF-b1, TGF-b2, TGF-b3), secreted noncovalently bound to latency-associated peptide, which dissociate to release activeTGF-b with acidification, oxidative stress, certain integ-rins, and the action of MMP-2 and MMP-9, so that TGFbis activated at sites of inflammation (Yu and Stamenkovic,2000). Activated TGF-b binds to activate TGF-b type I(T-bRI) via phosphorylation by T-bRII, which has Ser/Thr kinase activity. Phosphorylation of T-bRI thenrecruits Smad proteins, enabling Smad phosphorylationand nuclear translocation, resulting in activation ofvarious genes. There are eight Smad proteins in humans,including receptor-Smads (1, 2, 3, 5, 8), co-Smad4, andinhibitory Smads (6, 7). TGF-b signaling may also beactivated in a Smad-independent manner by MAPkinases, PI3K and Src-family kinases.

A. Role in Asthma and Chronic ObstructivePulmonary Disease

TGF-b plays a key role in the airway remodeling ofasthma (Aschner and Downey, 2016). TGF-b1 levels areincreased in asthmatic airways and there is increasedTGF-b in the submucosa of patients with asthma(Kokturk et al., 2003). TGF signaling may promote

Kinase Inhibitors in Airway Disease 805

Page 19: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

ECM deposition, airway smooth muscle proliferation,and collagen deposition after allergen challenge. TGF-bmay be derived from eosinophils, mast cells, macro-phages, epithelial cells, or fibroblasts (Minshall et al.,1997; Halwani et al., 2011). It is increased particularlyin severe asthma (Balzar et al., 2005). TGF-b inducesproliferation of human airway smooth muscle cells viaactivation of multiple kinases, including PI3K, ERK,JNK and by NF-kB activation (Goldsmith et al., 2006).There is increased expression of TGF-b in epithelial

cells from small airways of COPD patients (Takizawaet al., 2001; Wang et al., 2008) and this may becompounded by downregulation of inhibitory Smads(Smad6, Smad7) as a result of exposure to cigarettesmoke (Springer et al., 2004). TGF-b is also expressed inmacrophages in the walls of small airways of patientswith COPD (de Boer et al., 1998). Oxidative stress is apotent stimulus to increased TGF-b activation andexpression (Koli et al., 2008) and TGF-b is activatedfrom its latent precursor byMMP-9, which is secreted inCOPD (Yu and Stamenkovic, 2000; Dallas et al., 2002).

B. T-bRI Kinase Inhibitors

Several small molecule inhibitors of T-bRI kinase havebeen developed, such as SB-431542 [4-[4-(1,3-benzodioxol-5-yl)-5-(2-pyridinyl)-1H-imidazol-2-yl]benzamide] andSB-505154 [2-(5-benzo[1,3]dioxol-5-yl-2-tert-butyl-3H-imidazol-4-yl)-6-methylpyridine hydrochloride], whichinhibit TGF-b signaling and Smad activation (Lapinget al., 2002; DaCosta Byfield et al., 2004). For example,these drugs are effective in the bleomycin model of lungfibrosis (Koh et al., 2015). In a rat model of chronicallergen exposure, a T-bRI kinase inhibitor, SD-208 [2-(5-chloro-2-fluorophenyl)pteridin-4-yl]pyridin-4-yl-amine],reduced airway hyperresponsiveness and collagen de-position, but there was no reduction in airway smoothmuscle proliferation (Leung et al., 2006). However, aT-bRI kinase inhibitor was effective in reducing humanASM proliferation in vitro (Xie et al., 2007). Clinicaldevelopment of these inhibitors has proven problematicbecause of adverse effects, so no clinical trials appear tohave been conducted. A T-bRI kinase inhibitor, galuni-sertib, was entered into clinical trials for cancers but hadmarked cardiac toxicity that has slowed further develop-ment (Herbertz et al., 2015). Inhibiting TGF-b signalingmay have unwanted effects through inhibiting regulatoryT cells activity, so that targeting fibrosis may be betterachieved by targeting the downstream target connectivetissue growth factor (Jun and Lau, 2011).

XI. Protein Kinase C Inhibition

Protein kinase C (PKC) is a family of protein kinasesthat regulate a variety of downstream proteins via Ser/Thr phosphorylation. PKCs are activated by diacylgly-cerol, generated via phospholipase C activation, byphospholipids, or by calcium ions to regulate signal

propagation within cells (Rosse et al., 2010). There are15 subtypes of PKC, organized into three main families,determined by the method of activation. On activation,PKCs translocate from the cytoplasm to the cell mem-brane, where they remain activated for a long time andmediate myriad functions, including cell proliferation,receptor activation, and inhibition and contraction andproliferation of smooth muscle.

A. Role in Asthma and Chronic ObstructivePulmonary Disease

PKCs have multiple signaling roles relevant toairway disease (Dempsey et al., 2007). PKC isoformsare involved in bronchoconstriction, as well as pro-liferation of airway smoothmuscle cells. PKC activationby the phorbol ester phorbol 12,13-dibutyrate causes aslowly developing and sustained contraction of humanairway smooth muscle that is blocked by the PKCinhibitor staurosporine (Rossetti et al., 1995). SeveralPKC isoforms are detectable in ASM (Webb et al., 1997;Donnelly et al., 1995). PKC-z plays a role in theproliferation of ASM cells in response to PDGF (Carlinet al., 2000). PKC activation in response to phorbolesters and acetylcholine results in uncoupling ofb2-receptors in ASM, because diacylglycerol mediatesphosphorylation of the b2-receptor and the stimulatoryG-protein Gs (Grandordy et al., 1994). PKC is involvedin the activation of murine Th2 cells through thephosphorylation and activation of voltage-gated cal-cium channels (CaV1.1-1.4) and PKC inhibition inhibitscytokine release from Th2 cells (Robert et al., 2014).PKC-d is known to activate mTOR and suppression ofPKC-d by the inhibitor rottlerin inhibits PI3K-mTORsignaling after allergen exposure in a murine model ofasthma (Choi et al., 2013). PKC-d is involved in theactivation of mast cells via IgE-receptors and regulatesthe synthesis and secretion of cysteinyl-leukotrienes(Cho et al., 2004a,b). PKC-u is involved in activation ofT-cells and IL-2 secretion so its inhibition causesimmunosuppression (Chand et al., 2012). PKC-d andPKC-z are involved in eosinophil migration (Langloiset al., 2009). Allergen challenge in patients with asthmaresults in long-term activation of PKC-z in eosinophils(Evans et al., 1999). PKC is also involved in the mucussecretion response and expression of MUC5AC afterexposure to human neutrophil elastase (Shao andNadel, 2005).

B. Protein Kinase C Inhibitors

The multiple roles of PKC are bronchoconstriction,inflammatory cell activation, mucus secretion, andremodeling and suggest that PKC inhibitors may havetherapeutic potential in themanagement of asthma andCOPD. Nonselective inhibitors, such as staurosporine,are too toxic for human administration, but severalisoform-selective PKC inhibitors have been developedand entered clinical trials in various diseases, including

806 Barnes

Page 20: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

cancer, cardiovascular disease, diabetes, transplanta-tion, and neuropsychiatric diseases (Mochly-Rosenet al., 2012). Most clinical trials have been negativeand there have been several unexpected adverse effects.No clinical trials have been reported in asthma andCOPD, and there are no PKC drugs currently approvedfor human therapy.

XII. Other Kinase Targets

Several other kinases may be potential therapeutictargets because they are involved in chronic inflamma-tion or have effects on ASM.

A. Rho-Associated Protein Kinase Inhibition

Rho-associated protein kinases (ROCK1 and ROCK2)are Ser/Thr kinases that are mainly involved in regulat-ing cytoskeletal function and thus the shape and move-ment of cells (Riento and Ridley, 2003). ROCK istherefore important in migration of inflammatory cellsto sites of inflammation. ROCK1 is the major down-stream target of the small GTPase RhoA. ROCK1 andROCK2 phosphorylate actin and result in myosin lightchain phosphorylation, resulting in contraction and in-creased contractility of smooth muscle. Thus ROCKplays a key role in determining contractility in bovineASM (Gosens et al., 2004). ROCKs are thought tomediate some of the pleiotropic effects of statins.The ROCK inhibitor Y-27632 administered by inhala-

tion inhibits eosinophilic inflammation, AHR, and air-way remodeling after chronic allergen exposure insensitized guinea pigs (Schaafsma et al., 2008a; Possaet al., 2012; Righetti et al., 2014). This drug inhibitsAHR, bronchoconstriction, and eosinophilic inflamma-tion in a mouse model of acute allergic inflammation(Henry et al., 2005). Another ROCK inhibitor, fasudil,reduces airway inflammation and AHR induced by ozoneexposure inmice (Kasahara et al., 2015). There is growingevidence that ROCK inhibitors inhibit fibrosis and somayreduce airway remodeling in airway diseases (Knipe et al.,2015). This suggests that ROCK inhibitors might beuseful as bronchodilators and anti-inflammatory andantifibrotic therapy, but clinical development hasbeen slow (Fernandes et al., 2007; Schaafsma et al.,2008b). Y-27632 [(R)-(+)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide dihydrochloride] is notvery selective for ROCK and may inhibit other kinases. Ithas not been developed clinically because of toxicity.Fasudil is a more selective ROCK inhibitor that has beenapproved in Japan for the treatment of cerebral vaso-spasm. There are no reported clinical studies in asthma orCOPD. It is possible that there may be a positive in-teraction between a ROCK inhibitor and existing bron-chodilator therapy, but there are no studies exploring this.

B. Interleukin-1 Receptor-AssociatedKinase Inhibition

IL-1 receptor-associated kinases (IRAKs) are impor-tant enzymes in the signaling pathways from IL-1 andIL-18 receptors and from TLRs and are thereforeupstream of NF-kB and MAPK activation. Of the fourIRAKs identified, IRAK-4 appears to be the mostimportant in inflammation and is therefore a potentialtarget for anti-inflammatory therapy (Wang et al.,2009). In humans, mutations in IRAK-4 have beenassociated with susceptibility to bacterial infections.Small molecule IRAK-4 inhibitors are in clinical devel-opment and have been facilitated by the discovery of thecrystal structure of IRAK-4 (Chaudhary et al., 2015). Anaturally occurring compound lancemaside C inhibitsmacrophage activation induced by LPS via TLRthrough inhibition of IRAK-4 (Joh and Kim, 2010). AnIRAK-4 inhibitor is effective in suppressing release ofchemokines form airway epithelial cells of patients withCOPD in vitro (Fenwick et al., 2013). To date no clinicalstudies with IRAK-4 inhibitors have been reported.

C. Src Family Kinase Inhibition

Proto-oncogene tyrosine protein kinase Src is a non-receptor tyrosine kinase linked to cell proliferation andmalignancy but also involved in smoothmuscle functionand inflammation and may be activated by growthfactors, adhesion molecules, and oxidative stress(Roskoski, 2015). Src family kinases play a key role insensitizing airway smoothmuscle andmay play a role inAHR via activation of ROCK and is reversed by the Srcinhibitor PP2 (Shaifta et al., 2015). TNF-a disruptsairway epithelial barrier function and this is mediatedvia Src activation and prevented by a Src inhibitorSU6656 [2,3-dihydro-N,N-dimethyl-2-oxo-3-[(4,5,6,7-tetrahydro-1H-indol-2-yl)methylene]-1H-indole-5-sulfonamide] (Hardyman et al., 2013). The Src inhibitorPP2 and siRNA knockdown of Src prevents rhinovirus-induced release of CXCL8 from human airway epithelialcells (Bentley et al., 2007). Analysis of an asthma proteininteractome suggests that Src plays a central role andmay be a good target to inhibit (Randhawa and Bagler,2012). Several Src inhibitors, such as dasatinib, havebeen developed for the treatment of hematologic malig-nancies but inhibit other kinases and have frequent sideeffects and have not been tested in airway disease.

D. Hematopoietic Cell Kinase Inhibition

Hematopoietic cell kinase (Hck), a Src family kinase, isa myeloid-specific nonreceptor tyrosine kinase involvedin neutrophil production, adhesion and cytokine pro-duction. Hck overexpression in mice induces chronicneutrophilic inflammation and emphysema (Ernst,2002). Hck is involved in neutrophil and myeloid cellrecruitment into the lung, because Hck knockout micefail to recruit these cells in response to TNF-a and LPS

Kinase Inhibitors in Airway Disease 807

Page 21: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

(Mazzi et al., 2015). There is increased expression of Hckin circulating neutrophils from patients with COPD(Yanagisawa et al., 2009), suggesting that this kinasemay be a good target for inhibition. Several drugs thatinhibit Hck, including PP2 and dasatinib, are available,and new drugs are in development (Poh et al., 2015).Inhaled drugs that have inhibitory effects onHck are alsoin clinical development for the treatment of severeasthma and COPD (Norman, 2014a; Onions et al., 2016).

E. Bruton’s Tyrosine Kinase

Bruton’s tyrosine kinase (BTK) pays a critical role inB-lymphocyte development and in mast cell activationthrough high-affinity IgE receptors (Crofford et al., 2016).Mutations of BTK are associated with X-linked agamma-globulinemia, which causes immunodeficiency. SelectiveBTK inhibitors, such as ibrutinib, have been developed fortreatment of B cell malignancies, but so far have not beentested in airway disease (Foluso et al., 2016).

XIII. Future Developments

Although many hundreds of kinases have beenimplicated in chronic inflammatory diseases, relativelyfew have been successfully targeted therapeutically.Major problems involve specificity and adverse effects,often due to off-target effects and loss of efficacy overtime. Although over 30 kinase inhibitors have beenapproved for the treatment of various forms of malig-nancy, relatively few are approved for the treatment of

chronic inflammatory diseases and none so far forasthma and COPD therapy (Wu et al., 2015c).

A. Improving Selectivity and Safety

Most kinase inhibitors target the ATP-bindingpocket, where they compete with ATP in high concen-trations, and so need to be of at least nanomolar potencyto be effective. Because ATP binding is common to allkinases, it is difficult to develop kinases with specificityfor a particular kinase because of the great similarity inbinding site between different kinases. The off-targeteffects often account for sometimes unexpected adverseeffects that have severely limited the clinical develop-ment of these drugs. Although this is relatively lessimportant for treatment of life-threatening malignan-cies, it is unacceptable in long-term treatment of chronicinflammatory diseases, such as asthma and COPD.Understanding the cocrystal structure of the ligand-ATP-binding site of particular kinases may aid thediscovery of more selective inhibitors (Norman et al.,2012b). For example, this approach has been used todevelop selective inhibitors of FGFR tyrosine kinase(Norman et al., 2012a). Targeting allosteric bindingsites that lie outside the highly conserved ATP-bindingsite offers hope for greater potency and specificity andmay also protect against the development of drugresistance by mutation of the ATP-binding site (Wuet al., 2015b). Binding may be to an allosteric site closeor distant from the ATP-binding pocket. Over 10 allo-steric kinase inhibitors are now in clinical development

TABLE 1Kinase inhibitors in clinical development

Kinase Inhibitors Development Stage Disease Indication Comments

IKK (NF-kB) IMD-0354 Phase 2? COPD DiscontinuedBay 65-1942 Preclinical COPD Oral

p38 MAPK Dilmapimod Phase 2 COPD OralLosmapimod Phase 2 COPD OralPH-797804 Phase 2 COPD OralPF-03715455 Phase 2 Asthma, COPD InhaledAZD7624 Phase 2 COPD InhaledRV-568 Phase 2 COPD Inhaled

JNK SP600125 Preclinical Asthma, COPD? OralMEK1/ERK Trametinib Phase 3 Cancer OralASK-1 GS-4997 Phase 2 Diabetes OralPI3K Idelisib Approved B cell malignancy Oral PI3Kd-selective

TG100-115 Preclinical Asthma, COPD InhaledPI3Kg/d-selectiveGSK2269557 Phase 2 COPD Inhaled PI3Kd-selectiveRV-1729 Phase 2 Asthma, COPD Inhaled PI3Kg/d-selective

JAK LAS194046 Preclinical Asthma, COPD InhaledR256 Preclinical Asthma, COPD InhaledVR560 Preclinical Asthma, COPD Inhaled

SYK Fostamatinib Phase 3 Arthritis, lymphoma OralR112 Phase 2 Rhinitis Nasal, discontinuedR343 Phase 2 Asthma Inhaled, discontinuedLAS189386 Preclinical Asthma Inhaled

RTK BIBW 2948 Phase 2 COPD Inhaled EGFRMasitinib Phase 2 Severe asthma Oral, PDGFR, c-Kit

ROCK Fasudil Approved Cerebral vasospasm Oral

ASK, apoptosis signal-regulatory kinase; EGFR, epidermal growth factor receptor; ERK, extracellular regulatingkinase; IKK, inhibitor of kB kinase; JAK, Janus-activated kinase; JNK, Jun NH2-terminal kinase; MAPK, mitogen-activated protein kinase; NF-kB, nuclear factor-kappa B; PDGFR, platelet-derived growth factor receptor; PI3K,phosphoinositide-3-kinase; ROCK, Rho-associated protein kinase; RTK, receptor tyrosine kinase; Syk, spleen tyrosinekinase.

808 Barnes

Page 22: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

and somemay be combinedwith conventional inhibitorsof the same kinase.Another way to limit side effects is to deliver kinase

inhibitors topically and, in the case of airway disease, byinhalation, which should greatly reduce systemic expo-sure. This requires the development of drugs with highpotency that have low oral bioavailability and topicalactivity. It has so far proven difficult to develop suchdrugs, but inhaled p38 MAPK inhibitors are currentlyin clinical development for severe asthma and COPD(Millan, 2011; Norman, 2015). Inhaled PI3Kg/d inhibi-tors are also in development for severe asthma andCOPD because they may overcome the corticosteroidsresistance in these diseases (Norman, 2014a; Onionset al., 2016).

B. Targeting Several Kinases

Inhibitingmore than one kinasemay increase efficacybecause theremay be additive or synergistic effects seenby inhibiting more than one kinase. In addition, it maydecrease the risks of developing resistance by blockingpathways that may reduce the effect of monokinaseinhibition (Morphy, 2010). Several multikinase inhibi-tors have been developed for the treatment of malig-nancies (Garuti et al., 2015). Sorafenib targets severaltyrosine kinases (VEGFR, PDGFR, c-Raf, c-Kit), suni-tinib (VEGFR, PDGFR), vandetanib (VEGFR, EGFR,RET), and pazopanib (VEGFR, PDGFR, c-Kit). Thesedrugs are indicated for advanced malignancies, andadverse effects are common. Nintedanib inhibits sev-eral growth factor receptors (VEGFR, PDGFR, FGFR)and is approved for the treatment of idiopathic pulmo-nary fibrosis but has significant side effects (Keating,2015). There is a case for the development of inhaledmultikinase inhibitors for severe airway disease. Sev-eral narrow spectrum kinase inhibitors suitable forinhaled delivery have been developed for the treat-ment of corticosteroid-resistant inflammation in severeasthma and COPD (Onions et al., 2016). These drugsinhibit p38a, p38g and Hck and are more effective thanp38a inhibitors in suppressing the release of inflamma-tory cytokines from macrophages in vitro and in treat-ing inflammation in mice exposed to cigarette smoke,which is resistant to corticosteroids.

C. Overcoming Kinase Inhibitor Resistance

A major limitation to the use of kinase inhibitors inthe treatment of malignancy is mutation of the ATPbinding site, which overcomes the inhibitory effects ofkinases. This is addressed by the development of multi-kinase inhibitors and allosteric kinase inhibitors, whichmake development of resistance less likely. Covalentkinase inhibitors are another approach, and irrevers-ible inhibitors are now in clinical trials for malignantdisease. Loss of effect of kinase incubators is also seen inthe treatment of chronic inflammatory disease, such asrheumatoid arthritis and inflammatory bowel disease,

and tends to develop over several weeks of treatment(Barouch-Bentov and Sauer, 2011). This may arise ascompensatory pathways are upregulated to circumventthe pathway blocked by the kinase inhibitor and may beovercome by also blocking the compensatory pathway,for example with narrow spectrum kinase inhibitors(Onions et al., 2016).

D. Conclusions

Although this review highlighted the involvement ofmany different kinases in both asthma and COPD, sofar no kinase inhibitors have been approved for clinicaluse and have not even yet reached Phase 3 studies.Table 1 summarizes the clinical status of many of thedrugs that are discussed in the review. Many preclinicalstudies and early clinical trials are underway, sohopefully the field will advance. A major challenge hasbeen the development of potent inhaled kinase inhibi-tors to avoid systemic exposure and the lack of suchcompounds has slowed development in this area. Inview of the pressing unmet need for new treatments insevere asthma and COPD, it is hoped that there will besignificant investment for the pharmaceutical industryin this area in the future.

ReferencesAbraham SM, Lawrence T, Kleiman A, Warden P, Medghalchi M, Tuckermann J,Saklatvala J, and Clark AR (2006) Antiinflammatory effects of dexamethasone arepartly dependent on induction of dual specificity phosphatase 1. J Exp Med 203:1883–1889.

Adcock IM, Chung KF, Caramori G, and Ito K (2006) Kinase inhibitors and airwayinflammation. Eur J Pharmacol 533:118–132.

Adcock IM, Lane SJ, Brown CR, Lee TH, and Barnes PJ (1995) Abnormal gluco-corticoid receptor-activator protein 1 interaction in steroid-resistant asthma. J ExpMed 182:1951–1958.

Ajibade AA, Wang HY, and Wang RF (2013) Cell type-specific function of TAK1 ininnate immune signaling. Trends Immunol 34:307–316.

Alexander W (2011) Inhibiting the akt pathway in cancer treatment: three leadingcandidates. P&T 36:225–227.

Ali K, Camps M, Pearce WP, Ji H, Rückle T, Kuehn N, Pasquali C, Chabert C,Rommel C, and Vanhaesebroeck B (2008) Isoform-specific functions of phosphoi-nositide 3-kinases: p110 d but not p110 g promotes optimal allergic responsesin vivo. J Immunol 180:2538–2544.

Ameriks MK and Venable JD (2009) Small molecule inhibitors of phosphoinositide3-kinase (PI3K) delta and gamma. Curr Top Med Chem 9:738–753.

Anderson DR, Meyers MJ, Kurumbail RG, Caspers N, Poda GI, Long SA, Pierce BS,Mahoney MW, and Mourey RJ (2009) Benzothiophene inhibitors of MK2. Part 1:structure-activity relationships, assessments of selectivity and cellular potency.Bioorg Med Chem Lett 19:4878–4881.

Arthur JS and Ley SC (2013) Mitogen-activated protein kinases in innate immunity.Nat Rev Immunol 13:679–692.

Aschner Y and Downey GP (2016) Transforming growth factor-b: Master regulator ofthe respiratory system in health and disease. Am J Respir Cell Mol Biol 54:647–655.

Ashino S, Takeda K, Li H, Taylor V, Joetham A, Pine PR, and Gelfand EW (2014)Janus kinase 1/3 signaling pathways are key initiators of TH2 differentiation andlung allergic responses. J Allergy Clin Immunol 133:1162–1174.

Backers K, Blero D, Paternotte N, Zhang J, and Erneux C (2003) The termination ofPI3K signalling by SHIP1 and SHIP2 inositol 5-phosphatases. Adv Enzyme Regul43:15–28.

Balzar S, Chu HW, Silkoff P, Cundall M, Trudeau JB, Strand M, and Wenzel S (2005)Increased TGF-beta2 in severe asthma with eosinophilia. J Allergy Clin Immunol115:110–117.

Barnes PJ (2008a) The cytokine network in asthma and chronic obstructive pulmo-nary disease. J Clin Invest 118:3546–3556.

Barnes PJ (2008b) Immunology of asthma and chronic obstructive pulmonary dis-ease. Nat Rev Immunol 8:183–192.

Barnes PJ (2011) Pathophysiology of allergic inflammation. Immunol Rev 242:31–50.Barnes PJ (2013a) Corticosteroid resistance in patients with asthma and chronicobstructive pulmonary disease. J Allergy Clin Immunol 131:636–645.

Barnes PJ (2013b) New anti-inflammatory targets for chronic obstructive pulmonarydisease. Nat Rev Drug Discov 12:543–559.

Barnes PJ (2013c) Theophylline. Am J Respir Crit Care Med 188:901–906.Barnes PJ (2015a) Mechanisms of development of multimorbidity in the elderly. EurRespir J 45:790–806.

Kinase Inhibitors in Airway Disease 809

Page 23: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

Barnes PJ (2015b) Therapeutic approaches to asthma-chronic obstructive pulmonarydisease overlap syndromes. J Allergy Clin Immunol 136:531–545.

Barnes PJ (2016) Asthma-COPD overlap. Chest 149:7–8.Barnes PJ and Adcock IM (2009) Glucocorticoid resistance in inflammatory diseases.Lancet 373:1905–1917.

Barnes PJ, Burney PGJ, Silverman EK, Celli BR, Vestbo J, Wedzicha JA,and Wouters EFM (2015) Chronic obstructive pulmonary disease. Nat Rev DisPrimers 1:15076.

Barnes PJ and Karin M (1997) Nuclear factor-kappaB: a pivotal transcription factorin chronic inflammatory diseases. N Engl J Med 336:1066–1071.

Barouch-Bentov R and Sauer K (2011) Mechanisms of drug resistance in kinases.Expert Opin Investig Drugs 20:153–208.

Bateman ED, Reddel HK, van Zyl-Smit RN, and Agusti A (2015) The asthma-COPDoverlap syndrome: towards a revised taxonomy of chronic airways diseases? LancetRespir Med 3:719–728.

Beasley R, Crane J, Lai CK, and Pearce N (2000) Prevalence and etiology of asthma.J Allergy Clin Immunol 105:S466–S472.

Bentley JK, Newcomb DC, Goldsmith AM, Jia Y, Sajjan US, and Hershenson MB(2007) Rhinovirus activates interleukin-8 expression via a Src/p110b phosphati-dylinositol 3-kinase/Akt pathway in human airway epithelial cells. J Virol 81:1186–1194.

Berlin AA and Lukacs NW (2005) Treatment of cockroach allergen asthma modelwith imatinib attenuates airway responses. Am J Respir Crit Care Med 171:35–39.

Bhavsar P, Hew M, Khorasani N, Torrego A, Barnes PJ, Adcock I, and Chung KF(2008) Relative corticosteroid insensitivity of alveolar macrophages in severeasthma compared with non-severe asthma. Thorax 63:784–790.

Birrell MA, Wong S, Hardaker EL, Catley MC, McCluskie K, Collins M, Haj-Yahia S,and Belvisi MG (2006) IkappaB kinase-2-independent and -dependent in-flammation in airway disease models: relevance of IKK-2 inhibition to the clinic.Mol Pharmacol 69:1791–1800.

Bossé Y and Rola-Pleszczynski M (2008) FGF2 in asthmatic airway-smooth-muscle-cell hyperplasia. Trends Mol Med 14:3–11.

Braido F, Brusselle G, Ingrassia E, Nicolini G, Price D, Roche N, Soriano JB,and Worth H; LIAISON study group (2013) InternationaL cross-sectIonAl andlongItudinal assessment on aSthma cONtrol in European adult patients–theLIAISON study protocol. BMC Pulm Med 13:18.

Brook M, Tchen CR, Santalucia T, McIlrath J, Arthur JS, Saklatvala J, and Clark AR(2006) Posttranslational regulation of tristetraprolin subcellular localization andprotein stability by p38 mitogen-activated protein kinase and extracellular signal-regulated kinase pathways. Mol Cell Biol 26:2408–2418.

Brown JR (2015) Idelalisib has CLL on the run! Blood 126:2656–2657.Caramori G, Oates T, Nicholson AG, Casolari P, Ito K, Barnes PJ, Papi A, Adcock IM,and Chung KF (2009) Activation of NF-kappaB transcription factor in asthmadeath. Histopathology 54:507–509.

Caramori G, Romagnoli M, Casolari P, Bellettato C, Casoni G, Boschetto P, ChungKF, Barnes PJ, Adcock IM, and Ciaccia A, et al. (2003) Nuclear localisation of p65in sputum macrophages but not in sputum neutrophils during COPD exacerba-tions. Thorax 58:348–351.

Carlin S, Poronnik P, Cook DI, Carpenter L, Biden TJ, Johnson PR, and Black JL(2000) An antisense of protein kinase C-zeta inhibits proliferation of human airwaysmooth muscle cells. Am J Respir Cell Mol Biol 23:555–559.

Carracedo A and Pandolfi PP (2008) The PTEN-PI3K pathway: of feedbacks andcross-talks. Oncogene 27:5527–5541.

Catley MC, Sukkar MB, Chung KF, Jaffee B, Liao SM, Coyle AJ, Haddad B, BarnesPJ, and Newton R (2006) Validation of the anti-inflammatory properties of small-molecule IkappaB Kinase (IKK)-2 inhibitors by comparison with adenoviral-mediated delivery of dominant-negative IKK1 and IKK2 in human airways smoothmuscle. Mol Pharmacol 70:697–705.

Chand S, Mehta N, Bahia MS, Dixit A, and Silakari O (2012) Protein kinase C-thetainhibitors: a novel therapy for inflammatory disorders. Curr Pharm Des 18:4725–4746.

Chaudhary D, Robinson S, and Romero DL (2015) Recent advances in the discovery ofsmall molecule inhibitors of interleukin-1 receptor-associated kinase 4 (IRAK4) asa therapeutic target for inflammation and oncology disorders. J Med Chem 58:96–110.

Chiarini F, Evangelisti C, McCubrey JA, and Martelli AM (2015) Current treatmentstrategies for inhibiting mTOR in cancer. Trends Pharmacol Sci 36:124–135.

Cho SH, Woo CH, Yoon SB, and Kim JH (2004a) Protein kinase Cdelta functionsdownstream of Ca2+ mobilization in FcepsilonRI signaling to degranulation inmast cells. J Allergy Clin Immunol 114:1085–1092.

Cho SH, You HJ, Woo CH, Yoo YJ, and Kim JH (2004b) Rac and protein kinaseC-delta regulate ERKs and cytosolic phospholipase A2 in FcepsilonRI signaling tocysteinyl leukotriene synthesis in mast cells. J Immunol 173:624–631.

Choi YH, Jin GY, Li LC, and Yan GH (2013) Inhibition of protein kinase C deltaattenuates allergic airway inflammation through suppression of PI3K/Akt/mTOR/HIF-1 alpha/VEGF pathway. PLoS One 8:e81773.

Chung KF (2011) p38 mitogen-activated protein kinase pathways in asthma andCOPD. Chest 139:1470–1479.

Chung S, Sundar IK, Hwang JW, Yull FE, Blackwell TS, Kinnula VL, Bulger M, YaoH, and Rahman I (2011) NF-kB inducing kinase, NIK mediates cigarette smoke/TNFa-induced histone acetylation and inflammation through differential activa-tion of IKKs. PLoS One 6:e23488.

Chung KF and Wenzel S; European Respiratory Society/American Thoracic SocietySevere Asthma International Guidelines Task Force (2014) From the authors:International European Respiratory Society/American Thoracic Society guidelineson severe asthma. Eur Respir J 44:1378–1379.

Cohen P (2002) Protein kinases–the major drug targets of the twenty-first century?Nat Rev Drug Discov 1:309–315.

Cohen P (2014) Immune diseases caused by mutations in kinases and components ofthe ubiquitin system. Nat Immunol 15:521–529.

Comess KM, Sun C, Abad-Zapatero C, Goedken ER, Gum RJ, Borhani DW, ArgiriadiM, Groebe DR, Jia Y, and Clampit JE, et al. (2011) Discovery and characterizationof non-ATP site inhibitors of the mitogen activated protein (MAP) kinases. ACSChem Biol 6:234–244.

Condliffe AM, Davidson K, Anderson KE, Ellson CD, Crabbe T, Okkenhaug K,Vanhaesebroeck B, Turner M, Webb L, and Wymann MP, et al. (2005) Sequentialactivation of class IB and class IA PI3K is important for the primed respiratoryburst of human but not murine neutrophils. Blood 106:1432–1440.

Cosio BG, Tsaprouni L, Ito K, Jazrawi E, Adcock IM, and Barnes PJ (2004) Theo-phylline restores histone deacetylase activity and steroid responses in COPDmacrophages. J Exp Med 200:689–695.

Costa C, Martin-Conte EL, and Hirsch E (2011) Phosphoinositide 3-kinase p110g inimmunity. IUBMB Life 63:707–713.

Costa C, Rufino R, Traves SL, Lapa E Silva JR, Barnes PJ, and Donnelly LE (2008)CXCR3 and CCR5 chemokines in induced sputum from patients with COPD. Chest133:26–33.

Crofford LJ, Nyhoff LE, Sheehan JH, and Kendall PL (2016) The role of Bruton’styrosine kinase in autoimmunity and implications for therapy. Expert Rev ClinImmunol 4:1–11.

DaCosta Byfield S, Major C, Laping NJ, and Roberts AB (2004) SB-505124 is aselective inhibitor of transforming growth factor-b type I receptors ALK4, ALK5,and ALK7. Mol Pharmacol 65:744–752.

Dallas SL, Rosser JL, Mundy GR, and Bonewald LF (2002) Proteolysis of latenttransforming growth factor-b (TGF-b )-binding protein-1 by osteoclasts. A cellularmechanism for release of TGF-b from bone matrix. J Biol Chem 277:21352–21360.

Dean JL, Sully G, Clark AR, and Saklatvala J (2004) The involvement of AU-richelement-binding proteins in p38 mitogen-activated protein kinase pathway-mediated mRNA stabilisation. Cell Signal 16:1113–1121.

de Boer WI, Hau CM, van Schadewijk A, Stolk J, van Krieken JH, and Hiemstra PS(2006) Expression of epidermal growth factors and their receptors in the bronchialepithelium of subjects with chronic obstructive pulmonary disease. Am J ClinPathol 125:184–192.

de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk J, Hiemstra PS, and vanKrieken JH (1998) Transforming growth factor beta1 and recruitment of macro-phages and mast cells in airways in chronic obstructive pulmonary disease. Am JRespir Crit Care Med 158:1951–1957.

Dempsey EC, Cool CD, and Littler CM (2007) Lung disease and PKCs. PharmacolRes 55:545–559.

Desmet C, Gosset P, Pajak B, Cataldo D, Bentires-Alj M, Lekeux P, and Bureau F(2004) Selective blockade of NF-kappa B activity in airway immune cells inhibitsthe effector phase of experimental asthma. J Immunol 173:5766–5775.

Devereux G, Cotton S, Barnes P, Briggs A, Burns G, Chaudhuri R, Chrystyn H,Davies L, De Soyza A, and Fielding S, et al. (2015) Use of low-dose oral theoph-ylline as an adjunct to inhaled corticosteroids in preventing exacerbations ofchronic obstructive pulmonary disease: study protocol for a randomised controlledtrial. Trials 16:267.

Dienstmann R, Rodon J, Serra V, and Tabernero J (2014) Picking the point of in-hibition: a comparative review of PI3K/AKT/mTOR pathway inhibitors. Mol Can-cer Ther 13:1021–1031.Di Stefano A, Caramori G, Capelli A, Gnemmi I, RicciardoloFL, Oates T, Donner CF, Chung KF, Barnes PJ, and Adcock IM (2004) STAT4activation in smokers and patients with chronic obstructive pulmonary disease.Eur Respir J 24:78–85.

Di Stefano A, Caramori G, Oates T, Capelli A, Lusuardi M, Gnemmi I, Ioli F, ChungKF, Donner CF, and Barnes PJ, et al. (2002) Increased expression of nuclear factor-kappaB in bronchial biopsies from smokers and patients with COPD. Eur Respir J20:556–563.

Dong T, Li C, Wang X, Dian L, Zhang X, Li L, Chen S, Cao R, Li L, and Huang N,et al. (2015) Ainsliadimer A selectively inhibits IKKa/b by covalently binding aconserved cysteine. Nat Commun 6:6522.

Donnelly LE, Newton R, Kennedy GE, Fenwick PS, Leung RH, Ito K, Russell RE,and Barnes PJ (2004) Anti-inflammatory effects of resveratrol in lung epithelialcells: molecular mechanisms. Am J Physiol Lung Cell Mol Physiol 287:L774–L783.

Donnelly R, Yang K, Omary MB, Azhar S, and Black JL (1995) Expression of multipleisoenzymes of protein kinase C in airway smooth muscle. Am J Respir Cell Mol Biol13:253–256.

Doukas J, Eide L, Stebbins K, Racanelli-Layton A, Dellamary L, Martin M,Dneprovskaia E, Noronha G, Soll R, and Wrasidlo W, et al. (2009) Aerosolizedphosphoinositide 3-kinase gamma/delta inhibitor TG100-115 [3-[2,4-diamino-6-(3-hydroxyphenyl)pteridin-7-yl]phenol] as a therapeutic candidate for asthma andchronic obstructive pulmonary disease. J Pharmacol Exp Ther 328:758–765.

Duan W, Aguinaldo Datiles AM, Leung BP, Vlahos CJ, and Wong WS (2005a) Ananti-inflammatory role for a phosphoinositide 3-kinase inhibitor LY294002 in amouse asthma model. Int Immunopharmacol 5:495–502.

Duan W, Chan JH, McKay K, Crosby JR, Choo HH, Leung BP, Karras JG, and WongWS (2005b) Inhaled p38alpha mitogen-activated protein kinase antisense oligo-nucleotide attenuates asthma in mice. Am J Respir Crit Care Med 171:571–578.

Ernst M, Inglese M, Scholz GM, Harder KW, Clay FJ, and Bozinovski S, et al. (2002)Constitutive activation of the SRC family kinase Hck results in spontaneous pul-monary inflammation and an enhanced innate immune response. J Exp Med 196:589–604.

Evans DJ, Lindsay MA, Webb BL, Kankaanranta H, Giembycz MA, O’Connor BJ,and Barnes PJ (1999) Expression and activation of protein kinase C-zeta in eo-sinophils after allergen challenge. Am J Physiol 277:L233–L239.

Eynott PR, Nath P, Leung SY, Adcock IM, Bennett BL, and Chung KF (2003)Allergen-induced inflammation and airway epithelial and smooth muscle cellproliferation: role of Jun N-terminal kinase. Br J Pharmacol 140:1373–1380.

Fabbro D, Cowan-Jacob SW, and Moebitz H (2015) Ten things you shouldknow about protein kinases: IUPHAR Review 14. Br J Pharmacol 172:2675–2700.

810 Barnes

Page 24: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

Feltis BN, Wignarajah D, Zheng L, Ward C, Reid D, Harding R, and Walters EH(2006) Increased vascular endothelial growth factor and receptors: relationship toangiogenesis in asthma. Am J Respir Crit Care Med 173:1201–1207.

Fenwick PS, Macedo P, Kilty IC, Barnes PJ, and Donnelly LE (2015) Effect of JAKinhibitors on release of CXCL9, CXCL10 and CXCL11 from human airway epi-thelial cells. PLoS One 10:e0128757.

Fenwick P, Rao V, Kilty I, Lee K, Barnes PJ, and Donnelly D (2013) Inhibition ofIRAK4 suppresses chemokine release from human bronchial epithelial cells. EurRespir J 44:1033s

Fernandes LB, Henry PJ, and Goldie RG (2007) Rho kinase as a therapeutic target inthe treatment of asthma and chronic obstructive pulmonary disease. Ther AdvRespir Dis 1:25–33.

Fiore M, Forli S, and Manetti F (2016) Targeting mitogen-activated protein kinase-activated protein kinase 2 (MAPKAPK2, MK2): medicinal chemistry efforts to leadsmall molecule inhibitors to clinical trials. J Med Chem 59:3609–3634.

Firestein R, Blander G, Michan S, Oberdoerffer P, Ogino S, Campbell J,Bhimavarapu A, Luikenhuis S, de Cabo R, and Fuchs C, et al. (2008) The SIRT1 deace-tylase suppresses intestinal tumorigenesis and colon cancer growth. PLoS One 3:e2020.

Foluso O, Glick A, Stender M, and Jaiyesimi I (2016) Ibrutinib as a bruton kinaseinhibitor in the management of chronic lymphocytic leukemia: a new agent withgreat promise. Clin Lymphoma Myeloma Leuk 16:63–69.

Footitt J, Mallia P, Durham AL, Ho WE, Trujillo-Torralbo MB, Telcian AG, DelRosario A, Chang C, Peh HY, and Kebadze T, et al. (2016) Oxidative and nitro-sative stress and histone deacetylase-2 activity in exacerbations of COPD. Chest149:62–73.

Frémin C and Meloche S (2010) From basic research to clinical development ofMEK1/2 inhibitors for cancer therapy. J Hematol Oncol 3:8.

Gaestel M (2013) What goes up must come down: molecular basis of MAPKAP kinase2/3-dependent regulation of the inflammatory response and its inhibition. BiolChem 394:1301–1315.

Gaffey K, Reynolds S, Plumb J, Kaur M, and Singh D (2013) Increased phosphory-lated p38 mitogen-activated protein kinase in COPD lungs. Eur Respir J 42:28–41.

Galli SJ and Tsai M (2012) IgE and mast cells in allergic disease. Nat Med 18:693–704.

Garuti L, Roberti M, and Bottegoni G (2015) Multi-kinase inhibitors. Curr Med Chem22:695–712.

Geahlen RL (2014) Getting Syk: spleen tyrosine kinase as a therapeutic target.Trends Pharmacol Sci 35:414–422.

Ghoreschi K, Laurence A, and O’Shea JJ (2009) Janus kinases in immune cell sig-naling. Immunol Rev 228:273–287.

Goldsmith AM, Bentley JK, Zhou L, Jia Y, Bitar KN, Fingar DC, and Hershenson MB(2006) Transforming growth factor-beta induces airway smooth muscle hypertro-phy. Am J Respir Cell Mol Biol 34:247–254.

Göransson O, McBride A, Hawley SA, Ross FA, Shpiro N, Foretz M, Viollet B, HardieDG, and Sakamoto K (2007) Mechanism of action of A-769662, a valuable tool foractivation of AMP-activated protein kinase. J Biol Chem 282:32549–32560.

Gorska MM, Liang Q, Stafford SJ, Goplen N, Dharajiya N, Guo L, Sur S, Gaestel M,and Alam R (2007) MK2 controls the level of negative feedback in the NF-kappaBpathway and is essential for vascular permeability and airway inflammation. JExp Med 204:1637–1652.

Gosens R, Schaafsma D, Meurs H, Zaagsma J, and Nelemans SA (2004) Role of Rho-kinase in maintaining airway smooth muscle contractile phenotype. Eur J Phar-macol 483:71–78.

Grandordy BM, Mak JCW, and Barnes PJ (1994) Modulation of airway smoothmuscle b-adrenoceptor function by a muscarinic agonist. Life Sci 54:185–191.

Griego SD, Weston CB, Adams JL, Tal-Singer R, and Dillon SB (2000) Role of p38mitogen-activated protein kinase in rhinovirus-induced cytokine production bybronchial epithelial cells. J Immunol 165:5211–5220.

Guyer BJ, Shimamoto SR, Bradhurst AL, Grossbard EB, Dreskin SC, and Nelson HS(2006) Mast cell inhibitor R112 is well tolerated and affects prostaglandin D2 butnot other mediators, symptoms, or nasal volumes in a nasal challenge model ofallergic rhinitis. Allergy Asthma Proc 27:208–213.

Halayko AJ, Kartha S, Stelmack GL, McConville J, Tam J, Camoretti-Mercado B,Forsythe SM, Hershenson MB, and Solway J (2004) Phophatidylinositol-3 kinase/mammalian target of rapamycin/p70S6K regulates contractile protein accumula-tion in airway myocyte differentiation. Am J Respir Cell Mol Biol 31:266–275.

Hall DJ, Bates ME, Guar L, CronanM, Korpi N, and Bertics PJ (2005) The role of p38MAPK in rhinovirus-induced monocyte chemoattractant protein-1 production bymonocytic-lineage cells. J Immunol 174:8056–8063.

Halwani R, Al-Muhsen S, Al-Jahdali H, and Hamid Q (2011) Role of transforminggrowth factor-b in airway remodeling in asthma. Am J Respir Cell Mol Biol 44:127–133.

Hamilton LM, Torres-Lozano C, Puddicombe SM, Richter A, Kimber I, Dearman RJ,Vrugt B, Aalbers R, Holgate ST, and Djukanovi�c R, et al. (2003) The role of theepidermal growth factor receptor in sustaining neutrophil inflammation in severeasthma. Clin Exp Allergy 33:233–240.

Hardyman MA, Wilkinson E, Martin E, Jayasekera NP, Blume C, Swindle EJ,Gozzard N, Holgate ST, Howarth PH, and Davies DE, et al. (2013) TNF-a-medi-ated bronchial barrier disruption and regulation by src-family kinase activation. JAllergy Clin Immunol 132:665–675.

Hart L, Lim S, Adcock I, Barnes PJ, and Chung KF (2000) Effects of inhaled corti-costeroid therapy on expression and DNA-binding activity of nuclear factor kappaBin asthma. Am J Respir Crit Care Med 161:224–231.

Hart LA, Krishnan VL, Adcock IM, Barnes PJ, and Chung KF (1998) Activation andlocalization of transcription factor, nuclear factor-kappaB, in asthma. Am J RespirCrit Care Med 158:1585–1592.

Hatcher H, Planalp R, Cho J, Torti FM, and Torti SV (2008) Curcumin: from ancientmedicine to current clinical trials. Cell Mol Life Sci 65:1631–1652.

Hattori K, Naguro I, Runchel C, and Ichijo H (2009) The roles of ASK family proteinsin stress responses and diseases. Cell Commun Signal 7:9.

Hawkins PT and Stephens LR (2015) PI3K signalling in inflammation. BiochimBiophys Acta 1851:882–897.

Hayden MS and Ghosh S (2012) NF-kB, the first quarter-century: remarkableprogress and outstanding questions. Genes Dev 26:203–234.

Hegab AE, Sakamoto T, Nomura A, Ishii Y, Morishima Y, Iizuka T, Kiwamoto T,Matsuno Y, Homma S, and Sekizawa K (2007) Niflumic acid and AG-1478 reducecigarette smoke-induced mucin synthesis: the role of hCLCA1. Chest 131:1149–1156.

Hekking PP, Wener RR, Amelink M, Zwinderman AH, Bouvy ML, and Bel EH (2015)The prevalence of severe refractory asthma. J Allergy Clin Immunol 135:896–902.

Henry PJ, Mann TS, and Goldie RG (2005) A rho kinase inhibitor, Y-27632 inhibitspulmonary eosinophilia, bronchoconstriction and airways hyperresponsiveness inallergic mice. Pulm Pharmacol Ther 18:67–74.

Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST,Cleverly AL, Desaiah D, Guba SC, and Benhadji KA, et al. (2015) Clinical de-velopment of galunisertib (LY2157299 monohydrate), a small molecule in-hibitor of transforming growth factor-beta signaling pathway. Drug Des DevelTher 9:4479–4499.

Hirota JA, Ask K, Farkas L, Smith JA, Ellis R, Rodriguez-Lecompte JC, Kolb M,and Inman MD (2011) In vivo role of platelet-derived growth factor-BB in airwaysmooth muscle proliferation in mouse lung. Am J Respir Cell Mol Biol 45:566–572.

Hirst SJ, Barnes PJ, and Twort CHC (1996) PDGF isoform-induced proliferation andreceptor expression in human cultured airway smooth muscle cells. Am J Physiol270:L415–L428.

Hoshino M, Takahashi M, and Aoike N (2001) Expression of vascular endothelialgrowth factor, basic fibroblast growth factor, and angiogenin immunoreactivity inasthmatic airways and its relationship to angiogenesis. J Allergy Clin Immunol107:295–301.

Hua W, Liu H, Xia LX, Tian BP, Huang HQ, Chen ZY, Ju ZY, Li W, Chen ZH,and Shen HH (2015) Rapamycin inhibition of eosinophil differentiation attenuatesallergic airway inflammation in mice. Respirology 20:1055–1065.

Hubbard SR and Till JH (2000) Protein tyrosine kinase structure and function. AnnuRev Biochem 69:373–398.

Humbert M, de Blay F, Garcia G, Prud’homme A, Leroyer C, Magnan A, Tunon-de-Lara JM, Pison C, Aubier M, and Charpin D, et al. (2009) Masitinib, a c-kit/PDGF receptor tyrosine kinase inhibitor, improves disease control in severecorticosteroid-dependent asthmatics. Allergy 64:1194–1201.

Ichiyama T, Hasegawa S, Matsubara T, Hayashi T, and Furukawa S (2001) The-ophylline inhibits NF-k B activation and I k B a degradation in human pulmonaryepithelial cells. Naunyn Schmiedebergs Arch Pharmacol 364:558–561.

Ihn H (2002) Pathogenesis of fibrosis: role of TGF-b and CTGF. Curr Opin Rheumatol14:681–685.

Irusen E, Matthews JG, Takahashi A, Barnes PJ, Chung KF, and Adcock IM (2002)p38 Mitogen-activated protein kinase-induced glucocorticoid receptor phosphory-lation reduces its activity: role in steroid-insensitive asthma. J Allergy ClinImmunol 109:649–657.

Ito K, Caramori G, and Adcock IM (2007) Therapeutic potential of phosphatidyli-nositol 3-kinase inhibitors in inflammatory respiratory disease. J Pharmacol ExpTher 321:1–8.

Ito K, Lim S, Caramori G, Cosio B, Chung KF, Adcock IM, and Barnes PJ (2002) Amolecular mechanism of action of theophylline: Induction of histone deacetylaseactivity to decrease inflammatory gene expression. Proc Natl Acad Sci USA 99:8921–8926.

Jabara HH and Geha RS (2005) Jun N-terminal kinase is essential for CD40-mediated IgE class switching in B cells. J Allergy Clin Immunol 115:856–863.

Joh EH and Kim DH (2010) Lancemaside A inhibits lipopolysaccharide-induced in-flammation by targeting LPS/TLR4 complex. J Cell Biochem 111:865–871.

Johnson GL and Lapadat R (2002) Mitogen-activated protein kinase pathways me-diated by ERK, JNK, and p38 protein kinases. Science 298:1911–1912.

Johnson SC, Rabinovitch PS, and Kaeberlein M (2013) mTOR is a key modulator ofageing and age-related disease. Nature 493:338–345.

Jun JI and Lau LF (2011) Taking aim at the extracellular matrix: CCN proteins asemerging therapeutic targets. Nat Rev Drug Discov 10:945–963.

Kanazawa HTY, Asai K, and Hirata K (2014) Simultaneous assessment of hepatocytegrowth factor and vascular endothelial growth factor in epithelial lining fluid frompatients with COPD. Chest 146:1159–1165.

Kanazawa H, Asai K, Hirata K, and Yoshikawa J (2003) Possible effects of vascularendothelial growth factor in the pathogenesis of chronic obstructive pulmonarydisease. Am J Med 114:354–358.

Kankaanranta H, De Souza PM, Barnes PJ, Salmon M, Giembycz MA, and LindsayMA (1999) SB 203580, an inhibitor of p38 mitogen-activated protein kinase, en-hances constitutive apoptosis of cytokine-deprived human eosinophils. J Phar-macol Exp Ther 290:621–628.

Kankaanranta H, Ilmarinen P, Zhang X, Adcock IM, Lahti A, Barnes PJ, GiembyczMA, Lindsay MA, and Moilanen E (2014) Tumour necrosis factor-a regulates hu-man eosinophil apoptosis via ligation of TNF-receptor 1 and balance betweenNF-kB and AP-1. PLoS One 9:e90298.

Kasahara DI, Mathews JA, Park CY, Cho Y, Hunt G, Wurmbrand AP, Liao JK,and Shore SA (2015) ROCK insufficiency attenuates ozone-induced airwayhyperresponsiveness in mice. Am J Physiol Lung Cell Mol Physiol 309:L736–L746.

Keating GM (2015) Nintedanib: A review of its use in patients with idiopathic pul-monary fibrosis. Drugs 75:1131–1140.

Khorasani N, Baker J, Johnson M, Chung KF, and Bhavsar PK (2015) Reversal ofcorticosteroid insensitivity by p38 MAPK inhibition in peripheral blood mono-nuclear cells from COPD. Int J Chron Obstruct Pulmon Dis 10:283–291.

Kilty I and Jones LH (2015) TAK1 selective inhibition: state of the art and futureopportunities. Future Med Chem 7:23–33.

Kim DH, Jung WS, Kim ME, Lee HW, Youn HY, Seon JK, Lee HN, and Lee JS (2014)Genistein inhibits pro‑inflammatory cytokines in human mast cell activationthrough the inhibition of the ERK pathway. Int J Mol Med 34:1669–1674.

Kinase Inhibitors in Airway Disease 811

Page 25: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

Kim BS, Kim IK, Park YJ, Kim YS, Kim YJ, Chang WS, Lee YS, Kweon MN, ChungY, and Kang CY (2010) Conversion of Th2 memory cells into Foxp3+ regulatoryT cells suppressing Th2-mediated allergic asthma. Proc Natl Acad Sci USA 107:8742–8747.

Kim MS, Rådinger M, and Gilfillan AM (2008) The multiple roles of phosphoinositide3-kinase in mast cell biology. Trends Immunol 29:493–501.

Knipe RS, Tager AM, and Liao JK (2015) The Rho kinases: critical mediators ofmultiple profibrotic processes and rational targets for new therapies for pulmonaryfibrosis. Pharmacol Rev 67:103–117.

Kobayashi Y, Mercado N, Miller-Larsson A, Barnes PJ, and Ito K (2012) Increasedcorticosteroid sensitivity by a long acting b2 agonist formoterol via b2 adrenoceptorindependent protein phosphatase 2A activation. Pulm Pharmacol Ther 25:201–207.

Kobayashi Y, Wada H, Rossios C, Takagi D, Charron C, Barnes PJ, and Ito K (2013)A novel macrolide/fluoroketolide, solithromycin (CEM-101), reverses corticosteroidinsensitivity via phosphoinositide 3-kinase pathway inhibition. Br J Pharmacol169:1024–1034.

Koch A, Giembycz M, Ito K, Lim S, Jazrawi E, Barnes PJ, Adcock I, Erdmann E,and Chung KF (2004) Mitogen-activated protein kinase modulation of nuclearfactor-kappaB-induced granulocyte macrophage-colony-stimulating factor re-lease from human alveolar macrophages. Am J Respir Cell Mol Biol 30:342–349.

Koh RY, Lim CL, Uhal BD, Abdullah M, Vidyadaran S, Ho CC, and Seow HF (2015)Inhibition of transforming growth factor-b via the activin receptor-like kinase-5inhibitor attenuates pulmonary fibrosis. Mol Med Rep 11:3808–3813.

Kokturk N, Tatlicioglu T, Memis L, Akyurek N, and Akyol G (2003) Expression oftransforming growth factor beta1 in bronchial biopsies in asthma and COPD. JAsthma 40:887–893.

Koli K, Myllärniemi M, Keski-Oja J, and Kinnula VL (2008) Transforming growthfactor-beta activation in the lung: focus on fibrosis and reactive oxygen species.Antioxid Redox Signal 10:333–342.

Kono Y, Nishiuma T, Okada T, Kobayashi K, Funada Y, Kotani Y, Jahangeer S,Nakamura S, and Nishimura Y (2010) Sphingosine kinase 1 regulates mucinproduction via ERK phosphorylation. Pulm Pharmacol Ther 23:36–42.

Kontzias A, Kotlyar A, Laurence A, Changelian P, and O’Shea JJ (2012) Jakinibs: anew class of kinase inhibitors in cancer and autoimmune disease. Curr OpinPharmacol 12:464–470.

Kudlacz E, Conklyn M, Andresen C, Whitney-Pickett C, and Changelian P (2008) TheJAK-3 inhibitor CP-690550 is a potent anti-inflammatory agent in a murine modelof pulmonary eosinophilia. Eur J Pharmacol 582:154–161.

Lamming DW, Ye L, Sabatini DM, and Baur JA (2013) Rapalogs and mTOR inhib-itors as anti-aging therapeutics. J Clin Invest 123:980–989.

Lane SJ, Adcock IM, Richards D, Hawrylowicz C, Barnes PJ, and Lee TH (1998)Corticosteroid-resistant bronchial asthma is associated with increased c-fos ex-pression in monocytes and T lymphocytes. J Clin Invest 102:2156–2164.

Langlois A, Chouinard F, Flamand N, Ferland C, Rola-Pleszczynski M,and Laviolette M (2009) Crucial implication of protein kinase C (PKC)-delta, PKC-zeta, ERK-1/2, and p38 MAPK in migration of human asthmatic eosinophils. JLeukoc Biol 85:656–663.

Laping NJ, Grygielko E, Mathur A, Butter S, Bomberger J, Tweed C, Martin W,Fornwald J, Lehr R, and Harling J, et al. (2002) Inhibition of transforming growthfactor (TGF)-beta1-induced extracellular matrix with a novel inhibitor of the TGF-beta type I receptor kinase activity: SB-431542. Mol Pharmacol 62:58–64.

Lau C, Castellanos P, Ranev D, Wang X, and Chow CW (2011) HRV signaling inairway epithelial cells is regulated by ITAM-mediated recruitment and activationof Syk. Protein Pept Lett 18:518–529.

Lau C, Wang X, Song L, North M, Wiehler S, Proud D, and Chow CW (2008) Sykassociates with clathrin and mediates phosphatidylinositol 3-kinase activationduring human rhinovirus internalization. J Immunol 180:870–880.

Leaker BR, Barnes PJ, O’Connor BJ, Ali FY, Tam P, Neville J, Mackenzie LF,and MacRury T (2014) The effects of the novel SHIP1 activator AQX-1125 onallergen-induced responses in mild-to-moderate asthma. Clin Exp Allergy 44:1146–1153.

Lee YC, Chuang CY, Lee PK, Lee JS, Harper RW, Buckpitt AB, Wu R, and Oslund K(2008) TRX-ASK1-JNK signaling regulation of cell density-dependent cytotoxicityin cigarette smoke-exposed human bronchial epithelial cells. Am J Physiol LungCell Mol Physiol 294:L921–L931.

Lee KY, Ito K, Hayashi R, Jazrawi EP, Barnes PJ, and Adcock IM (2006b) NF-kappaB and activator protein 1 response elements and the role of histone modifi-cations in IL-1b-induced TGF-b1 gene transcription. J Immunol 176:603–615.

Lee KS, Jeong JS, Kim SR, Cho SH, Kolliputi N, Ko YH, Lee KB, Park SC, Park HJ,and Lee YC (2016) Phosphoinositide 3-kinase-d regulates fungus-induced allergiclung inflammation through endoplasmic reticulum stress. Thorax 71:52–63.

Lee KS, Lee HK, Hayflick JS, Lee YC, and Puri KD (2006a) Inhibition of phosphoi-nositide 3-kinase delta attenuates allergic airway inflammation and hyper-responsiveness in murine asthma model. FASEB J 20:455–465.

Lee JS, Park SJ, Cho YS, Huh JW, Oh YM, and Lee SD (2015) Role of AMP-activatedprotein kinase (AMPK) in smoking-Induced lung Inflammation and emphysema.Tuberc Respir Dis (Seoul) 78:8–17.

Lemmon MA and Schlessinger J (2010) Cell signaling by receptor tyrosine kinases.Cell 141:1117–1134.

Lennartsson J and Rönnstrand L (2012) Stem cell factor receptor/c-Kit: from basicscience to clinical implications. Physiol Rev 92:1619–1649.

Leung SY, Niimi A, Noble A, Oates T, Williams AS, Medicherla S, Protter AA,and Chung KF (2006) Effect of transforming growth factor-beta receptor I kinaseinhibitor 2,4-disubstituted pteridine (SD-208) in chronic allergic airway in-flammation and remodeling. J Pharmacol Exp Ther 319:586–594.

Levings MK, Bacchetta R, Schulz U, and Roncarolo MG (2002) The role of IL-10 andTGF-beta in the differentiation and effector function of T regulatory cells. Int ArchAllergy Immunol 129:263–276.

Li LB, Leung DY, and Goleva E (2015) Activated p38 MAPK in peripheral bloodmonocytes of steroid resistant asthmatics. PLoS One 10:e0141909.

Li N, Li Q, Zhou XD, Kolosov VP, and Perelman JM (2012) Chronic mechanical stressinduces mucin 5AC expression in human bronchial epithelial cells through ERKdependent pathways. Mol Biol Rep 39:1019–1028.

Lin JH, Zhang JJ, Lin SL, and Chertow GM (2015) Design of a phase 2 clinical trial ofan ASK1 inhibitor, GS-4997, in patients with diabetic kidney disease. Nephron129:29–33.

Lomas DA, Lipson DA, Miller BE, Willits L, Keene O, Barnacle H, Barnes NC,and Tal-Singer R; Losmapimod Study Investigators (2012) An oral inhibitor of p38MAP kinase reduces plasma fibrinogen in patients with chronic obstructive pul-monary disease. J Clin Pharmacol 52:416–424.

Lombardi V, Singh AK, and Akbari O (2010) The role of costimulatory molecules inallergic disease and asthma. Int Arch Allergy Immunol 151:179–189.

Louie CY, DiMaio MA, Matsukuma KE, Coutre SE, Berry GJ, and Longacre TA(2015) Idelalisib-associated enterocolitis: clinicopathologic features and distinctionfrom other enterocolitides. Am J Surg Pathol 39:1653–1660.

Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, AdairT, Aggarwal R, and Ahn SY, et al. (2012) Global and regional mortality from235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis forthe Global Burden of Disease Study 2010. Lancet 380:2095–2128.

MacNee W, Allan RJ, Jones I, De Salvo MC, and Tan LF (2013) Efficacy and safety ofthe oral p38 inhibitor PH-797804 in chronic obstructive pulmonary disease: arandomised clinical trial. Thorax 68:738–745.

Maneechotesuwan K, Xin Y, Ito K, Jazrawi E, Lee KY, Usmani OS, Barnes PJ,and Adcock IM (2007) Regulation of Th2 cytokine genes by p38 MAPK-mediatedphosphorylation of GATA-3. J Immunol 178:2491–2498.

Maneechotesuwan K, Yao X, Ito K, Jazrawi E, Usmani OS, Adcock IM,and Barnes PJ (2009) Suppression of GATA-3 nuclear import and phosphory-lation: a novel mechanism of corticosteroid action in allergic disease. PLoS Med6:e1000076.

Mannick JB, Del Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B,Lonetto MA, Maecker HT, Kovarik J, and Carson S, et al. (2014) mTOR inhibitionimproves immune function in the elderly. Sci Transl Med 6:268ra179.

Manning G, Whyte DB, Martinez R, Hunter T, and Sudarsanam S (2002) The proteinkinase complement of the human genome. Science 298:1912–1934.

Marks-Konczalik J, Costa M, Robertson J, McKie E, Yang S, and Pascoe S (2015) Apost-hoc subgroup analysis of data from a six month clinical trial comparing theefficacy and safety of losmapimod in moderate-severe COPD patients with #2%and .2% blood eosinophils. Respir Med 109:860–869.

Martin-Montalvo A, Mercken EM, Mitchell SJ, Palacios HH, Mote PL, Scheibye-Knudsen M, Gomes AP, Ward TM, Minor RK, and Blouin MJ, et al. (2013) Met-formin improves healthspan and lifespan in mice. Nat Commun 4:2192.

Marwick JA, Caramori G, Casolari P, Mazzoni F, Kirkham PA, Adcock IM, ChungKF, and Papi A (2010a) A role for phosphoinositol 3-kinase delta in the impairmentof glucocorticoid responsiveness in patients with chronic obstructive pulmonarydisease. J Allergy Clin Immunol 125:1146–1153.

Marwick JA, Caramori G, Stevenson CS, Casolari P, Jazrawi E, Barnes PJ, Ito K,Adcock IM, Kirkham PA, and Papi A (2009) Inhibition of PI3Kdelta restores glu-cocorticoid function in smoking-induced airway inflammation in mice. Am J RespirCrit Care Med 179:542–548.

Marwick JA, Chung KF, and Adcock IM (2010b) Phosphatidylinositol 3-kinase iso-forms as targets in respiratory disease. Ther Adv Respir Dis 4:19–34.

Ma�slanka T, Otrocka-Domagała I, Zu�ska-Prot M, Mikiewicz M, Przybysz J, JasieckaA, and Jaroszewski JJ (2016) IkB kinase b inhibitor, IMD-0354, prevents allergicasthma in a mouse model through inhibition of CD4(+) effector T cell responses inthe lung-draining mediastinal lymph nodes. Eur J Pharmacol 775:78–85.

Massagué J (2012) TGFb signalling in context. Nat Rev Mol Cell Biol 13:616–630.Matsubara S, Koya T, Takeda K, Joetham A, Miyahara N, Pine P, Masuda ES,Swasey CH, and Gelfand EW (2006a) Syk activation in dendritic cells is essentialfor airway hyperresponsiveness and inflammation. Am J Respir Cell Mol Biol 34:426–433.

Matsubara S, Li G, Takeda K, Loader JE, Pine P, Masuda ES, Miyahara N, MiyaharaS, Lucas JJ, and Dakhama A, et al. (2006b) Inhibition of spleen tyrosine kinaseprevents mast cell activation and airway hyperresponsiveness. Am J Respir CritCare Med 173:56–63.

Matsunaga Y, Inoue H, Fukuyama S, Yoshida H, Moriwaki A, Matsumoto T,Matsumoto K, Asai Y, Kubo M, and Yoshimura A, et al. (2011) Effects of a Januskinase inhibitor, pyridone 6, on airway responses in a murine model of asthma.Biochem Biophys Res Commun 404:261–267.

Matthews JG, Irusen E, Chung KF, Barnes PJ, and Adcock IM (1999) p38 Mitogenactivated protein kinase mediates IL-2 and IL-4 mediated changes in glucocorti-coid receptor affinity. Am J Respir Crit Care Med 157:A336.

Mazzi P, Caveggion E, Lapinet-Vera JA, Lowell CA, and Berton G (2015) The Src-Family Kinases Hck and Fgr Regulate Early Lipopolysaccharide-Induced MyeloidCell Recruitment into the Lung and Their Ability To Secrete Chemokines. JImmunol 195:2383–2395.

McMillan DH, Baglole CJ, Thatcher TH, Maggirwar S, Sime PJ, and Phipps RP(2011) Lung-targeted overexpression of the NF-kB member RelB inhibits cigarettesmoke-induced inflammation. Am J Pathol 179:125–133.

Medicherla S, Fitzgerald M, Spicer D, Woodman P, Ma JY, Kapoun AM, ChakravartyS, Dugar S, Protter AA, and Higgins LS (2007) p38alpha-selective mitogen-activated protein kinase inhibitor SD-282 reduces inflammation in a subchronicmodel of tobacco smoke-induced airway inflammation. J Pharmacol Exp Ther 324:921–929.

Medina-Tato DA, Ward SG, and Watson ML (2007) Phosphoinositide 3-kinase sig-nalling in lung disease: leucocytes and beyond. Immunology 121:448–461.

Meltzer EO, Berkowitz RB, and Grossbard EB (2005) An intranasal Syk-kinase in-hibitor (R112) improves the symptoms of seasonal allergic rhinitis in a park en-vironment. J Allergy Clin Immunol 115:791–796.

812 Barnes

Page 26: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

Menet CJ, Rompaey LV, and Geney R (2013) Advances in the discovery of selectiveJAK inhibitors. Prog Med Chem 52:153–223.

Mercado N, Hakim A, Kobayashi Y, Meah S, Usmani OS, Chung KF, Barnes PJ,and Ito K (2012) Restoration of corticosteroid sensitivity by p38 mitogen activatedprotein kinase inhibition in peripheral blood mononuclear cells from severeasthma. PLoS One 7:e41582.

Mercado N, Ito K, and Barnes PJ (2015) Accelerated ageing of the lung in COPD: newconcepts. Thorax 70:482–489.

Mercado N, To Y, Ito K, and Barnes PJ (2011a) Nortriptyline reverses corticosteroidinsensitivity by inhibition of PI3K-d. J Pharmacol Exp Ther 337:465–470.

Mercado N, To Y, Kobayashi Y, Adcock IM, Barnes PJ, and Ito K (2011b) p38mitogen-activated protein kinase-g inhibition by long-acting b2 adrenergic ago-nists reversed steroid insensitivity in severe asthma. Mol Pharmacol 80:1128–1135.

Millan DS (2011) What is the potential for inhaled p38 inhibitors in the treatment ofchronic obstructive pulmonary disease? Future Med Chem 3:1635–1645.

Millan DS, Bunnage ME, Burrows JL, Butcher KJ, Dodd PG, Evans TJ, Fairman DA,Hughes SJ, Kilty IC, and Lemaitre A, et al. (2011) Design and synthesis of inhaledp38 inhibitors for the treatment of chronic obstructive pulmonary disease. J MedChem 54:7797–7814.

Minshall EM, Leung DY, Martin RJ, Song YL, Cameron L, Ernst P, and Hamid Q(1997) Eosinophil-associated TGF-beta1 mRNA expression and airways fibrosis inbronchial asthma. Am J Respir Cell Mol Biol 17:326–333.

Mitani A, Ito K, Vuppusetty C, Barnes PJ, and Mercado N (2015) Inhibition of mTORrestores corticosteroid sensitivity in chronic obstructive pulmonary disease. Am JRespir Crit Care Med 193:143–153.

Mochly-Rosen D, Das K, and Grimes KV (2012) Protein kinase C, an elusive thera-peutic target? Nat Rev Drug Discov 11:937–957.

Mócsai A, Ruland J, and Tybulewicz VL (2010) The SYK tyrosine kinase: a crucialplayer in diverse biological functions. Nat Rev Immunol 10:387–402.

Moretto N, Bertolini S, Iadicicco C, Marchini G, Kaur M, Volpi G, Patacchini R, SinghD, and Facchinetti F (2012) Cigarette smoke and its component acrolein augmentIL-8/CXCL8 mRNA stability via p38 MAPK/MK2 signaling in human pulmonarycells. Am J Physiol Lung Cell Mol Physiol 303:L929–L938.

Morphy R (2010) Selectively nonselective kinase inhibition: striking the right bal-ance. J Med Chem 53:1413–1437.

Moy LY, Jia Y, Caniga M, Lieber G, Gil M, Fernandez X, Sirkowski E, Miller R,Alexander JP, and Lee HH, et al. (2013) Inhibition of spleen tyrosine kinase at-tenuates allergen-mediated airway constriction. Am J Respir Cell Mol Biol 49:1085–1092.

Mushaben EM, Brandt EB, Hershey GK, and Le Cras TD (2013) Differential effectsof rapamycin and dexamethasone in mouse models of established allergic asthma.PLoS One 8:e54426.

Myou S, Leff AR, Myo S, Boetticher E, Tong J, Meliton AY, Liu J, Munoz NM,and Zhu X (2003) Blockade of inflammation and airway hyperresponsiveness inimmune-sensitized mice by dominant-negative phosphoinositide 3-kinase-TAT. JExp Med 198:1573–1582.

Nadel JA and Burgel PR (2001) The role of epidermal growth factor in mucus pro-duction. Curr Opin Pharmacol 1:254–258.

Nakamaru Y, Vuppusetty C, Wada H, Milne JC, Ito M, Rossios C, Elliot M, Hogg J,Kharitonov S, and Goto H, et al. (2009) A protein deacetylase SIRT1 is a negativeregulator of metalloproteinase-9. FASEB J 23:2810–2819.

Nashed BF, Zhang T, Al-Alwan M, Srinivasan G, Halayko AJ, Okkenhaug K,Vanhaesebroeck B, Hayglass KT, and Marshall AJ (2007) Role of the phosphoi-nositide 3-kinase p110delta in generation of type 2 cytokine responses and allergicairway inflammation. Eur J Immunol 37:416–424.

Nath P, Eynott P, Leung SY, Adcock IM, Bennett BL, and Chung KF (2005) Potentialrole of c-Jun NH2-terminal kinase in allergic airway inflammation and remodel-ling: effects of SP600125. Eur J Pharmacol 506:273–283.

Nath P, Leung SY, Williams A, Noble A, Chakravarty SD, Luedtke GR, Medicherla S,Higgins LS, Protter A, and Chung KF (2006) Importance of p38 mitogen-activatedprotein kinase pathway in allergic airway remodelling and bronchial hyper-responsiveness. Eur J Pharmacol 544:160–167.

Newton R, Holden NS, Catley MC, Oyelusi W, Leigh R, Proud D, and Barnes PJ(2007) Repression of inflammatory gene expression in human pulmonary epithelialcells by small-molecule IkappaB kinase inhibitors. J Pharmacol Exp Ther 321:734–742.

Ngkelo A, Hoffmann RF, Durham AL, Marwick JA, Brandenburg SM, de Bruin HG,Jonker MR, Rossios C, Tsitsiou E, and Caramori G, et al. (2015) Glycogen synthasekinase-3b modulation of glucocorticoid responsiveness in COPD. Am J PhysiolLung Cell Mol Physiol 309:L1112–L1123.

Nijjar JS, Tindell A, McInnes IB, and Siebert S (2013) Inhibition of spleen tyrosinekinase in the treatment of rheumatoid arthritis. Rheumatology (Oxford) 52:1556–1562.

Norman P (2014a) Respivert’s multikinase inhibitors, an evaluation ofWO2014033446 to WO2014033449. Expert Opin Ther Pat 24:1397–1407.

Norman P (2014b) Spleen tyrosine kinase inhibitors: a review of the patent literature2010 - 2013. Expert Opin Ther Pat 24:573–595.

Norman P (2015) Investigational p38 inhibitors for the treatment of chronic ob-structive pulmonary disease. Expert Opin Investig Drugs 24:383–392.

Norman RA, Schott AK, Andrews DM, Breed J, Foote KM, Garner AP, Ogg D, OrmeJP, Pink JH, and Roberts K, et al. (2012a) Protein-ligand crystal structures canguide the design of selective inhibitors of the FGFR tyrosine kinase. J Med Chem55:5003–5012.

Norman RA, Toader D, and Ferguson AD (2012b) Structural approaches to obtainkinase selectivity. Trends Pharmacol Sci 33:273–278.

Onions ST, Ito K, Charron CE, Brown RJ, Colucci M, Frickel F, Hardy G, Joly K,King-Underwood J, and Kizawa Y, et al. (2016) Discovery of narrow spectrumkinase inhibitors: new therapeutic agents for the treatment of COPD and steroid-resistant asthma. J Med Chem 59:1727–1746.

O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, and Laurence A(2015) The JAK-STAT pathway: impact on human disease and therapeutic in-tervention. Annu Rev Med 66:311–328.

Papi A, Contoli M, Adcock IM, Bellettato C, Padovani A, Casolari P, Stanciu LA,Barnes PJ, Johnston SL, and Ito K, et al. (2013) Rhinovirus infection causes steroidresistance in airway epithelium through nuclear factor kB and c-Jun N-terminalkinase activation. J Allergy Clin Immunol 132:1075–1085.e6.

Paredi P and Barnes PJ (2009) The airway vasculature: recent advances and clinicalimplications. Thorax 64:444–450.

Park CS, Bang BR, Kwon HS, Moon KA, Kim TB, Lee KY, Moon HB, and Cho YS(2012) Metformin reduces airway inflammation and remodeling via activation ofAMP-activated protein kinase. Biochem Pharmacol 84:1660–1670.

Partridge MR, van der Molen T, Myrseth SE, and Busse WW (2006) Attitudes andactions of asthma patients on regular maintenance therapy: the INSPIRE study.BMC Pulm Med 6:13.

Paschalaki KE, Starke RD, Hu Y, Mercado N, Margariti A, Gorgoulis VG, Randi AM,and Barnes PJ (2013) Dysfunction of endothelial progenitor cells from smokers andCOPD patients due to increased DNA damage and senescence. Stem Cells 31:2813–2826.

Penton PC, Wang X, Amatullah H, Cooper J, Godri K, North ML, Khanna N, ScottJA, and Chow CW (2013) Spleen tyrosine kinase inhibition attenuates airwayhyperresponsiveness and pollution-induced enhanced airway response in a chronicmouse model of asthma. J Clin Immunol 131:512–520.

Pera T, Atmaj C, van der Vegt M, Halayko AJ, Zaagsma J, and Meurs H (2012) Rolefor TAK1 in cigarette smoke-induced proinflammatory signaling and IL-8 releaseby human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 303:L272–L278.

Pera T, Sami R, Zaagsma J, and Meurs H (2011) TAK1 plays a major role in growthfactor-induced phenotypic modulation of airway smooth muscle. Am J PhysiolLung Cell Mol Physiol 301:L822–L828.

Pernis AB and Rothman PB (2002) JAK-STAT signaling in asthma. J Clin Invest109:1279–1283.

Poh AR, O’Donoghue RJ, and Ernst M (2015) Hematopoietic cell kinase (HCK) as atherapeutic target in immune and cancer cells. Oncotarget 6:15752–15771.

Possa SS, Charafeddine HT, Righetti RF, da Silva PA, Almeida-Reis R, Saraiva-Romanholo BM, Perini A, Prado CM, Leick-Maldonado EA, and Martins MA, et al.(2012) Rho-kinase inhibition attenuates airway responsiveness, inflammation,matrix remodeling, and oxidative stress activation induced by chronic in-flammation. Am J Physiol Lung Cell Mol Physiol 303:L939–L952.

Postma DS and Rabe KF (2015) The Asthma-COPD Overlap Syndrome. N Engl JMed 373:1241–1249.

Rajendrasozhan S, Yang SR, Kinnula VL, and Rahman I (2008) SIRT1, an antiin-flammatory and antiaging protein, is decreased in lungs of patients with chronicobstructive pulmonary disease. Am J Respir Crit Care Med 177:861–870.

Ramis I, Calama E, Domènech A, Carreño C, Calaf E, Cordoba M, Alberti J, De AlbaJ, Bach J, and Prats N, et al. (2014) New inhaled JAK inhibitor LAS194046 in-hibits allergen-induced airway inflammation in Brown Norway rats. Eur Respir J44 (Suppl 58):P1508.

Ramis I, Otal R, Carreño C, Domènech A, Eichhorn P, Orellana A, Maldonado M, DeAlba J, Prats N, and Fernández JC, et al. (2015) A novel inhaled Syk inhibitorblocks mast cell degranulation and early asthmatic response. Pharmacol Res 99:116–124.

Randhawa V and Bagler G (2012) Identification of SRC as a potent drug target forasthma, using an integrative approach of protein interactome analysis and in silicodrug discovery. OMICS 16:513–526.

Rastrick JM, Stevenson CS, Eltom S, Grace M, Davies M, Kilty I, Evans SM,Pasparakis M, Catley MC, and Lawrence T, et al. (2013) Cigarette smoke inducedairway inflammation is independent of NF-kB signalling. PLoS One 8:e54128.

Reber L, Da Silva CA, and Frossard N (2006) Stem cell factor and its receptor c-Kit astargets for inflammatory diseases. Eur J Pharmacol 533:327–340.

Renda T, Baraldo S, Pelaia G, Bazzan E, Turato G, Papi A, Maestrelli P, Maselli R,Vatrella A, and Fabbri LM, et al. (2008) Increased activation of p38 MAPK inCOPD. Eur Respir J 31:62–69.

Riccaboni M, Bianchi I, and Petrillo P (2010) Spleen tyrosine kinases: biology,therapeutic targets and drugs. Drug Discov Today 15:517–530.

Riento K and Ridley AJ (2003) Rocks: multifunctional kinases in cell behaviour. NatRev Mol Cell Biol 4:446–456.

Righetti RF, Pigati PA, Possa SS, Habrum FC, Xisto DG, Antunes MA, Leick EA,Prado CM, Martins MdeA, and Rocco PR, et al. (2014) Effects of Rho-kinase in-hibition in lung tissue with chronic inflammation. Respir Physiol Neurobiol 192:134–146.

Robert V, Triffaux E, Paulet PE, Guéry JC, Pelletier L, and Savignac M (2014)Protein kinase C-dependent activation of CaV1.2 channels selectively controlshuman TH2-lymphocyte functions. J Allergy Clin Immunol 133:1175–1183.

Roskoski R Jr (2015) Src protein-tyrosine kinase structure, mechanism, and smallmolecule inhibitors. Pharmacol Res 94:9–25.

Rosse C, Linch M, Kermorgant S, Cameron AJ, Boeckeler K, and Parker PJ (2010)PKC and the control of localized signal dynamics. Nat Rev Mol Cell Biol 11:103–112.

Rossetti M, Savineau JP, Crevel H, and Marthan R (1995) Role of protein kinase C innonsensitized and passively sensitized human isolated bronchial smooth muscle.Am J Physiol 268:L966–L971.

Rückle T, Schwarz MK, and Rommel C (2006) PI3Kgamma inhibition: towards an‘aspirin of the 21st century’? Nat Rev Drug Discov 5:903–918.

Russell P, Cass L, and Ito K (2013) Russell P, Cass L, Ito K, Safety, pharmacokineticand pharmacodynamic profile of RV568, a narrow spectrum kinase inhibitor, fol-lowing repeat inhaled dosing in COPD patients. Eur Resp J 2013;42:1371. EurRespir J 42:1371.

Ruwanpura SM, McLeod L, Miller A, Jones J, Vlahos R, Ramm G, Longano A, BardinPG, Bozinovski S, and Anderson GP, et al. (2012) Deregulated Stat3 signaling

Kinase Inhibitors in Airway Disease 813

Page 27: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

dissociates pulmonary inflammation from emphysema in gp130 mutant mice. Am JPhysiol Lung Cell Mol Physiol 302:L627–L639.

Saccani S, Pantano S, and Natoli G (2002) p38-Dependent marking of inflammatorygenes for increased NF-k B recruitment. Nat Immunol 3:69–75.

Salt IP and Palmer TM (2012) Exploiting the anti-inflammatory effects of AMP-activated protein kinase activation. Expert Opin Investig Drugs 21:1155–1167.

Sapey E, Greenwood H, Walton G, Mann E, Love A, Aaronson N, Insall RH, StockleyRA, and Lord JM (2014) Phosphoinositide 3-kinase inhibition restores neutrophilaccuracy in the elderly: toward targeted treatments for immunosenescence. Blood123:239–248.

Sato S, Sanjo H, Takeda K, Ninomiya-Tsuji J, Yamamoto M, Kawai T, Matsumoto K,Takeuchi O, and Akira S (2005) Essential function for the kinase TAK1 in innateand adaptive immune responses. Nat Immunol 6:1087–1095.

Scanlon ST and McKenzie AN (2012) Type 2 innate lymphoid cells: new players inasthma and allergy. Curr Opin Immunol 24:707–712.

Scarpulla RC (2011) Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network. Biochim Biophys Acta 1813:1269–1278.

Schaafsma D, Bos IS, Zuidhof AB, Zaagsma J, and Meurs H (2008a) The inhaled Rhokinase inhibitor Y-27632 protects against allergen-induced acute bronchocon-striction, airway hyperresponsiveness, and inflammation. Am J Physiol Lung CellMol Physiol 295:L214–L219.

Schaafsma D, Gosens R, Zaagsma J, Halayko AJ, and Meurs H (2008b) Rho kinaseinhibitors: a novel therapeutical intervention in asthma? Eur J Pharmacol 585:398–406.

Schlapbach A and Huppertz C (2009) Low-molecular-weight MK2 inhibitors: a toughnut to crack! Future Med Chem 1:1243–1257.

Schwartz DM, Bonelli M, Gadina M, and O’Shea JJ (2016) Type I/II cytokines, JAKs,and new strategies for treating autoimmune diseases. Nat Rev Rheumatol 12:25–36.

Schwartz S, Wongvipat J, Trigwell CB, Hancox U, Carver BS, Rodrik-OutmezguineV, Will M, Yellen P, de Stanchina E, and Baselga J, et al. (2015) Feedback sup-pression of PI3Ka signaling in PTEN-mutated tumors is relieved by selective in-hibition of PI3Kb. Cancer Cell 27:109–122.

Shaifta Y, Irechukwu N, Prieto-Lloret J, MacKay CE, Marchon KA, Ward JP,and Knock GA (2015) Divergent modulation of Rho-kinase and Ca(2+) influxpathways by Src family kinases and focal adhesion kinase in airway smoothmuscle. Br J Pharmacol 172:5265–5280.

Shao MX and Nadel JA (2005) Neutrophil elastase induces MUC5AC mucin pro-duction in human airway epithelial cells via a cascade involving protein kinase C,reactive oxygen species, and TNF-alpha-converting enzyme. J Immunol 175:4009–4016.

Shen H, Yoshida H, Yan F, Li W, Xu F, Huang H, Jono H, and Li JD (2008) Syn-ergistic induction of MUC5AC mucin by nontypeable Haemophilus influenzae andStreptococcus pneumoniae. Biochem Biophys Res Commun 365:795–800.

Shigeta A, Tada Y, Wang JY, Ishizaki S, Tsuyusaki J, Yamauchi K, Kasahara Y,Iesato K, Tanabe N, and Takiguchi Y, et al. (2012) CD40 amplifies Fas-mediatedapoptosis: a mechanism contributing to emphysema. Am J Physiol Lung Cell MolPhysiol 303:L141–L151.

Shinkai M, López-Boado YS, and Rubin BK (2007) Clarithromycin has an immuno-modulatory effect on ERK-mediated inflammation induced by Pseudomonas aer-uginosa flagellin. J Antimicrob Chemother 59:1096–1101.

Shuto T, Xu H, Wang B, Han J, Kai H, Gu XX, Murphy TF, Lim DJ, and Li JD (2001)Activation of NF-kappa B by nontypeable Hemophilus influenzae is mediated bytoll-like receptor 2-TAK1-dependent NIK-IKK alpha /beta-I kappa B alpha andMKK3/6-p38 MAP kinase signaling pathways in epithelial cells. Proc Natl Acad SciUSA 98:8774–8779.

Simeone-Penney MC, Severgnini M, Rozo L, Takahashi S, Cochran BH, and SimonAR (2008) PDGF-induced human airway smooth muscle cell proliferation requiresSTAT3 and the small GTPase Rac1. Am J Physiol Lung Cell Mol Physiol 294:L698–L704.

Singh D, Siew L, Christensen J, Plumb J, Clarke GW, Greenaway S, Perros-HuguetC, Clarke N, Kilty I, and Tan L (2015) Oral and inhaled p38 MAPK inhibitors:effects on inhaled LPS challenge in healthy subjects. Eur J Clin Pharmacol 71:1175–1184.

Singh D, Smyth L, Borrill Z, Sweeney L, and Tal-Singer R (2010) A randomized,placebo-controlled study of the effects of the p38 MAPK inhibitor SB-681323 onblood biomarkers of inflammation in COPD patients. J Clin Pharmacol 50:94–100.

Smith SJ, Fenwick PS, Nicholson AG, Kirschenbaum F, Finney-Hayward TK,Higgins LS, Giembycz MA, Barnes PJ, and Donnelly LE (2006) Inhibitory effect ofp38 mitogen-activated protein kinase inhibitors on cytokine release from humanmacrophages. Br J Pharmacol 149:393–404.

Sommers CD, Thompson JM, Guzova JA, Bonar SL, Rader RK, Mathialagan S,Venkatraman N, Holway VW, Kahn LE, and Hu G, et al. (2009) Novel tight-binding inhibitory factor-kappaB kinase (IKK-2) inhibitors demonstrate target-specific anti-inflammatory activities in cellular assays and following oral and localdelivery in an in vivo model of airway inflammation. J Pharmacol Exp Ther 330:377–388.

Song MS, Salmena L, and Pandolfi PP (2012) The functions and regulation of thePTEN tumour suppressor. Nat Rev Mol Cell Biol 13:283–296.

Springer J, Scholz FR, Peiser C, Groneberg DA, and Fischer A (2004) SMAD-signaling in chronic obstructive pulmonary disease: transcriptional down-regulation of inhibitory SMAD 6 and 7 by cigarette smoke. Biol Chem 385:649–653.

Starosyla SA, Volynets GP, Lukashov SS, Gorbatiuk OB, Golub AG, Bdzhola VG,and Yarmoluk SM (2015) Identification of apoptosis signal-regulating kinase1 (ASK1) inhibitors among the derivatives of benzothiazol-2-yl-3-hydroxy-5-phenyl-1,5-dihydro-pyrrol-2-one. Bioorg Med Chem 23:2489–2497.

Steinberg GR and Kemp BE (2009) AMPK in health and disease. Physiol Rev 89:1025–1078.

Stenton GR, Kim MK, Nohara O, Chen CF, Hirji N, Wills FL, Gilchrist M, HwangPH, Park JG, and Finlay W, et al. (2000) Aerosolized Syk antisense suppresses Syk

expression, mediator release from macrophages, and pulmonary inflammation. JImmunol 164:3790–3797.

Stenton GR, Mackenzie LF, Tam P, Cross JL, Harwig C, Raymond J, Toews J,Chernoff D, MacRury T, and Szabo C (2013a) Characterization of AQX-1125, asmall-molecule SHIP1 activator: Part 2. Efficacy studies in allergic and pulmonaryinflammation models in vivo. Br J Pharmacol 168:1519–1529.

Stenton GR, Mackenzie LF, Tam P, Cross JL, Harwig C, Raymond J, Toews J, Wu J,Ogden N, and MacRury T, et al. (2013b) Characterization of AQX-1125, a small-molecule SHIP1 activator: Part 1. Effects on inflammatory cell activation andchemotaxis in vitro and pharmacokinetic characterization in vivo. Br J Pharmacol168:1506–1518.

Stenton GR, Ulanova M, Déry RE, Merani S, Kim MK, Gilchrist M, Puttagunta L,Musat-Marcu S, James D, and Schreiber AD, et al. (2002) Inhibition of allergicinflammation in the airways using aerosolized antisense to Syk kinase. J Immunol169:1028–1036.

Sugita A, Ogawa H, Azuma M, Muto S, Honjo A, Yanagawa H, Nishioka Y, Tani K,Itai A, and Sone S (2009) Antiallergic and anti-inflammatory effects of a novel IkappaB kinase beta inhibitor, IMD-0354, in a mouse model of allergic in-flammation. Int Arch Allergy Immunol 148:186–198.

Sun L, Deng L, Ea CK, Xia ZP, and Chen ZJ (2004) The TRAF6 ubiquitin ligase andTAK1 kinase mediate IKK activation by BCL10 and MALT1 in T lymphocytes.MolCell 14:289–301.

Sun SC (2012) The noncanonical NF-kB pathway. Immunol Rev 246:125–140.Suzuki J, Ogawa M, Muto S, Itai A, Isobe M, Hirata Y, and Nagai R (2011) Novel IkBkinase inhibitors for treatment of nuclear factor-kB-related diseases. Expert OpinInvestig Drugs 20:395–405.

Szatmáry Z, Garabedian MJ, and Vilcek J (2004) Inhibition of glucocorticoidreceptor-mediated transcriptional activation by p38 mitogen-activated protein(MAP) kinase. J Biol Chem 279:43708–43715.

Takahashi-Yanaga F (2013) Activator or inhibitor? GSK-3 as a new drug target.Biochem Pharmacol 86:191–199.

Takeyama K, Dabbagh K, Jeong Shim J, Dao-Pick T, Ueki IF, and Nadel JA (2000)Oxidative stress causes mucin synthesis via transactivation of epidermal growthfactor receptor: role of neutrophils. J Immunol 164:1546–1552.

Takeyama K, Jung B, Shim JJ, Burgel PR, Dao-Pick T, Ueki IF, Protin U, Kroschel P,and Nadel JA (2001) Activation of epidermal growth factor receptors is responsiblefor mucin synthesis induced by cigarette smoke. Am J Physiol Lung Cell MolPhysiol 280:L165–L172.

Takezawa K, Ogawa T, Shimizu S, and Shimizu T (2016) Epidermal growth factorreceptor inhibitor AG1478 inhibits mucus hypersecretion in airway epithelium. AmJ Rhinol Allergy 30:1–6.

Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, Satoh M, Okada Y, YamasawaF, Nakahara K, and Umeda A (2001) Increased expression of transforming growthfactor-beta1 in small airway epithelium from tobacco smokers and patients withchronic obstructive pulmonary disease (COPD). Am J Respir Crit Care Med 163:1476–1483.

Taraseviciene-Stewart L, Douglas IS, Nana-Sinkam PS, Lee JD, Tuder RM, NicollsMR, and Voelkel NF (2006) Is alveolar destruction and emphysema in chronicobstructive pulmonary disease an immune disease? Proc Am Thorac Soc 3:687–690.

Thomas MJ, Smith A, Head DH, Milne L, Nicholls A, Pearce W, Vanhaesebroeck B,Wymann MP, Hirsch E, and Trifilieff A, et al. (2005) Airway inflammation:chemokine-induced neutrophilia and the class I phosphoinositide 3-kinases. Eur JImmunol 35:1283–1291.

Thorarensen A and Kaila N (2014) New spleen tyrosine kinase inhibitors: patentapplications published during 2011-2013. Pharm Pat Anal 3:523–541.

To Y, Ito K, Kizawa Y, Failla M, Ito M, Kusama T, Elliot M, Hogg JC, Adcock IM,and Barnes PJ (2010) Targeting phosphoinositide-3-kinase-delta with theophyllinereverses corticosteroid insensitivity in chronic obstructive pulmonary disease. AmJ Respir Crit Care Med 182:897–904.

Trempolec N, Dave-Coll N, and Nebreda AR (2013) SnapShot: p38 MAPK substrates.Cell 152:924–924.e1.

Tudhope SJ, Catley MC, Fenwick PS, Russell RE, Rumsey WL, Newton R, BarnesPJ, and Donnelly LE (2007) The role of IkappaB kinase 2, but not activation ofNF-kappaB, in the release of CXCR3 ligands from IFN-g-stimulated humanbronchial epithelial cells. J Immunol 179:6237–6245.

Tudhope SJ, Finney-Hayward TK, Nicholson AG, Mayer RJ, Barnette MS, BarnesPJ, and Donnelly LE (2008) Different mitogen-activated protein kinase-dependentcytokine responses in cells of the monocyte lineage. J Pharmacol Exp Ther 324:306–312.

van der Velden JL, Hoffman SM, Alcorn JF, Tully JE, Chapman DG, Lahue KG,Guala AS, Lundblad LK, Aliyeva M, and Daphtary N, et al. (2014) Absence of c-JunNH2-terminal kinase 1 protects against house dust mite-induced pulmonaryremodeling but not airway hyperresponsiveness and inflammation. Am J PhysiolLung Cell Mol Physiol 306:L866–L875.

Vanhaesebroeck B and Khwaja A (2014) PI3Kd inhibition hits a sensitive spot inB cell malignancies. Cancer Cell 25:269–271.

Vanhaesebroeck B, Stephens L, and Hawkins P (2012) PI3K signalling: the path todiscovery and understanding. Nat Rev Mol Cell Biol 13:195–203.

Vanhaesebroeck B, Whitehead MA, and Piñeiro R (2016) Molecules in medicine mini-review: isoforms of PI3K in biology and disease. J Mol Med (Berl) 94:5–11.

von Mehren M (2006) Beyond imatinib: second generation c-KIT inhibitors for themanagement of gastrointestinal stromal tumors. Clin Colorectal Cancer 6 (Suppl1):S30–S34.

Wang D, Boerner SA, Winkler JD, and LoRusso PM (2007) Clinical experience ofMEK inhibitors in cancer therapy. Biochim Biophys Acta 1773:1248–1255.

Wang B, Cleary PP, Xu H, and Li JD (2003) Up-regulation of interleukin-8 by novelsmall cytoplasmic molecules of nontypeable Haemophilus influenzae via p38 andextracellular signal-regulated kinase pathways. Infect Immun 71:5523–5530.

814 Barnes

Page 28: Kinases as Novel Therapeutic Targets in Asthma …pharmrev.aspetjournals.org/.../pharmrev/68/3/788.full.pdfof protein kinase inhibitors in inflammatory airway diseases. II. Role of

Wang X, Khanna N, Wu J, Godri Pollitt K, Evans GJ, Chow CW, and Scott JA (2015)Syk mediates airway contractility independent of leukocyte function. Allergy 70:429–435.

Wang IM, Stepaniants S, Boie Y, Mortimer JR, Kennedy B, Elliott M, Hayashi S, LoyL, Coulter S, and Cervino S, et al. (2008) Gene expression profiling in patients withchronic obstructive pulmonary disease and lung cancer. Am J Respir Crit Care Med177:402–411.

Wang Z, Wesche H, Stevens T, Walker N, and Yeh WC (2009) IRAK-4 inhibitors forinflammation. Curr Top Med Chem 9:724–737.

Watz H, Barnacle H, Hartley BF, and Chan R (2014) Efficacy and safety of the p38MAPK inhibitor losmapimod for patients with chronic obstructive pulmonarydisease: a randomised, double-blind, placebo-controlled trial. Lancet Respir Med 2:63–72.

Webb BJL, Lindsay MA, Barnes PJ, and Giembycz MA (1997). Protein kinase Cisoenzymes in airway smooth muscle. Biochem J 324:167–175.

Wiegman CH, Russell KE, Sieffert J, Rossios C, Adcock IM, Rothaul A, Main M,and Morgan F (2015) Rhe selective pan-Janus kinase (JAK) inhibitor VR588demonstrates potent anti-inflammatory activity in a murne chronic house dustmite model. Am J Respir Crit Care Med 191:A6435.

Wilkinson JE, Burmeister L, Brooks SV, Chan CC, Friedline S, Harrison DE, Hejt-mancik JF, Nadon N, Strong R, and Wood LK, et al. (2012) Rapamycin slows agingin mice. Aging Cell 11:675–682.

Williams AS, Issa R, Durham A, Leung SY, Kapoun A, Medicherla S, Higgins LS,Adcock IM, and Chung KF (2008) Role of p38 mitogen-activated protein kinase inozone-induced airway hyperresponsiveness and inflammation. Eur J Pharmacol600:117–122.

Wilson R, Cahn AAD, McSherry L, Rambaran C, Sousa A, and Wilbraham D (2013)Safety, tolerability and pharmacokinetics (PK) of single and repeat nebulised dosesof a novel phosphoinositide 3-kinase d inhibitor (PI3Kd), GSK2269557, adminis-tered to healthy male subjects in a phase I study. Eur Respir J 42 (Suppl 57):P729.

Winkler DG, Faia KL, DiNitto JP, Ali JA, White KF, Brophy EE, Pink MM, ProctorJL, Lussier J, and Martin CM, et al. (2013) PI3K-d and PI3K-g inhibition by IPI-145 abrogates immune responses and suppresses activity in autoimmune and in-flammatory disease models. Chem Biol 20:1364–1374.

Woodruff PG, Wolff M, Hohlfeld JM, Krug N, Dransfield MT, Sutherland ER, CrinerGJ, Kim V, Prasse A, and Nivens MC, et al. (2010) Safety and efficacy of an inhaledepidermal growth factor receptor inhibitor (BIBW 2948 BS) in chronic obstructivepulmonary disease. Am J Respir Crit Care Med 181:438–445.

Worby CA and Dixon JE (2014) PTEN. Annu Rev Biochem 83:641–669.Wu P, Clausen MH, and Nielsen TE (2015b) Allosteric small-molecule kinase in-hibitors. Pharmacol Ther 156:59–68.

Wu HM, Fang L, Shen QY, and Liu RY (2015a) SP600125 promotes resolution ofallergic airway inflammation via TLR9 in an OVA-induced murine acute asthmamodel. Mol Immunol 67 (2 Pt B):311–316.

Wu P, Nielsen TE, and Clausen MH (2015c) FDA-approved small-molecule kinaseinhibitors. Trends Pharmacol Sci 36:422–439.

Wu W, Wang T, Dong JJ, Liao ZL, and Wen FQ (2012) Silencing of c-kit with smallinterference RNA attenuates inflammation in a murine model of allergic asthma.Int J Mol Med 30:63–68.

Xie S, Sukkar MB, Issa R, Khorasani NM, and Chung KF (2007) Mechanisms ofinduction of airway smooth muscle hyperplasia by transforming growth factor-beta. Am J Physiol Lung Cell Mol Physiol 293:L245–L253.

Yahiaoui L, Villeneuve A, Valderrama-Carvajal H, Burke F, and Fixman ED (2006)Endothelin-1 regulates proliferative responses, both alone and synergistically withPDGF, in rat tracheal smooth muscle cells. Cell Physiol Biochem 17:37–46.

Yamamoto N, Takeshita K, Shichijo M, Kokubo T, Sato M, Nakashima K, Ishi-mori M, Nagai H, Li YF, and Yura T, et al. (2003) The orally available spleentyrosine kinase inhibitor 2-[7-(3,4-dimethoxyphenyl)-imidazo[1,2-c]pyrimidin-5-ylamino]nicotinamide dihydrochloride (BAY 61-3606) blocks antigen-induced airway inflammation in rodents. J Pharmacol Exp Ther 306:1174–1181.

Yanagisawa S, Sugiura H, Yokoyama T, Yamagata T, Ichikawa T, Akamatsu K,Koarai A, Hirano T, Nakanishi M, and Matsunaga K, et al. (2009) The possiblerole of hematopoietic cell kinase in the pathophysiology of COPD. Chest 135:94–101.

Yang F, Oz HS, Barve S, de Villiers WJ, McClain CJ, and Varilek GW (2001) Thegreen tea polyphenol (-)-epigallocatechin-3-gallate blocks nuclear factor-kappa Bactivation by inhibiting I kappa B kinase activity in the intestinal epithelial cellline IEC-6. Mol Pharmacol 60:528–533.

Yew-Booth L, Birrell MA, Lau MS, Baker K, Jones V, Kilty I, and Belvisi MG (2015)JAK-STAT pathway activation in COPD. Eur Respir J 46:843–845.

Yu Q and Stamenkovic I (2000) Cell surface-localized matrix metalloproteinase-9proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis.Genes Dev 14:163–176.

Yum HY, Cho JY, Miller M, and Broide DH (2011) Allergen-induced coexpression ofbFGF and TGF-b1 by macrophages in a mouse model of airway remodeling: bFGFinduces macrophage TGF-b1 expression in vitro. Int Arch Allergy Immunol 155:12–22.

Ziegelbauer K, Gantner F, Lukacs NW, Berlin A, Fuchikami K, Niki T, Sakai K, InbeH, Takeshita K, and Ishimori M, et al. (2005) A selective novel low-molecular-weight inhibitor of IkappaB kinase-beta (IKK-beta) prevents pulmonary in-flammation and shows broad anti-inflammatory activity. Br J Pharmacol 145:178–192.

Kinase Inhibitors in Airway Disease 815