Molecularly targeted therapies for asthma: Current development, challenges and potential clinical translation

Molecularly targeted therapies for asthma: Current development, challenges and potential clinical translation

Accepted Manuscript Molecularly targeted therapies for asthma: Current development, challenges and potential clinical translation Ibrahim Sulaiman, Li...

3MB Sizes 0 Downloads 42 Views

Accepted Manuscript Molecularly targeted therapies for asthma: Current development, challenges and potential clinical translation Ibrahim Sulaiman, Lim Chee Woei, Soo Hon Liong, Johnson Stanslas PII:

S1094-5539(16)30056-6

DOI:

10.1016/j.pupt.2016.07.005

Reference:

YPUPT 1551

To appear in:

Pulmonary Pharmacology & Therapeutics

Received Date: 9 May 2016 Revised Date:

14 July 2016

Accepted Date: 20 July 2016

Please cite this article as: Sulaiman I, Woei LC, Liong SH, Stanslas J, Molecularly targeted therapies for asthma: Current development, challenges and potential clinical translation, Pulmonary Pharmacology & Therapeutics (2016), doi: 10.1016/j.pupt.2016.07.005. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT MOLECULARLY TARGETED THERAPIES FOR ASTHMA: CURRENT DEVELOPMENT, CHALLENGES AND POTENTIAL CLINICAL TRANSLATION

3

Ibrahim Sulaiman, Lim Chee Woei, Soo Hon Liong and Johnson Stanslas.

4 5

Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.

30 31 32 33

SC M AN U TE D EP

29

Corresponding Author: Prof Johnson Stanslas, Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia. Email: [email protected]; [email protected]; Phone: 603-89472310; Fax: 603 89472789

AC C

6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28

RI PT

1 2

34 35 36

1

ACCEPTED MANUSCRIPT Abstract:

38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Extensive research into the therapeutics of asthma has yielded numerous effective interventions over the past few decades. However, adverse effects and ineffectiveness of most of these medications especially in the management of steroid resistant severe asthma necessitate the development of better medications. Numerous drug targets with inherent airway smooth muscle tone modulatory role have been identified for asthma therapy. This article reviews the latest understanding of underlying molecular aetiology of asthma towards design and development of better antiasthma drugs. New drug candidates with their putative targets that have shown promising results in the preclinical and/or clinical trials are summarised. Examples of these interventions include restoration of Th1/Th2 balance by the use of newly developed immunomodulators such as toll-like receptor-9 activators (CYT003QbG10 and QAX-935). Clinical trials revealed the safety and effectiveness of chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) antagonists such as OC0000459, BI-671800 and ARRY-502 in the restoration of Th1/Th2 balance. Regulation of cytokine activity by the use of newly developed biologics such as benralizumab, reslizumab, mepolizumab, lebrikizumab, tralokinumab, dupilumab and brodalumab are at the stage of clinical development. Transcription factors are potential targets for asthma therapy, for example SB010, a GATA-3 DNAzyme is at its early stage of clinical trial. Other candidates such as inhibitors of Rho kinases (Fasudil and Y-27632), phosphodiesterase inhibitors (GSK256066, CHF 6001, roflumilast, RPL 554) and proteinase of activated receptor-2 (ENMD-1068) are also discussed. Preclinical results of blockade of calcium sensing receptor by the use of calcilytics such as calcitriol abrogates cardinal signs of asthma. Nevertheless, successful translation of promising preclinical data into clinically viable interventions remains a major challenge to the development of novel anti-asthmatics.

61 62

Keywords:

63

Asthma, Inflammation, Airway Remodelling, Anti-Asthmatics, Targets

64

Chemical compounds studied in this article:

65 66 67 68 69

Bis-(5-amidino-2-benzimidazolyl)-methane (PubChem CID: 46936860); Calcitriol (PubChem CID: 5280453); JNJ-39758979 (PubChem CID: 24994634); JNJ-7777120 (PubChem CID: 4908365); PF-3893787 (PubChem CID: 24745335); Y-27632 (PubChem CID: 448042); Fasudil (PubChem CID: 3547); Roflumilast (PubChem CID: 449193), YM-341619 (PubChem CID: 10321901); Afzelin (PubChem CID: 5316673).

AC C

EP

TE D

M AN U

SC

RI PT

37

2

ACCEPTED MANUSCRIPT 1.

Introduction

71

Asthma is an intricate inflammatory airway disease that results from activation of numerous

72

inflammatory and structural cells, often leading to partially or fully reversible airway

73

obstruction, airway inflammation, mucus hypersecretion and acute hyperresponsiveness

74

(Prado et al., 2014). The aetiology of asthma may be genetic, environmental or an interplay of

75

both factors. It places enormous economic burden on patients, families, and the health care

76

system by causing loss of productivity, high number of missed school and/or workdays, high

77

medical bills, and premature death (Barnes et al., 2015). About 334 million people globally

78

were estimated to be asthmatic (Global Asthma Report, 2014; Murray et al., 2013) and this

79

figure was projected to escalate to 400 million by the year 2025 (Global Asthma Report,

80

2014). Statistics have placed asthma as the 14th most important disorder in the world, it

81

affects about 14%, 8.6% and 4.5% of world children, young adults and world population

82

respectively (Murray et al., 2013; To et al., 2012; Global Asthma Report, 2014). Severe

83

uncontrolled asthma account for 5% to 10% of total asthma cases globally, and 50% of total

84

asthma cost (Al-Hajjaj, 2011; Dheda et al., 2015). In Europe alone, the direct and indirect

85

cost of both controlled and uncontrolled asthma in patients between the age of 15-64 was

86

estimated to be €19 billion/annum (Domínguez-Ortega et al., 2015). Asthma, as a global

87

disorder is a source of concern to both developed and developing nations (Behera and Sehgal,

88

2015). Although improvement in asthma conditions can be achieved by the use of currently

89

available conventional therapies such as topical bronchodilators and corticosteroids, these

90

drugs have presented a number of unwanted side effects, possible resistance with long term

91

use and absolute non-responsiveness in some cases (Abramson et al., 2003; Chung et al.,

92

2014). Only a few effective anti-asthma controllers and relievers are currently available

93

(example include steroids such as budesonide and fluticasone, bronchodilators such as

94

salmeterol and levalbuterol). This may be due to inadequate knowledge of the underlying

95

cause of asthma, difficulties in developing inhalable preparations for topical delivery and

96

inordinate animal models used in testing new treatments that mostly fail clinical translation

97

and relevance. This article discusses targets that could aid in developing new interventions

98

with minimal adverse effects as compared to current medications. The role of certain

99

receptors, enzymes and cytokines in the aetiology of asthma was further emphasized.

AC C

EP

TE D

M AN U

SC

RI PT

70

3

ACCEPTED MANUSCRIPT 2.

Pathophysiology of Asthma

101

The cardinal pathophysiological manifestations of asthma are airway inflammation,

102

intermittent obstruction and acute hyperresponsiveness (AHR) (Holgate, 2008). Depending

103

on cellular and molecular characterization of the inflammatory cascade, asthma may either be

104

allergic or non-allergic endotype. Allergic asthma is usually eosinophilic, whereas non-

105

allergic endotypes, such as aspirin, infection and exercise induced asthma may present with

106

neutrophilic or paucigranulocytic phenotype. Most non-allergic asthma is severe, and steroid-

107

resistant (Lötvall et al., 2011).

108

Fundamental in vitro and in vivo studies have revealed several cellular and molecular events

109

involved in development of asthma. These include increased immunoglobulin E (IgE)

110

production by B cells, shift in T helper-type 1/T helper-type 2 (Th1/Th2) paradigm,

111

upregulation of Th2 cytokines, airway cellular infiltrations, dysfunctional activated

112

inflammatory cells, mitochondrial damage, increased production of reactive oxygen species

113

(ROS) and reactive nitrite species (RNS) (Aguilera-Aguirre et al., 2009).

114

Aeroallergens such as house dust mites, pollens and animal dander exert proteolytic and LPS-

115

like activity that permits them to penetrate through the airway epithelium and attach to toll

116

like receptors (TLR) to activate some cascade of events (Wilson et al., 2012). Allergen-TLR

117

interaction induces release of IL-25, IL-33, Thymic stromal lymphopoietin (TSLP),

118

macrophage inflammatory protein-3 (MIP3A) and monocyte chemotactic protein 1 (MCP1)

119

by the airway epithelial cells. IL-25, IL-33 and TSLP enhance the development of Th2 cells

120

while MIP3A (CCL20) and MCP1 (CCL2) are involved in the recruitment and activation of

121

dendritic cells. Activated dendritic cells recognizes and present fragments of allergen peptides

122

to lymph nodal naïve T lymphocytes (Th0) through its major histocompatibility complex

123

(MHC) class II (Kallinich et al., 2007). The differentiation of Th0 into Th1 or Th2 depends on

124

the cytokine milieu. Usually, increased IL-4 (induced by mast cell) and low IL-2 promotes

125

differentiation of Th0 to Th2 cells. Th2 cells constitutively expresses GATA3, a crucial

126

transcription factor that enhances Th2 production of IL-4, IL-5, IL-9 and IL-13 which further

127

improve the suitability of the cytokine milieu for further Th0 - Th2 differentiation (Kaiko et

128

al., 2008). Furthermore, IL-4 and IL-13 incites atopy by enhancing the production of IgE

129

from B lymphocytes, influences secretion of eosinophil and Th2 recruiting cytokines such as

130

IL-4, IL-5, IL-13, granulocyte macrophage colony stimulating factor (GM-CSF) and potent

131

chemoattactants such as eotaxin 1 (CCL11), eotaxin 2 (CCL24) and RANTES (CCL5)

AC C

EP

TE D

M AN U

SC

RI PT

100

4

ACCEPTED MANUSCRIPT (Wenzel, 2013a). Interleukin9 (IL-9), mainly secreted by Th9 (a subset of Th2 cells) promotes

133

mast cell airway infiltration, maturation and degranulation, while IL-5 induces and augment

134

pulmonary eosinophil homing (Fulkerson and Rothenberg, 2013). IL-4 further enhances

135

eosinophil migration by activating airway vascular cell adhesion molecule 1 (VCAM-1).

136

Pulmonary eosinophils secrete wide array of inflammatory mediators such as cysteinyl

137

leukotrienes (LTC4, LTD4, LTE4) and cytotoxic agents such as major basic protein (MBP),

138

eosinophil derived neurotoxin, eosinophil cationic protein and eosinophil peroxidases which

139

all together further aggravate airway inflammation and oxidative stress damage observed in

140

asthma (Hall and Agrawal, 2014; Pelaia et al., 2015). Although eosinophilic asthma is of

141

allergic endotype, non-allergic adult onset asthma can present with airway eosinophilia whose

142

pattern of development is independent of Th2, rather, it depends on combined transcriptional

143

activity of GATA3/RORα transcription factors and type 2 innate lymphoid cells (ILC2) (Woo

144

et al., 2014). Nevertheless, both allergic and non-allergic eosinophilic asthma shows elevated

145

type 2 cytokine levels, thereby indicating the critical role of IL-4, IL-5, IL-13 in the

146

pathogenesis of eosinophilic asthma (Pelaia et al., 2015; Lambrecht and Hammad, 2015;

147

Brusselle et al., 2013).

148

Apart from airway eosinophilia due to Th2 or ILC2, asthma may present with neutrophilic

149

phenotype, which is widely mediated by Th17, a subset of T helper cells that secretes IL-17.

150

This category of asthma is often expressed in severe/uncontrolled asthmatics, steroid

151

insensitive and it may be triggered by allergens or non-allergenic triggers such as microbes,

152

cigarette smoke, diesel exhaust particles and other environmental pollutants (Polosa and

153

Thomson, 2013; Newcomb and Peebles, 2013; Vroman et al., 2015). Differentiation of

154

thymocytes into Th17 cells depends on RAR-related orphan receptor gammaT (RORɣt), a

155

transcription factor that requires IL-1β, IL-6 and transforming growth factor (TGF-β) milieu

156

for its upregulation and consequent Th17 production (Dong, 2008; Vroman et al., 2015). The

157

heterogeneous nature of asthma pathogenesis has rendered blanket approach to treatment and

158

diagnosis of different asthma endotypes ineffective. Therefore, proper understanding of

159

asthma pathogenesis is essential for successful drug discovery and repositioning. Figure 1

160

describes the cellular pathways involved in the pathogenesis of eosinophilic and neutrophilic

161

asthma endotypes.

AC C

EP

TE D

M AN U

SC

RI PT

132

5

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

162

Figure 1: Pathophysiology of asthma: Depending on the trigger and cytokine milieu, pathogenesis of asthma may occur through an eosinophilic or neutrophilic pathway. Antigens are presented to dendritic cells (DC) for onward processing and subsequent presentation to the naïve T-helper cells (Th0). Depending on the cytokine milieu, the Th0 may differentiate into T-helper cells type 1 (Th1), Th17 or Th2. A Th independent pathway occurs through the ILC2 to induce eosinophilic asthma. Treg: T regulatory cells, MMP: matrix metalloproteinase, IL: interleukin, TGF-β: Transforming growth factor-beta., LT: leukotriene, MHC: major histocompatibility II, TSLP: Tissue stromal lymphopoietin, GM-CSF: Granulocyte macrophage-colony stimulating factor, PAF: Platelet-activating factor, ILC2: Innate lymphoid cells 2, MHC-II: Major histocompatibility complex II, GR0-α: Growth regulated protein alpha, Cyst-LTs: cysteinyl leukotrienes,15-HETE: 15-Hydroxyicosatetraenoic acid.

172

TE D

163 164 165 166 167 168 169 170 171

3.

Current Asthma Therapies

174

The overall aim of asthma management is to prevent or control acute and chronic symptoms

175

(such as exacerbations, airway mucus accumulation, narrowing, squeezing and swelling of

176

the airway smooth muscles), stabilize pulmonary function, improve asthma-related quality of

177

life, and provide optimum therapeutic interventions with minimal adverse effects (EPR3,

178

2007). Current anti-asthmatic drugs function as either bronchodilators or anti-inflammatory,

179

or a combination of both. Alternatively, some of the medications are defined according to

180

their targets and mode of action, e.g. the leukotriene modifiers, mast cell stabilizer and IgE

181

inhibitors. Conventionally, asthma is managed by use of single or multiple agents of

182

bronchodilators such as short-acting β2 agonists (e.g pirbuterol, salbutamol and levalbuterol),

183

long-acting β2 agonists (e.g salmeterol and formeterol), anti-cholinergics (e.g tiotropium,

184

oxitropium and ipratropium bromides), and inhaled corticosteroids (ICS) (Barnes, 2006;

AC C

EP

173

6

ACCEPTED MANUSCRIPT Chanez et al., 2004; Feltis et al., 2007). These approaches have recorded wide range of

186

adverse effects, including sympathomimetic side effects (such as anxiety, tremor and

187

tachycardia) ascribed to β2 agonists activity (Abramson et al., 2003; Henderson et al., 2005)

188

while anti-cholinergics were associated with induction of xerostomia (Kato et al., 2006;

189

Rodrigo and Castro-Rodriguez, 2005).

190

Although, steroids are currently the most effective therapy for asthma, they were reported to

191

cause a number of unwanted side effects such as glaucoma, cataract, skin bruise,

192

telangiectasia, osteoporosis, sore throat, dysphonia (due to laryngeal oedema and muscular

193

hypertrophy), hyperglycaemia, adrenal suppression and topical candidiasis (Reddel et al.,

194

2015; Saag et al., 2011; Dahl, 2006; Israel et al., 2001). In addition to aforementioned

195

adverse effects, a group of asthmatics are resistant to steroid intervention due to variety of

196

factors such as alteration in glucocorticoid receptor expression, unstable glucocorticoid

197

receptor activity and failure in nuclear translocation of glucocorticoid-receptor complexes.

198

Inaccurate binding of complexes to glucocorticoid DNA response elements and faulty RNA

199

splicing leading to aberrant cytokine expression or distorted extracellular matrix were also

200

suggested to cause steroid resistance (Chung et al., 2014; Beck et al., 2009). The

201

unavailability of better alternatives to steroid has made the management of steroid resistant

202

asthma to be very challenging.

203

Although, omalizumab, a 95% humanized IgE monoclonal antibodies used in moderate to

204

severe persistent allergic asthma was proven to be effective in management of uncontrolled

205

asthma (Cox, 2009), the antibody has the potential of eliciting anaphylaxis and

206

atherothrombotic events such as myocardial infarction and stroke (Ali and Hartzema, 2012).

207

Few other classes of drugs used in asthma therapy include the mast cell stabilizers (e.g.

208

cromolyn sodium and nedocromil sodium) and leukotriene modifiers (e.g. montelukast,

209

pranlukast, zafirlukast and zileuton) all of which are less effective as compared to steroid

210

therapy (Fanta, 2009; del Giudice et al., 2009). Identification of safe substitute for current

211

asthma medications through targeted therapy is paramount for improved asthma control,

212

management and possible cure.

213

4.

214

Despite the unmet medical needs in prophylaxis and treatment of asthma, very few new

215

classes of drugs were introduced and considered safe and effective over the past 40 years. In

216

general, development of new drugs in the field of respiratory medicine is slow and scanty due

AC C

EP

TE D

M AN U

SC

RI PT

185

Emerging Drug Targets

7

ACCEPTED MANUSCRIPT to fewer drug candidates, higher failure rate and the unsuccessful translation of many positive

218

preclinical findings into clinically relevant data (Barnes et al., 2015). The probability for a

219

newly discovered respiratory drug to enter into the market phase of drug development is only

220

3%, as against to 6–14% in other diseases such as HIV/AIDS, cardiovascular disorders,

221

cancer, dermatological, haematological and neurological diseases (Mestre-Ferrandiz et al.,

222

2012). Meaningful advancements recorded in the understanding of pathophysiology and

223

factors associated with asthma development are critical for the successful design and

224

development of new and more effective candidate drugs. Thus, targeting of critical cytokines,

225

receptors and enzymes implicated in aetiology and progression of specific asthma endotype is

226

critical to successful development of novel interventions.

227

4.1. Target Receptors

228

4.1.1. Toll-like Receptor Activators

229

Toll like receptors (TLRs) are pattern recognition receptors (PRRs) capable of detecting and

230

responding to microbial and endogenously derived signature molecules. Some TLRs are

231

found expressed on surfaces of epithelium, mast cells, fibroblasts, monocytes, macrophages

232

and dendritic cells while others are endosomal or phagosomal in nature. Induction of TLR

233

signalling promotes Th1 and downregulates Th2 responses, thereby countering allergic

234

asthma responses (Hennessy et al., 2010; Meng et al., 2011; Fonseca and Kline, 2009).

235

Considering the percentages of asthmatics that are of allergic endotype (50% and 80% of

236

asthmatic adults and children respectively), TLR targeting will be beneficial in reducing

237

asthma burden (Knudsen et al., 2009).

238

Toll like receptor subtypes TLR3, TLR7, TLR8, and TLR9 are strong inducers of Th1

239

responses (Hennessy et al., 2010). TLR9 agonist QAX-935 (IMO-2134) was reported to

240

decrease airway resistance and inflammation by activating several interferon-dependent genes

241

involved in effective Th1 response (Panter, et al., 2009; Kline and Krieg, 2010). Furthermore,

242

QbG10, a TLR9 agonist, improved overall asthma symptoms in patients with persistent

243

allergic asthma following controlled steroid withdrawal (Casale et al., 2015; Lassen, et al.,

244

2014; Beeh, et al., 2013). Similarly, synthetic TLR9 ligands [unmethylated cytosine-guanine

245

oligodeoxynucleotides (CpG ODNs)] induced Th1 responses and counterbalanced Th2

246

phenotype via Th1 cytokine surge (IFN-γ, IL-10 and IL-12) induced by CpG mediated

247

activation of natural killer (NK) cells through dendritic cell stimulation (Gupta and Agrawal,

248

2010). In a related development, CpG ODNs analogs (1018 ISS and ASM8) suppressed

AC C

EP

TE D

M AN U

SC

RI PT

217

8

ACCEPTED MANUSCRIPT airway eosinophilia, induced T regulatory cell (Tregs) activity and Th2 cytokine production

250

in allergic asthma (Senti et al., 2009; Campbell et al., 2014).

251

A potent TLR8 agonist (vTX-1463) reportedly stimulated the production of Th1 polarizing

252

cytokines (IFNγ and IL-12), decreased airway eosinophila and pulmonary congestion (figure

253

2). Likewise, upregulation and activation of TLR7 markedly suppressed asthma symptoms

254

and exacerbations through a Tregs dependent pathway (Hatchwell et al., 2015; Pham Van et

255

al., 2011). Immunotherapeutic approach by the use of house dust mite (HDM) sublingual

256

allergen immunotherapy (SLIT) tablet was recently reported to reduce the risk of moderate to

257

severe asthma exacerbations among adults with HDM allergy–related asthma that respond

258

poorly to ICS mono or combination therapy with no report of severe systemic adverse

259

reaction (Virchow et al., 2016). This improvement could be credited to allergen

260

desensitization and improved allergen tolerance due to shift in Th2 to Treg paradigm.

261

Although

262

immunotherapy (AIT) is yet to be fully elucidated, the ability of TLR to skew Th2-Th1

263

balance towards Th1-treg profile suggests its likely involvement in AIT. Furthermore, unlike

264

TLR-4, stimulation of TLR-7 and TLR-9 failed to break allergen-specific T-cell tolerance

265

induced by AIT (Akdis and Akdis, 2015). Although, TLR mediated immunotherapy appear

266

safe, the potential adverse effects of the intervention include autoimmunity, hypersensitivity,

267

exaggerated immune reactions and risk of perpetuating vicious inflammatory cycles as

268

obtainable in rheumatoid arthritis (Goh and Midwood, 2012).

269

4.1.2. Chemoattractant receptor-homologous molecule expressed on Th2 cells

270

(CRTH2)

271

Chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) otherwise

272

termed D prostanoid receptor 2 (DP2), is a G protein–coupled receptor that has a potential

273

role in the pathogenesis of allergic disorders such as asthma. The receptor is highly expressed

274

on Th2 cells, while its natural ligand [prostaglandin D2 (PGD2)] was found to be elevated in

275

the bronchoalveolar lavage fluid (BALF) of asthmatics and causes a characteristic cough (Fajt

276

et al., 2013; Stinson et al., 2015; Maher et al., 2015). Activation of CRTH2 enhances airway

277

remodelling, goblet metaplasia, MUC5AC expression and cellular migration as revealed by

278

transbronchial biopsy of severe asthma patients (Stinson et al., 2015). The resultant effects of

279

CRTH2 activation were blocked by AZD6430 (a potent CRTH2 antagonist), suggesting

280

CRTH2 inhibition as remedy to some asthma symptoms. AZD6430 decreased goblet

detailed

molecular

mechanism

behind

desensitization

in

allergen

AC C

EP

TE D

the

M AN U

SC

RI PT

249

9

ACCEPTED MANUSCRIPT metaplasia through MUC5AC repression and ameliorated cellular infiltration through

282

inhibition of PGD2 driven chemotaxic effects on eosinophils, mast cells and Th2

283

lymphocytes (Stinson et al., 2015). Furthermore, downregulation of CRTH2 activity by

284

OC0000459 suppressed airway hyperreactivity, improved forced expiratory volume in 1

285

second (FEV1), reduced total IgE level, reversed sputum eosinophilia and decreased

286

expression of proinflammatory genes in inflamed airways (Lukacs et al., 2008; Barnes et al.,

287

2012; Pettipher et al., 2012). According to a randomised, double-blind, placebo-controlled,

288

two-way crossover clinical trial study (NCT01056692), OC0000459 inhibited allergic airway

289

inflammation by preventing PGD2 mediated airway and serum eosinophilia. In addition,

290

improvement of FEV1 among asthma patients was also reported (Singh et al., 2012).

291

Moreover, administration of oral CRTH2 antagonist BI 671800 to ICS-naïve patients and

292

those on ICS improved asthma symptom by increasing FEV1 in both montelukast and ICS

293

treated patients (Hall et al., 2015). Thus, CRTH2 inhibition is potentially suitable for patients

294

whose asthma conditions are inadequately controlled by ICS monotherapy.

295

4.1.3. Proteinase-activated receptor 2 (PAR-2)

296

PAR2 is a G-protein coupled receptor (GPCR) commonly expressed in airway smooth

297

muscle, leukocytes and bronchial epithelial cells. It is usually activated by proteinases that

298

arises from aeroallergens, invading pathogens, or by endogenously released proteinases such

299

as trypsin, house dust mite (HDM) protein Derp9 and mast cell tryptase (Boitano et al.,

300

2011). The attention given to inhibition of protease activity as potential drug target is

301

currently on the rise (Alam, 2014; Harvima et al., 2014). Many triggers of asthma (e.g. house

302

dust mite, fungal allergens and ovalbumin) exhibit proteinase activity (Corry and

303

Kheradmand, 2009; Kauffman and van der Heide, 2003). The activation of PAR2 occurs by

304

cleavage of its extracellular N terminus to reveal its tethered ligand (SLIGRL-NH2/SLIGKV-

305

NH2) that auto-activates the receptor. Upon PAR2 activation, classical GPCR and β-arrestin

306

signalling pathways are triggered. The latter is pro-inflammatory and occurs due to immense

307

activation of PAR2 while the former is bronchoprotective and occur due to mild activation of

308

PAR2 (Nichols et al., 2012).

309

Prior reports showed 38% reduction and 52% increase in airway hyper-reactivity following

310

methacholine challenge in asthmatic mice lacking PAR2 and those overexpressing PAR2

311

respectively as compared to wild-type (Schmidlin et al., 2002). This implicated the

312

involvement of PAR2 in pathogenesis of hyperresponsiveness in asthma. Activated PAR2

AC C

EP

TE D

M AN U

SC

RI PT

281

10

ACCEPTED MANUSCRIPT stimulates airway epithelium to release cellular mediators that promotes asthma, these

314

mediators include the granulocyte macrophage- colony stimulating factors (GM-CSF),

315

eotaxin and matrix metalloproteinase 9 (Vliagoftis et al., 2000). Bronchial smooth muscles

316

(BSM) PAR2 stimulation increased the proliferation of BSM cells from asthmatic subjects

317

only, while BSM cells from non-asthmatic subjects showed no hyper-proliferative property

318

even after lentiviral over-expression of PAR2. Such increased proliferation could be

319

accounted for by increased basal level of PAR2 in asthmatics airway (Allard et al., 2014).

320

Furthermore,

321

tryptase inhibitor, inhibitors of tryptase (a PAR2 substrate) were found to prevent

322

bronchoconstriction in asthmatics (Sylvin et al., 2002; Hernández-Hernández et al., 2012). It

323

could therefore be suggested that mast cells mediated induction of airway hyper

324

responsiveness is not solely dependent on released histamine but also the endpoint effect of

325

mast cell tryptases on PAR2.

326

4.1.4. Calcium Sensing Receptor (CaSR) Antagonism

327

Calcium-sensing receptor (CaSR) is a calcium-binding G protein-coupled receptor (GPCR),

328

that regulates systemic calcium homeostasis (Breitweiser, 2014). Airway smooth muscle cells

329

contain elevated [Ca2+]i levels while CaSR are upregulated and constitutively activated in

330

airways of asthmatics (Yarova et al., 2015). Increase in [Ca2+]i enhances bronchoconstriction

331

that leads to AHR and induces chronic genomic effects that increases ASM cell proliferation

332

and extracellular matrix components depositions that additively results in airway remodelling

333

(Koopmans et al., 2014; Mahn et al., 2010). Activation of CaSRs is therefore considered a

334

major climactic event leading to asthma pathogenesis. Eosinophilic cationic proteins and

335

major basic proteins released by infiltrating eosinophils are considered to primarily induce

336

airway narrowing in asthma (Pégorier et al., 2006), these polycations exert their effect

337

through activation of airway CaSRs which is normally involved in ASM homeostasis of

338

divalent cations and polycations (figure 3). Blockade of this receptor by the use of calcilytics

339

(e.g calcityrol), were found to reduce AHR and airway inflammation in mouse asthma models

340

(Yarova et al., 2015). Although the concept of using calcilytics as novel asthma intervention

341

it still at its initial stage, the intervention appears promising especially in asthmatics that are

342

non-sensitive to current asthma management strategies (Tanday, 2015).

RI PT

313

and

leech-derived

AC C

EP

TE D

M AN U

SC

APC-366, Bis-(5-amidino-2-benzimidazolyl)-methane

11

ACCEPTED MANUSCRIPT 4.1.5. H4 Receptor Antagonism

344

Plasma and airway histamine levels are heightened among asthmatics especially during

345

exacerbations. The biogenic amine mediates through four GPCR subtypes, the histamine H1-,

346

H2-, H3, and H4-receptors (H1-4R). H4R is considered a promising drug target for

347

inflammatory and allergic disorders due to its broad expression in hematopoietic cells (Seifert

348

et al., 2013; Brimblecombe et al., 2010; Zhang et al., 2007). The mediator is usually released

349

by mast cells and it functions by activating DCs, enhance Th2 polarization as well as airway

350

inflammatory cells homing (Reher et al., 2012; Caron et al., 2001). In vivo cellular

351

characterization of H4R in experimental allergic asthma models identified the role of the

352

receptor in the regulation of dendritic cell (DC) activation during sensitization and its role in

353

eosinophil activation during effector phase. This was further proved by the inability of

354

effector T cells to induce eosinophilic infiltration upon adoptive transfer of DCs devoid of

355

H4R at vitro sensitization reaction stage (Hartwig et al., 2015).

356

H4R selective antagonist, JNJ -7777120 H4R ((5-chloro-1H-indol-2-yl)-(4-methyl-piperazin-

357

1-yl)-methanone) ameliorated asthma symptoms in ovalbumin mouse asthma model.

358

Furthermore, H4R gene knock-out caused decreased allergic pulmonary inflammation,

359

prevented Th2 responses and bronchoconstriction, inhibited airway eosinophil and

360

lymphocyte infiltration (Neumann et al., 2013; Dunford et al., 2006; Somma et al., 2013).

361

The interesting results obtained from preclinical assessment of H4R antagonists prompted

362

further exploration of its clinical application in asthma. H4R selective antagonists showed

363

excellent safety profile following preclinical toxicity studies. Likewise, phase 1 clinical trial

364

has shown that selective H4R antagonists such as UR-63325, JNJ-39758979 [(R)-4-(3-amino-

365

pyrrolidin-1-yl)-6-isopropyl-pyrimidin-2-ylamine]

366

(cyclopropylmethyl)-6-(3-(methylamino)pyrrolidin-1-yl)pyrimidine-2,4-diamine] are safe for

367

use in humans. In addition, JNJ-39758979 dose dependently inhibited histamine induced

368

eosinophil morphological changes (Thurmond et al., 2014; Salcedo, 2013; Mowbray et al.,

369

2011). Unfortunate incidence of agranulocytosis observed in two out of eighty-eight Japanese

370

patients that enrolled for safety and efficacy study of JNJ-39758979 in moderate atopic

371

dermatitis led to the withdrawal of a phase 2 clinical trial of JNJ-39758979 in patients with

372

uncontrolled, persistent asthma prior to enrolment (Murata et al., 2015). This adverse effect

373

may be due to off target effect of the intervention or likely due to unascertained

374

pharmacogenomic variation among subjects. Nevertheless, the intervention was found to be

375

effective before the premature discontinuation of the trial.

and

PF-3893787

[(R)-N4-

AC C

EP

TE D

M AN U

SC

RI PT

343

12

Figure 2: Toll like (TLR) and histamine 4 receptor (H4R) targeting in asthma treatment. TLR7, TLR8 and TLR9 engages their Toll–IL-1-resistence (TIR) domains to myeloid differentiation primary-response protein 88 (MYD88). This in turn stimulates downstream IL-1R-associated kinase 4 (IRAK4) and TNF receptor-associated factor 6 (TRAF6) thereby leading the activation of interferon-regulatory factor 7 (IRF-7), which induced type I interferon (IFN-α). IFN-α provide the cytokine milieu that enhances Th1 polarization by inducing IL-12 secretion from monocyte derived dendritic cells (mDC). Histamine 4 receptor (H4R) activation results in mast cell chemotaxis, downstream Ca2+ surge from endoplasmic reticulum (ER) and upregulation of markers such as malonyldialdehyde (MDA), 8-hydroxy-2’-deoxyguanosine (8OHdG) and transforming growth factor β (TGF-β). Alternatively, H4R activation may lead to decreased adenyl cyclase (AC) activity through Gi protein activation. This decreases intracellular cyclic adenosine monophosphate (cAMP) level, which in turnresults in protein kinase A (PKA) activation. Active PKA phosphorylates cAMP-responsive element-binding protein (CREB), which modulates gene transcription.

EP

377 378 379 380 381 382 383 384 385 386 387 388

AC C

376

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

389

13

ACCEPTED MANUSCRIPT 4.2.

Enzymatic Targets

391

4.2.1. Inhibitors of Rho-Kinases

392

Rho, a member of the Ras superfamily of guanosine triphosphatases (GTPases) and a

393

monomeric G protein associated with regulation of smooth muscle contraction, modulation of

394

myosin light chain phosphorylation and stress fibre formation (primarily actin and non-

395

muscle myosin II fibres). Rho also controls few other physiological events such as

396

cytokinesis, focal adhesion, cell migration, motility, migration and regulation of downstream

397

extracellular signals, such as lysophosphatidic acid (Taki et al., 2007). The contraction of

398

ASM is triggered by phosphorylation of the myosin light chain (MLC) by Ca2+ calmodulin

399

(CaM)-activated MLC kinase in the presence of intracellular calcium, thereby allowing actin-

400

myosin cross-linkage and subsequent shortening of sarcomeres. Whereas, ASM relaxation is

401

caused by MLC dephosphorylation by Rho kinase regulated myosin phosphatase (Chiba et

402

al., 2010; Schaafsma et al., 2006; Burdyga et al., 2003). Phosphorylation of myosin binding

403

subunit of myosin phosphatase by Rho kinases (ROCK) exerts inhibitory effect on MLC

404

dephosphorylation, thereby limiting smooth muscle relaxation (Chiba et al., 2010). Rho-

405

kinases are overexpressed in airway of asthmatics, therefore contributing to intermittence or

406

persistence AHR symptoms in asthma. The additive effect of Rho kinase upregulation results

407

in persistent MLC phosphorylation, contributing to asthma-related cellular remodelling

408

(Holgate, 2008).

409

Aside from ASM tone regulation via MLC phosphatase pathway, Rho kinases also play a

410

vital role in regulating inflammation related cellular infiltration and migrations via their

411

inherent spatiotemporal regulatory potential on the actin cytoskeleton (Biro et al., 2014). In

412

support of this, Rho-kinase inhibition (by use of Y-27632) blocks the cellular expression of

413

NF-κB, an essential transcription factor that contributes to asthma progression by activating

414

genes coding for inflammatory cytokines (Tong and Tergaonkar, 2014).

415

Fasudil (HA-1077), an isoquinoline-based compound is a first generation and the only

416

clinically available ROCK inhibitor that proved to be effective in management of pulmonary

417

arterial hypertension (PAH) and cerebral vasospasm through blockage of ATP dependent

418

domain of Rho-kinase and consequent smooth muscle relaxation (Raja, 2012; Velat et al.,

419

2011; Bain et al., 2007). Fasudil inhibits ROCK induced actomyosin dynamics by

420

competitively binding to the ribose region in the ATP-binding motif of ROCK (Nagumo et

421

al., 2000). It reportedly induced substantial decrease in cellular infiltration, lung

AC C

EP

TE D

M AN U

SC

RI PT

390

14

ACCEPTED MANUSCRIPT inflammatory

index,

423

responsiveness, in addition to downregulation of IL-17, IL-13, and IL-4 in lungs of OVA-

424

challenged mice. Reduction in allergic airway inflammation and mucus hypersecretion by

425

fasudil-mediated inhibition of Rho kinase was reported to occur as a result of decreased

426

expression and phosphorylation of two transcription factors that were critically implicated in

427

asthma, namely; STAT6 and NFκB (Xie et al., 2015; Taki et al., 2007).

428

Another

429

cyclohexane

430

bronchodilation in guinea pigs while maintaining minimal cardiopulmonary side effects

431

(Righetti et al., 2014; Possa et al., 2012). Many studies have shown that administration of Y-

432

27632 by either inhalation or intranasal drops prevented and/or reduced airway

433

hyperresponsiveness and pulmonary eosinophilia in different animal models of asthma (Possa

434

et al., 2012; Schaafsma et al., 2008; Henry et al., 2005; Gosens et al., 2004). Recent reports

435

revealed that co-administration of Y-27632 with corticosteroids preferentially decreased

436

exhaled NO level, bronchial inflammation, airway oxidative stress, extracellular matrix

437

remodelling, airway collagen level as well as TIMP-1 positive cells, alveolar septa

438

eosinophilia, IL-2, 8-iso-PGF2α, IFN-γ and NF-κB activity in distal parenchyma, as

439

compared to corticosteroid or Rho-kinase inhibitor monotherapy (Pigati et al., 2015). Thus,

440

Rho-kinase inhibition (either as monotherapy or combination therapy) is a potential

441

therapeutic tool for asthma management and control.

442

4.2.2. Phosphodiesterase inhibitors

443

Elevation of phosphodiesterase (PDE) activity and alteration in cAMP-PDE pathway in

444

asthmatic airway smooth muscle cells is an indication that PDEs are involved in the

445

regulation of bronchial tone, airway hyperplasia and airway remodeling (Burgess et al., 2006;

446

Trian et al., 2011). PDE4 inhibitor roflumilast, decreased airway muscular contraction,

447

eosinophilia, neutrophilia, bronchial RSV infection and MUC5AC expression (Mata et al.,

448

2013; Gauvreau et al., 2011a). Another PDE4 inhibitor, 3-[4-(3-chlorophenyl)-1-ethyl-7-

449

methyl-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl] propanoic acid (ASP3258), was reported to

450

have ameliorated airway eosinophilia in a chronic murine asthma model (Kobayashi et al.,

451

2012). Repeated doses of roflumilast (a potent oral PDE4 inhibitor that is well known for its

452

anti-inflammatory property and treatment of chronic obstructive pulmonary disorder)

453

decreased sputum eosinophilia, FeNO, leukotriene (LTE4), improved FEV1 and attenuated

454

exercise-induced asthma (Bardin et al., 2015). Although, roflumilast is considered well

selective

goblet

cell

Rho-kinase

inhibitor,

(Y-27632),

MUC5AC

expression,

and

airway

(+)-(R)-trans–4-(1-aminoethyl)-N-(4-pyridyl)

a

pyridine

derivative,

reportedly

induced

AC C

EP

TE D

M AN U

SC

carboxamide

hyperplasia,

RI PT

422

15

ACCEPTED MANUSCRIPT tolerated by most asthmatics, few unwanted adverse effects majority of which are related to

456

gastrointestinal disturbances (such as nausea and diarrhoea), insomnia and headache were

457

reported (Bardin et al., 2015; Parikh and Chakraborti, 2016; Chervinsky et al., 2015; Fabbri

458

et al., 2009).

459

In addition to induction of rapid and sustained bronchodilation among asthmatics, reduction

460

in sputum cell titre was recorded in lipopolysaccharide challenged healthy volunteers that

461

were given RPL554, a novel inhalable PDE3/4 inhibitor induced (Franciosi et al., 2013).

462

Unlike most other PDE inhibitors, the safety profile of RPL554 was within the tolerable range

463

and did not produce GI effects often associated with most PDE inhibitors (Franciosi et al.,

464

2013).

465

Xanthines were formally used in asthma treatment, however, considering their non-selectivity

466

in PDE inhibition, narrow therapeutic window, severe side effects (such as cardiac

467

arrhythmias, seizures, nausea, vomiting, and headaches) and following the discovery of better

468

bronchodilators (such as β2 agonists and muscarinic receptor antagonists), the use of

469

xanthines

470

Theophylline (dimethylxanthine) was commonly used as bronchodilator when administered at

471

relatively high dose. However, recent advances have shown that it possesses anti-

472

inflammatory effects in asthma when administered at lower doses (Barnes, 2013).

473

Theophylline molecularly induces bronchodilation by inhibition of PDE3, while it exerts its

474

anti-inflammatory effect by PDE4 inhibition and concomitant upregulation of histone

475

deacetylase-2 (HDAC-2), allowing deactivation of asthma activated inflammatory genes

476

(Barnes, 2013). HDAC-2 is critically lowered in steroid resistant severe asthmatics. The

477

upregulation of HDAC2 by theophylline reverses corticosteroid resistance in steroid

478

insensitive asthma endotype (To et al., 2010). Theophylline mediates HDAC-2 increase is

479

achievable through inhibition of phosphoinositide-3-kinase-δ (PI3Kδ) which is activated by

480

oxidative stress (To et al., 2010; Tobinick, 2009). The co-administration of theophylline with

481

salmeterol/fluticasone propionate combination to asthma patients significantly reduced

482

frequency and number of patients with asthma exacerbations, improved the forced expiratory

483

flow, decreased sputim eosinophil cationic protein concentration and generally improved

484

airway inflammation (Nie et al., 2013). Similarly, an improvement in asthma symptoms was

485

reported in asthmatic smokers that are nonresponsive to ICS following low theophylline dose

486

(Spears et al., 2009). These therefore indicate the antiasthmatic potential of theophylline,

487

especially in control of steroid resistant endotype.

asthma

therapy

gradually

faded

out

(Page,

2014).

AC C

EP

TE D

in

M AN U

SC

RI PT

455

16

ACCEPTED MANUSCRIPT 4.2.3. iNOS and Arginase Inhibition

489

The enzymatic breakdown of the guanidine group of L-arginine to release of NO and L-

490

citrulline results in generation of nitric oxide, a critical marker of airway inflammation

491

(Verini et al., 2010). This process is catalysed by nitric oxide synthase (NOS). Among the

492

three basic types of NOS, inducible nitric oxide synthase (iNOS) is overexpressed

493

inflammatory conditions. Conversely, NO from iNOS influences localized vasodilatation,

494

plasma extravasation, mucus hypersecretion, promotes eosinophilic homing and consequent

495

activation of Th2 cells, thereby aggravating airway inflammation (Prado et al., 2011; Reis et

496

al., 2012; Souza et al., 2013). Virtually, all cells in the respiratory tract of asthmatics exhibits

497

intensified expression of iNOS, which is correlated with perpetuating and amplified airway

498

inflammation in asthma (Prado et al., 2014). Potent partial and full inhibitors of iNOS

499

effectively decreased the FeNO among asthmatics (Singh et al., 2007). Nevertheless,

500

GW274150, a selective iNOS inhibitor did not effectively ameliorate airway reactivity and

501

inflammation (Singh et al., 2007). The non-desired response observed in upper airway may

502

be due to the paradoxically opposing functions of NO in upper and lower airway. L-NG-

503

Nitroarginine Methyl Ester (L-NAME), a commercially available precursor of NOS inhibitor,

504

was found to increase airway resistance, compliance and elastance, while it decreased both

505

properties (resistance, elastance and compliance) in lung parenchyma following its bio-

506

activation to L-NG-Nitroarginine; NG-nitro-L-Arginine (L-NNA), a non-specific NOS

507

inhibitor. This provide support to the fact that NO is a potent bronchodilator of proximal

508

airways (by acting through the NANC pathways) and as constrictor of distal pulmonary

509

parenchyma. Acute L-NAME therapy was also found to decreased airway eosinophilia (Prado

510

et al., 2005). Alternative treatment with specific iNOS inhibitor (e.g. 1400-W) provided better

511

control of hyperresponsiveness and airway inflammation in the same murine model used

512

(Starling et al., 2009; Marques et al., 2012; Souza et al., 2013).

513

Apart from the NO metabolite of L-arginine produced through the NOS pathway, arginase

514

catabolic cascade appears to play role in development and progression of asthma (Vonk et al.,

515

2010). However unlike NOS, the activity of arginases does not lead to the production of NO,

516

it catabolizes L-arginine to L-ornithine and urea (Ricciardolo et al., 2004). The L-ornithine

517

(product of arginase activity) is further catabolized into polyamines by ornithine

518

decarboxylase. These L-ornithine–derived polyamines are the prime agents through which

519

arginases contribute to AHR. They promote collagen synthesis and cellular proliferation

520

thereby contributing to asthma related airway remodelling (Prado et al., 2014). Moreover,

AC C

EP

TE D

M AN U

SC

RI PT

488

17

ACCEPTED MANUSCRIPT plasma polyamine surge was reported in asthmatics (North et al., 2013). Up-regulation of

522

arginase further complicates AHR by competitively upregulating pro-inflammatory

523

peroxynitrite release due to iNOS activity (Maarsingh et al., 2009). Studies have shown high

524

airway activity and expression of arginases (ARG1 and ARG2) following allergen challenge

525

in mouse asthma models (Vonk et al., 2010). 2(S)-amino-6-boronohexanoic acid, a selective

526

arginase inhibitor, was reported to reduce bronchial hypersensitivity to allergens, prevents

527

airway obstruction and reversed airway inflammation in allergen challenged guinea pigs

528

(Maarsingh et al., 2008). Additionally, S-(2-boronoethyl)-L-cysteine, another potent arginase

529

inhibitor, was reported to have decreased AHR in murine models of allergic asthma (North et

530

al., 2009). Likewise, AHR was reversed in murine model of IL-13-induced AHR by RNA

531

interference intervention against ARG1 (Yang et al., 2006). Thus, the application of specific

532

ARG inhibitors may supposedly be a promising intervention for successful amelioration of

533

asthma symptoms (figure 3).

535 536 537 538 539 540 541 542

M AN U

AC C

EP

TE D

534

SC

RI PT

521

Figure 3: Control of airway remodelling. Ca2+ signalling through calcium sensing receptor (CaSR) and Rho kinase pathways appear critical for asthma control. Inhibition of CaSR by calcilytics reverses bronchoconstriction by deactivating downstream signalling pathways such as the MAPK and the Rho/Rac/Cdc42 pathways. Direct inhibition of Rho kinases permits the dephosphorylation of myosin light chain (MLC) by active myosin phosphatase thereby inducing smooth muscle relaxation. Polyamine and proline induced bronchoconstriction and goblet metaplasia can be averted by inhibiting arginase activity. ODC: ornithine decarboxylase, OAT: Ornithine acetyltransferase, NOS: Nitric oxide synthase.

543 18

ACCEPTED MANUSCRIPT 4.3.

Biologics

545

Mast cells, bronchial epithelium and macrophages are important sources of cytokines,

546

chemokines, and a number of chemical mediators involved in asthma progression (Desai and

547

Brightling, 2009). Targeting individual cytokines that play dominant roles in the

548

pathophysiology of asthma may provide better biologics for prophylaxis and/or treatment of

549

asthma. So far, some few antibodies targeting cytokines are at developmental stages (see table

550

1).

551

Targeting of cytokine effect/activity can be achieved directly by cytokine targeting or

552

indirectly through cytokine receptor modulation. IL-13 is central to development of AHR in

553

asthma and its effect is mediated through its binding to low affinity IL-13 receptor alpha1

554

(IL-13Rα1) and IL-4 receptor alpha (IL-4Rα) complexes (IL-13Rα1/ IL-4Rα complex).

555

While IL-4 also exerts its effect through IL-4Rα. Dupilumab, a human monoclonal antibody

556

against IL-4α/IL-13α receptor complex, inhibited the downstream signalling events induced

557

by IL-4 and IL-13 by binding to the alpha subunit of the IL-4 receptor of the complexes

558

(Hambly and Nair, 2014; Vatrella et al., 2013; Wenzel et al., 2013b; Prado et al., 2014). Add

559

on therapy of dupilumab/ICS/LABA combination was recently reported to effectively

560

improve FEV1, patient’s quality of life and decreased recurrent asthma exacerbations in

561

patients with uncontrolled persistent asthma regardless of baseline blood eosinophil level.

562

Additionally, the antibody has the tendency of ameliorating comorbidities associated with

563

uncontrolled persistent asthma (e.g nasal polyps and acute dermatitis) (Wenzel et al., 2016).

564

Likewise, pitrakinra, a dual IL-4/IL-13 antagonist was evaluated for potential asthma therapy

565

and it was found to dampen the progression of early and late asthmatic response in allergen

566

challenge models (Antoniu, 2010; Wenzel et al., 2007). Pitrakinra competitively inhibited IL-

567

4Rα activity to impede IL-4 and IL-13 activities. It also significantly improved asthma

568

symptoms and decreased exacerbations in patients with airway eosinophilia, nevertheless,

569

phase IIb trial failed to demonstrate clinical efficacy of the intervention in the overall study

570

population (Hambly and Nair, 2014).

571

Effort to target IL-13 alone using lebrikizumab (a humanized anti-IL-13 antibody) suggests

572

asthma endotype specificity in IL-13 dependent asthma treatment. Significant improvement in

573

lung function was reported following treatment with lebrikizumab in patients with high serum

574

periostin (an indicator of eosinophilia in asthmatics) and high fractional exhaled nitric oxide

575

(FeNO) levels (Corren, et al., 2011; Jia et al.,2012; Noonan et al., 2013). Serum periostin

AC C

EP

TE D

M AN U

SC

RI PT

544

19

ACCEPTED MANUSCRIPT level can be used as a critical determinant of lebrikizumab efficacy. Recent randomised,

577

double-blind, placebo-controlled trial revealed reduction in asthma exacerbations rate and

578

improvement in FEV1 among periostin-high patients with moderate-to-severe asthma than in

579

the periostin-low patients following 24 weeks of treatment with different doses of

580

lebrikizumab (37.5, 125, 250 mg every four weeks) (Hanania et al., 2015). Tralokinumab

581

administration dose dependently improved lung function (as measured by FEV1) of patients

582

with moderate-severe asthma. Safety and toxicity studies revealed no serious adverse effect in

583

its phase II trial (Piper et al., 2013; Hanania et al., 2015).

584

An IL-13Rα1 isomer, IL-13 receptor alpha2 (IL-13Rα2) possesses high affinity to IL-13 as

585

compared to IL-13R α1. However, it was proposed to perform a non-signalling decoy role in

586

IL-13 functionality. Upregulation of its soluble isomer (sIL-13Rα2) reportedly inhibited IL-

587

13 induced airway inflammation in murine asthma models (Daines et al., 2006; Chen et al.,

588

2013). Conversely, mouse has both its soluble (sIL-13Rα2) and membrane bound (memIL-

589

13Rα2) forms of IL-13Rα2, humans only have the memIL-13Rα2 form. This may contribute

590

to failure in translation of preclinical trials involving IL-13Rα2 immunomodulation for

591

control of human asthma (Chen et al., 2013).

592

Considering the role of IL-5 in airway eosinophil homing, as well stimulation of mediator

593

release from eosinophil and its effect on tissue survival (Garcia et al., 2013), monoclonal

594

antibodies against interleukin 5 (anti-IL-5) were tried for use in asthma. Anti-IL-5

595

monoclonal antibodies such as mepolizumab and reslizumab revealed beneficial effect in

596

management of persistent airways eosinophilia among corticosteroid resistant subjects. In a

597

randomized, double blind and double dummy trial, reduction in rate of asthma exacerbations

598

and sputum eosinophilia was recorded upon repeated administration of mepolizumab to

599

insensitive asthma patients (Ortega et al., 2014).

600

mepolizumab is an exciting advancement especially for patients with uncontrolled severe

601

eosinophilic asthma because it is safe and effective option that could replace oral

602

corticosteroids (Bel et al 2014; Haldar et al., 2009; Nair et al., 2009; Pavord et al., 2012).

603

According to recent trials on reslizumab, the antibody exhibited tolerable safety profile and

604

improved FEV1, forced vital capacity (FVC), forced expiratory flow (FEF), asthma control,

605

patient’s quality of life and rescued SABA use in uncontrolled asthma with eosinophilic

606

endotype (Corren et al., 2016; Markham, 2016; Castro et al., 2015). Equally, benralizumab

607

(MEDI-563), an afucosylated monoclonal antibody against IL-5Rα, was reported to reduce

608

peripheral eosinophils levels in moderate asthma patients and sustained its effect for about 8–

AC C

EP

TE D

M AN U

SC

RI PT

576

20

The corticosteroid sparing effect of

ACCEPTED MANUSCRIPT 12 weeks through antibody dependent cell-mediated cytotoxicity (Ghazi et al., 2012; Busse et

610

al., 2010; Kolbeck, et al., 2010).

611

Th17 lymphocytes have demonstrated the ability to recruit both eosinophils and neutrophils

612

to the airway through IL-17A and IL-17F release (Tesmer et al., 2008). Although IL-17

613

targeting using secukinumab, and ixekizumab seems promising, there are few clinical trial

614

reports on the use of the antibodies in asthma. However, brodalumab, a monoclonal antibody

615

that targets IL-17RA, produced clinically meaningful responses in moderate to severe

616

asthmatic patients with high bronchodilator reversibility (Busse et al., 2013). Considering the

617

equivocal nature of available data on use of brodalumab in asthma control (Bauer et al.,

618

2015), further studies on its effect on different asthma sub-populations is highly

619

recommended.

620

Tumor necrotic factor-alpha (TNF-α) is mainly secreted by lymphocytes, mast cells, and

621

macrophages. It promotes bronchial hyperresponsiveness and sputum neutrophilia, this make

622

it an attractive target for severe asthma (Wenzel et al., 2009). A number of anti-TNF-α agents

623

exist in the market and they were proven effective in some inflammatory diseases such as

624

rheumatoid arthritis and Crohn disease. It is apparent that the use of inhibitors of TNF-α may

625

be valuable in reversal of asthma symptoms. Etanercept (anti TNF-α agent) showed short-

626

term and modest efficacy for severe and mild asthma respectively (Antoniu, 2009). Slight

627

reduction in number of exacerbations and improvement in wheezing was observed in patients

628

with moderate uncontrolled asthma originally on ICS monotherapy following infliximab or

629

adalimumab administration (Erin et al., 2006; Stoll et al., 2009). Upon withdrawal of TNF-α

630

inhibition, wheezing triumphed (Stoll et al., 2009). Safety concerns may limit the use of anti-

631

TNF-α in asthma therapy. Substantial adverse reactions were observed without achieving

632

fundamental objectives of reducing asthma exacerbations and improving FEV1 among

633

subjects (see table 2). Golimumab lowered risk of asthma exacerbations, nevertheless, its trial

634

on severe persistent asthma ended at phase II due to incidences of malignancies and

635

infections such as pneumonia (Wenzel et al., 2009). Given the role of TNF- α in severe,

636

refractory, or steroid-resistant asthma, future studies on the use of this anti-TNF- are needed

637

to identify whether the long-term risk-benefit profile favours use in asthma.

638

The results of trials involving agents targeting IL-4, IL-13, IL-9, IL-12, IL-10, interferon-γ,

639

granulocyte-macrophage colony-stimulating factor, and even Th17 cells are anticipated

640

(Hansbro et al., 2011; Desai and Brightling, 2009). As understanding of the cytokine

AC C

EP

TE D

M AN U

SC

RI PT

609

21

ACCEPTED MANUSCRIPT 641

networks continues to evolve, so too will the potential targets for antiasthma therapy.

642

4.4.

643

Airway inflammation in asthma is associated with several transcription factors such as the

644

NFkB, nuclear factor of activated T-cells (NF-AT) and glucocorticoid receptor (GR). Cyclic

645

AMP response element binding protein (CREB), activator protein-1 (AP-1), peroxisome

646

proliferator-activated

647

CCAAT/enhancer binding protein (C/EBP), signal transducer and activator of transcription 6

648

(STAT6), GATA-3, doublesex and mab-3 related transcription factor 1 (DMRT1) and nuclear

649

factor erythroid 2-related factor 2 (Nrf2) were also implicated in pathogenesis of asthma

650

(Roth and Black, 2006). Although, most transcription factors play important role in tissue and

651

organ homeostasis, modulation of some of the transcription factors (especially GATA-3,

652

NFkB, Nrf2, NF-AT and STAT6) appeared to be a valid therapeutic approach to control of

653

asthma symptoms. Control of lung inflammatory responses is achievable by application of

654

decoy or antisense oligonucleotides specific for targeted airway transcription factor of

655

interest. Nevertheless, long term suppression or overexpression of these transcription factors

656

could lead to wide array of adverse effects, thus, cell type specific delivery of inhibitors or

657

activators of target factors is critical to obtaining minimal adverse effect that may arise from

658

this approach (Roth and Black, 2006; Haley et al., 2011; Schieck et al., 2016)

659

The characteristic Th2 molecular endotype of allergic asthma is controlled by a zinc finger

660

transcription factor GATA3, which is essential for Th2-cell differentiation and immune

661

pathway activation (Wenzel, 2012). GATA-3 is reportedly overexpressed in broncholaveolar

662

lavage and lung biopsies from severe asthma patients (Bergqvist et al., 2013). Thus,

663

interventions that target GATA3 and its downstream effectors were suggested for treatment

664

of asthma (Holgate, 2012). Afzelin, a flavonol glycoside, was reported to suppress airway

665

inflammation, eosinophil infiltration, airway hyper-responsiveness and BALF Th2 cytokines

666

in murine asthma model by inhibition of GATA-3 activity and increasing T-bet/GATA-3

667

ratio. Considering the role of T-bet transcription factor in regulation of Th1 differentiation

668

and activation, the increase in T-bet/GATA-3 ratio supposedly restored Th1/Th2 balance

669

(Jenner et al., 2009; Zhou and Nie, 2015). In another preclinical study, GATA-3 specific

670

catalytically active, single stranded, synthetic antisense DNA enzyme (DNAzyme) hgd40

671

containing SB010 (active drug component) was tested for efficacy in reduction of Th2

672

cytokine level via inhibition of GATA-3 mRNA and protein level (Turowska et al., 2013; Sel

(PPAR),

runt-related

transcription

factors (RUNX),

AC C

EP

TE D

M AN U

SC

receptor

RI PT

Targeting Transcription Factors

22

ACCEPTED MANUSCRIPT et al., 2008). The results of the preclinical tests showed remarkable improvement in asthma

674

symptoms with no observable off-target effect therefore allowing for further clinical trial of

675

the DNAzyme (Homburg et al., 2015; Fuhst et al., 2013). In a randomized, double-blind,

676

placebo-controlled, multicenter clinical trial, 10mg of aerosolized SB010 or placebo was

677

administered to asthma patients once daily for 28 days, and a significant improvement in lung

678

function was reported. Furthermore, the intervention attenuated allergen-induced sputum

679

eosinophilia, reduced sputum tryptase and negated plasma IL-5 levels (Krug et al., 2015).

680

GATA-3 activity is linked to few other asthma related transcription factor signalling such as

681

the JAK/STAT6 signalling pathway. Boswellic acid, a specific and non-redox inhibitor of 5-

682

lipoxygenase enzyme, was revealed to suppress allergic airway inflammation by indirectly

683

abrogating GATA3 expression through inhibition of IL-4 dependent pSTAT6 activity (Liu et

684

al, 2015). Furthermore, in an effort to unveil the determinants of asthma development in

685

atopic subjects with or without asthma and identify asthma associated drug targets in HDM-

686

specific T-helper memory responses, a differential gene network analysis revealed that most

687

asthma-associated genes were enriched with targets of STAT6 signaling (Troy et al., 2016).

688

AS1517499 and YM-341619 are small-molecule inhibitors of STAT6 activity, they were

689

found to modulate asthma symptoms by selectively inhibiting Th2 differentiation (Vale,

690

2016; Nagashima et al., 2009). In another development, Ursolic acid reportedly decreased

691

airway eosinophilia, antigen-induced Penh, pulmonary inflammation, pro-inflammatory

692

cytokine production as well as serum antigen-specific IgE by increasing PPARγ expression

693

and decreasing GATA-3/STAT6 expression through NF-kB pathway modulation (Kim et al.,

694

2013). After all, previous studies revealed that ursolic acid suppresses NF-kB activation by

695

inhibition of IκBα kinase and p65 phosphorylation (Shishodia et al., 2003 and Zeng et al.,

696

2012). More recently, integromics studies revealed GRB2-associated binding protein 1

697

(GAB1) as a novel modulator of asthma due to its NF-kB regulatory activity (Sharma et al.,

698

2015).

699

Control of STAT6 activity can be achieved by JAK modulation. Preclinical studies have

700

validated the suitability of JAKs as a therapeutic target for asthma. A pan-JAK inhibitor,

701

Pyridone 6 (P6), suppressed asthmatic symptoms through Th2 inhibition (Matsunaga et al.,

702

2011). Tofacitinib (CP-690,550), a STAT-6 and pan-JAK inhibitor was suggested to reduce

703

eosinophilia, eotaxins and IL-13 by inhibition of IL-4 signaling pathway in murine model of

704

pulmonary eosinophilia (Kudlacz et al., 2008). However, it is established that tofacitinib

705

enhances pulmonary bacterial growth in models of latent tuberculosis infection, thereby

AC C

EP

TE D

M AN U

SC

RI PT

673

23

ACCEPTED MANUSCRIPT posing the risk of reactivation of latent form of tuberculosis in asthma patients with

707

comorbidities (Maiga et al., 2012). It is for this reason that development of safer selective

708

JAK inhibitors was proposed. R256, an inhalable selective JAK/1/3 inhibitor proved to be a

709

safer option over inhibitors of pan-JAKs, due to their selective inhibition of Th2

710

differentiation without affecting Th1 and/or Th17 differentiation (Vale, 2016; Ashino et al.,

711

2014). Figure 4 illustrates the targeted JAK-STAT pathway inhibition as it relates to GATA3

712

and CRTh2 signalling pathways in asthma.

AC C

EP

TE D

M AN U

SC

RI PT

706

713 714 715 716 717 718 719

Figure 4: Th2 surface receptor directed therapy. Prevention of airway allergic inflammatory cell migration is achievable through modulation of key surface receptors and downstream targets such as intracellular receptors, enzymes and transcription factors. Interaction of Th2 cytokine such as IL-4 with its receptors (IL-4R) results in activation of the JAK/STAT6 pathway that leads to activation and production of GATA3 protein. Prostaglandin D2 (PGD2) released from mast cells interact with CRTh2 to induce inhibition of adenylyl cyclase (AC) and consequent decrease in intracellular cyclic adenosine monophosphate (cAMP) levels through Gi

24

ACCEPTED MANUSCRIPT 720 721 722 723 724 725 726 727

protein activation. This also results in release of βɣ subunit of the G protein, which directly stimulates phospholipase Cβ (PLCβ) isoforms. PLBβ breakdown phosphatidylinositol-4,5-bisphosphate (PIP2) to produce inositol triphosphate (IP3) which mobilizes Ca2+ from endoplasmic-reticulum stores. The Ca2+ activates calcineurin, which then dephosphorylate NFAT to its active state (NFATc). Transcriptional activation of both GATA3 and NFAT exert a net effect of increased Th2 cytokine production, thereby providing the cytokine milieu that is required for aggravated allergic asthma response in a vicious manner. Inhibition of IL-4R, CRTh2, JAK, STAT-6, GATA3, and cytokine inhibition showed remarkable reduction in chemotaxis, goblet metaplasia, Th1/Th2 imbalance, bronchoconstriction and hyperresponsiveness.

RI PT

728

5.

Conclusion

730

Acute asthma exacerbations are arbitrary, unsettling and detrimental to patient’s quality of

731

life. Prevention of exacerbations through adherence to controller drug regimens, avoidance of

732

exposure to known triggers, annual immunization against influenza and pneumococcal

733

polysaccharide (for asthmatics aged 19 years or older) are imperative. This review has

734

highlighted some potential drug targets for the treatment of asthma. However, few more

735

potential candidates for asthma therapy including calcium sensing receptor (CaSR)

736

antagonist, phosphatidylinositol 3-kinase (PI3K) inhibition through direct inhibitors or

737

indirect inhibitors (e.g. theophylline should also be considered. Asthma symptoms are

738

infrequent, short-lived and mild, occasional administration of a quick-acting bronchodilator to

739

reverse bronchoconstriction is an acceptable approach. However, as symptoms become more

740

frequent or more severe, the emphasis changes to prevention of symptoms (and of asthmatic

741

attacks). Inhaled corticosteroid reduces the frequency of episodic bronchoconstriction and

742

lessens the risk of asthmatic attacks through its anti-inflammatory activities. In low-to-

743

moderate doses, corticosteroids administered by inhalation are safe for long-term use, even in

744

young children.

745

Whilst considerable advancement in the understanding pathophysiology of asthma promoted

746

the discovery of novel therapeutic targets, only few are successfully translated into clinical

747

application. The heterogeneous nature of asthma calls for multifaceted approach to its

748

treatment, thus, further investigations into potential therapies are paramount. On a general

749

note, the pleitrophic nature of kinases and their ability to modulate wide array of cellular

750

events increases risk of adverse effects (e.g haematological disorders and cardiovascular

751

complications) as observed in various protein kinase inhibitors currently used for

752

management of cancer. It is therefore pertinent to apply caution while considering kinase

753

inhibitors for asthma therapy. This may include ensuring minimal side effects by

754

administering low doses of inhalable preparations.

AC C

EP

TE D

M AN U

SC

729

25

ACCEPTED MANUSCRIPT Most of the candidate drugs discussed in this article have shown potential to fit into stepwise

756

asthma management strategy taking into account the patient’s response to available

757

medication, onset of the disorder (childhood or adult onset asthma), effectiveness and safety

758

of the candidate drug with respect to patient’s asthma endotytpe. Administration of TLR

759

agonist is suitable during ICS step down treatment in patients on medium to high dose ICS

760

whose asthma has been successfully controlled. this is because TLR agonist may contribute to

761

sustaining asthma control during such steroid reduction regime, thereby preventing sudden

762

relapse. The use of CRTh2 antagonist may best be indicated at stages where patients respond

763

poorly to ICS monotherapy (step 2 to 4 of GINA treatment guidelines) and in ICS naïve

764

asthmatics as add on therapy to reliever medications. Additionally, CRTh2 targeted therapies

765

may be more effective in Th2-high asthma subphenotype. The combined bronchodilatory and

766

anti-inflammatory potential of dual PDE3/4 inhibitors may enhance its use as LABA

767

replacement or as add-on therapy to reduced dose of ICS/LABAs controller medication,

768

thereby complementing safety concerns related to long term use of LABAs. While most

769

biologics based interventions will best be administered to patients with uncontrolled

770

persistent asthma that failed to respond to standard therapy. This may include the

771

administration of such biologics as add on therapy to ICS or oral corticosteroids (OCS) to

772

relieve severe recurrent asthma exacerbations

773

Acknowledgment

774

This work was supported by National Key Economic Area Research Grant Scheme

775

(NRGS/NH1014D026), provided by the Ministry of Agriculture, Malaysia.

AC C

EP

TE D

M AN U

SC

RI PT

755

26

ACCEPTED MANUSCRIPT

Table 1: Summary of emerging molecular targets for the treatment of asthma and developmental stage

Drug name

Highest Development phase

Receptor Modulators

CRTH2 antagonist

OC000459

2

CRTH2 antagonist

BI-671800

2

CRTH2 antagonist

ARRY-502

CCR3 receptor blocker

TPI ASM8

TLR-3 antagonists

CNTO 3157

urotensin II receptor antagonist

GSK1440115

Oxagen Ltd

Barnes et al., 2012 NCT01057927 Hall et al., 2015 NCT01103349

2

Array BioPharma Inc.

Wenzel et al., 2014 NCT01561690

2

Pharmaxis,

Gauvreau et al., 2008; Gauvreau et al., 2011b NCT00264966/ NCT01158898

1

Janssen Research and Development, LLC

NCT01704040

1

GlaxoSmithKline

Portnoy et al., 2013 NCT01202214

M AN U

TE D

Reference/NCT number

Boehringer Ingelheim

AC C

EP

Sponsor

RI PT

Mechanism of action

SC

Therapeutic Class

27

ACCEPTED MANUSCRIPT

Mechanism of action

Drug name

Highest Development phase

Enzymatic Targets

PI3K/mTOR inhibitor

GSK2269557

1

PDE4 inhibitor

GSK256066

2

PDE4 inhibitor

CHF 6001 (inhaled)

2

PDE4 inhibitor

roflumilast

PDE4/3 inhibitor

RPL 554

Leukotriene A4 hydrolase inhibitor

JNJ-40929837

IL-5Rα mAb

IL-5 mAb IL-5 mAb

Reference/NCT number

Stark et al., 2015 NCT02294734

GlaxoSmithKline,

Singh et al., 2010 NCT00380354

Chiesi Farmaceutici S.p.A.

Matera et al., 2014 NCT01689571

2

Takeda; Nycomed and Pfizer

NCT01765192/ NCT01365533

2

Verona Pharma

NCT02427165

2

Johnson and Johnson Pharmaceutical Research and Development, L.L.C.

Barchuk et al., 2014 (NCT01241422)

Benralizumab

2/3

AstraZeneca

Nowak et al., 2015 NCT01914757

Reslizumab

3

Teva Pharmaceutical Industries

Castro et al., 2015 NCT01287039/ NCT01285323

Mepolizumab

3

GlaxoSmithKline

Ortega et al., 2014 NCT01691521

M AN U

TE D

EP

SC

GlaxoSmithKline

AC C

Anti-interleukin agents

Sponsor

RI PT

Therapeutic Class

28

ACCEPTED MANUSCRIPT

Highest Development phase

IL-13 mAb

Lebrikizumab

2

IL-13 mAb

Tralokinumab

3

IL-4Rα mAb

Dupilumab

2

IL-17RA

Brodalumab

1

29

Reference/NCT number

Genentech

Hanania et al., 2015 NCT01545440/ NCT01545453

AstraZeneca

Brightling et al., 2015 NCT02281357

Sanofi and Regeneron Pharmaceuticals Inc.

Wang et al., 2015 NCT01854047

Amgen Kyowa Hakko Kirin Company Limited and AstraZeneca

NCT01902290

Sterna Biologicals GmbH and Co. KG

Homburg et al., 2015 NCT01743768

M AN U

TE D

SB010

2 (trial terminated during Phase 2)

EP

GATA-3 Transcription DNAzyme Factor NCT: National Clinical Trial

Sponsor

RI PT

Drug name

SC

Mechanism of action

AC C

Therapeutic Class

ACCEPTED MANUSCRIPT

Table 2: Summary of adverse effects of selected anti-asthma candidate drugs as revealed by clinical trials

Mechanism of action

Route of administration

Adverse effects

Reference/Trial number

CYT003

TLR-9 Agonist

Subcutaneous

Injection site reactions, Pyrexia Nasopharyngitis, Head ache.

Casale et al., 2015, NCT01673672

QbG10

TLR-9 Agonist

Subcutaneous

BI 671800

CRTH2 antagonist

Oral

H4R antagonist

SC M AN U

Oral

Oral

Toxic hepatitis in patients with prior elevated hepatic transaminases♯

Hall et al., 2015, NCT01092148 and NCT01103349

Headache♯

Sidharta et al., 2014, EudraCT number 2006-006777-25 Diamant et al., 2014, EudraCT number 2008-001209-41

Drug-induced agranulocytosis*

Murata et al., 2015, NCT01493882, NCT01497119

Headache♯, nausea, diarrhoea, asthenia and dyspepsia

Bateman et al., 2016, NCT01765192. Chervinsky et al., 2015, NCT00073177, NCT00076076, NCT00163527 Gauvreau et al., 2011a, NCT01365533



Roflumilast

PDE-4 inhibitor

AC C

EP

JNJ 39758979

CRTH2 antagonist

Beeh, et al., 2013, NCT00890734

Injection site reaction

TE D

Setipiprant,

RI PT

Candidate drug

Oral

30

ACCEPTED MANUSCRIPT

Mechanism of action

Route of administration

Adverse effects

RI PT

Candidate drug

Reference/Trial number Franciosi et al., 2013, EudraCT, numbers 2008005048-17, 2011-001698-22, 2010-023573-18, and 2012000742-34.

Dual PDE-3 and PDE-4 inhibitor

Inhalation

Headache♯, Palpitations, Paraesthesia. Rhinorrhoea, Somnolence

Dupilumab

IL-4Rα antagonist

Subcutaneous

Upper respiratory tract infection, Headache, Injection-site erythema, Sinusitis, Influenza

Wenzel et al., 2016, NCT01854047, EudraCT number 2013-000856-16

Lebrikizumab

IL-13 inhibitor

Subcutaneous

Injection site reaction, Injection-site erythema, Upper respiratory tract infection

Hanania et al., 2015, NCT01545440, NCT01545453

Headache, Injection-site reaction, nasopharyngitis, cough, arthralgia

Pouliquen et al., 2015, NCT01366521 Ortega et al., 2014, NCT01691521

Intravenous/Subcutaneous

Upper respiratory tract infections, headache, nasopharyngitis myalgia, oropharyngeal pain, anaphylaxis and sinusitis

Corren et al., 2016, NCT01508936 Castro et al., 2015 NCT01287039/ NCT01285323 Markham, 2016, NCT02452190, NCT02501629, NCT02559791

Subcutaneous

Injection site erythema, upper respiratory tract infection, sinusitis, nasopharyngitis and oral candidiasis

Busse et al., 2013, NCT01199289

M AN U

TE D

IL-5 inhibitor

Intravenous/Subcutaneous

IL-5 receptor antagonist

Brodalumab

IL-17 receptorA antagonist

AC C

Reslizumab

EP

Mepolizumab

SC

RPL554

31

ACCEPTED MANUSCRIPT

Adverse effects

Reference/Trial number

Subcutaneous

Injection-site reaction, upper respiratory tract infections, headache, migraine, sinusitis and asthenia

Holgate et al., 2011, NCT00141791

Pneumonia*♯, cellulitis, sepsis, upper respiratory tract infections, chest pain, sinusitis, nausea, and injection-site erythema

Wenzel et al., 2009, NCT00207740

Golimumab

TNF-α inhibitor

Subcutaneous

SB010

GATA-3 DNAzyme

Inhalation

RI PT

TNF-α inhibitor

Route of administration

SC

Etanercept

Mechanism of action

M AN U

Candidate drug

Mild oropharyngeal pain, back pain and bronchospasm♯

Homburg et al., 2015, NCT01743768, NCT01577953

AC C

EP

TE D

*Serious Adverse event, ♯Treatment emergent adverse event, EudraCT: European Clinical Trials Database, NCT: National Clinical Trial

32

ACCEPTED MANUSCRIPT References Abramson, M. J., Walters, J., and Walters, E. H. (2003). Adverse effects of βagonists. American Journal of Respiratory Medicine, 2 (4), 287-297. http://dx.doi.org/10.1007/bf03256657

RI PT

Aguilera-Aguirre, L., Bacsi, A., Saavedra-Molina, A., Kurosky, A., Sur, S., and Boldogh, I. (2009). Mitochondrial dysfunction increases allergic airway inflammation. The Journal of Immunology, 183 (8), 5379-5387. http://dx.doi.org/10.4049/jimmunol.0900228 Akdis, C. A., and Akdis, M. (2015). Mechanisms of allergen-specific immunotherapy and immune tolerance to allergens. The World Allergy Organization Journal, 8(1), 17. http://doi.org/10.1186/s40413-015-0063-2

SC

Alam, A. (2014). Serine Proteases of Malaria Parasite Plasmodium falciparum: Potential as Antimalarial Drug Targets. Interdisciplinary Perspectives on Infectious Diseases, 2014, 453186. http://doi.org/10.1155/2014/453186

M AN U

Al-Hajjaj, M. S. (2011). Difficult-to-treat asthma, is it really difficult? Annals of Thoracic Medicine, 6(1), 1. http://dx.doi.org/10.4103/1817-1737.39633 Ali, A. K., and Hartzema, A. G. (2012). Assessing the association between omalizumab and arteriothrombotic events through spontaneous adverse event reporting. Journal of Asthma and Allergy, 5(1), 1-9. http://dx.doi.org/10.2147/jaa.s29811 Allard, B., Bara, I., Gilbert, G., Carvalho, G., Trian, T., Ozier, A., ... and Berger, P. (2014). Protease activated receptor-2 expression and function in asthmatic bronchial smooth muscle. PloS ONE, 9 (2), e86945. http://dx.doi.org/10.1371/journal.pone.0086945

TE D

Antoniu, S. A. (2009). Cytokine Antagonists for the Treatment of Asthma. BioDrugs, 23 (4), 241-251. http://dx.doi.org/10.2165/11317130-000000000-00000

EP

Antoniu, S. A. (2010). Pitrakinra, a dual IL-4/IL-13 antagonist for the potential treatment of asthma and eczema. Current Opinion in Investigational Drugs (London, England: 2000), 11 (11), 1286-1294. http://dx.doi.org/10.1517/14712598.2010.524203

AC C

Ashino, S., Takeda, K., Li, H., Taylor, V., Joetham, A., Pine, P. R., and Gelfand, E. W. (2014). Janus kinase 1/3 signaling pathways are key initiators of Th2 differentiation and lung allergic responses. Journal of Allergy and Clinical Immunology, 133 (4), 1162-1174. http://dx.doi.org/10.1016/j.jaci.2013.10.036 Bain, J., Plater, L., Elliott, M., Shpiro, N., Hastie, C., Mclauchlan, H., ... and Cohen, P. (2007). The selectivity of protein kinase inhibitors: a further update. The Biochemical Journal, 408 (3), 297-315. http://dx.doi.org/10.1042/bj20070797 Barchuk, W., Lambert, J., Fuhr, R., Jiang, J. Z., Bertelsen, K., Fourie, A., ... and Thurmond, R. (2014). Effects of JNJ-40929837, a leukotriene A 4 hydrolase inhibitor, in a bronchial allergen challenge model of asthma. Pulmonary Pharmacology and Therapeutics, 29 (1), 1523. http://dx.doi.org/10.1016/j.pupt.2014.06.003 Bardin, P., Kanniess, F., Gauvreau, G., Bredenbröker, D., and Rabe, K. F. (2015). Roflumilast for asthma: Efficacy findings in mechanism of action studies. Pulmonary Pharmacology and Therapeutics, 35, S4-S10. http://dx.doi.org/10.1016/j.pupt.2015.08.006

33

ACCEPTED MANUSCRIPT Barnes, N., Pavord, I., Chuchalin, A., Bell, J., Hunter, M., Lewis, T., ... and Perkins, C. M. (2012). A randomized, double‐blind, placebo‐controlled study of the CRTH2 antagonist OC000459 in moderate persistent asthma. Clinical and Experimental Allergy, 42 (1), 38-48. http://dx.doi.org/10.1111/j.1365-2222.2011.03813.x Barnes, P. J. (2006). How corticosteroids control inflammation: quintiles prize lecture 2005. British Journal of Pharmacology, 148 (3), 245-254. http://dx.doi.org/10.1038/sj.bjp.0706736

RI PT

Barnes, P. J. (2013). Theophylline. American Journal of Respiratory and Critical Care Medicine, 188 (8), 901-906. http://dx.doi.org/10.1164/rccm.201302-0388pp Barnes, P. J., Bonini, S., Seeger, W., Belvisi, M. G., Ward, B., and Holmes, A. (2015). Barriers to new drug development in respiratory disease. European Respiratory Journal, 45 (5), 1197-1207. http://dx.doi.org/10.1183/09031936.00007915

M AN U

SC

Bateman, E. D., Goehring, U. M., Richard, F., and Watz, H. (2016). Roflumilast combined with montelukast versus montelukast alone as add-on treatment in patients with moderate-tosevere asthma. Journal of Allergy and Clinical Immunology. (in press). http://dx.doi.org/10.1016/j.jaci.2015.11.035 Bauer, E., Lucier, J., and Furst, D. E. (2015). Brodalumab-an IL-17RA monoclonal antibody for psoriasis and psoriatic arthritis. Expert Opinion on Biological Therapy, 15 (6), 883-893. http://dx.doi.org/10.1517/14712598.2015.1045410

TE D

Beck, I. M., Vanden Berghe, W., Vermeulen, L., Yamamoto, K. R., Haegeman, G., and De Bosscher, K. (2009). Crosstalk in inflammation: the interplay of glucocorticoid receptorbased mechanisms and kinases and phosphatases. Endocrine Reviews, 30 (7), 830-882. http://dx.doi.org/10.1210/er.2009-0013 Beeh, K. M., Kanniess, F., Wagner, F., Schilder, C., Naudts, I., Hammann-Haenni, A., ... and Renner, W. A. (2013). The novel TLR-9 agonist QbG10 shows clinical efficacy in persistent allergic asthma. Journal of Allergy and Clinical Immunology, 131 (3), 866-874. http://dx.doi.org/10.1016/j.jaci.2012.12.1561

EP

Behera, D., and Sehgal, I. S. (2015). Bronchial asthma-issues for the developing world. The Indian Journal of Medical Research, 141 (4), 380. http://dx.doi.org/10.4103/09715916.159237

AC C

Bel, E. H., Wenzel, S. E., Thompson, P. J., Prazma, C. M., Keene, O. N., Yancey, S. W., ... and Pavord, I. D. (2014). Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. New England Journal of Medicine, 371 (13), 1189-1197. http://dx.doi.org/10.1056/nejmoa1403291 Bergqvist, A., Andersson, C. K., Hoffmann, H. J., Mori, M., Shikhagaie, M., Krohn, I. K., ... and Erjefält, J. S. (2013). Marked epithelial cell pathology and leukocyte paucity in persistently symptomatic severe asthma. American Journal of Respiratory and Critical Care Medicine, 188 (12), 1475-1477. http://dx.doi.org/10.1164/rccm.201308-1444le Biro, M., Munoz, M. A., and Weninger, W. (2014). Targeting Rho‐GTPases in immune cell migration and inflammation. British Journal of Pharmacology, 171 (24), 5491-5506. http://dx.doi.org/10.1111/bph.12658

34

ACCEPTED MANUSCRIPT Boitano, S., Flynn, A. N., Schulz, S. M., Hoffman, J., Price, T. J., and Vagner, J. (2011). Potent agonists of the protease activated receptor 2 (PAR2). Journal of Medicinal Chemistry, 54(5), 1308-1313. http://dx.doi.org/10.1021/jm1013049 Brimblecombe, R. W., Duncan, W. A. M., Durant, G. J., Ganellin, C. R., Parsons, M. E., and Black, J. W. (2010). The pharmacology of cimetidine, a new histamine H2‐receptor antagonist. British Journal of Pharmacology, 160(s1), S52-S53. http://dx.doi.org/10.1111/j.1476-5381.2010.00854.x

RI PT

Brusselle, G. G., Maes, T., and Bracke, K. R. (2013). Eosinophils in the spotlight: eosinophilic airway inflammation in nonallergic asthma. Nature M edicine, 19 (8), 977-979. http://dx.doi.org/10.1038/nm.3300

SC

Burdyga, T., Mitchell, R. W., Ragozzino, J., and Ford, L. E. (2003). Force and myosin light chain phosphorylation in dog airway smooth muscle activated in different ways. Respiratory Physiology and Neurobiology, 137 (2), 141-149. http://dx.doi.org/10.1016/s15699048(03)00143-5

M AN U

Busse, W. W., Holgate, S., Kerwin, E., Chon, Y., Feng, J., Lin, J., and Lin, S. L. (2013). Randomized, double-blind, placebo-controlled study of brodalumab, a human anti–IL-17 receptor monoclonal antibody, in moderate to severe asthma. American Journal of Respiratory and Critical Care Medicine, 188(11), 1294-1302. http://doi.org/10.1164/rccm.201212-2318OC Busse, W. W., Katial, R., Gossage, D., Sari, S., Wang, B., Kolbeck, R., ... and Molfino, N. A. (2010). Safety profile, pharmacokinetics, and biologic activity of MEDI-563, an anti–IL-5 receptor α antibody, in a phase I study of subjects with mild asthma. Journal of Allergy and Clinical Immunology, 125 (6), 1237-1244. http://dx.doi.org/10.1016/j.jaci.2010.04.005

TE D

Campbell, J. D., Kell, S. A., Kozy, H. M., Lum, J. A., Sweetwood, R., Chu, M., ... and Hessel, E. M. (2014). A limited CpG-containing oligodeoxynucleotide therapy regimen induces sustained suppression of allergic airway inflammation in mice. Thorax, 69(6), 565573. http://dx.doi.org/10.1136/thoraxjnl-2013-204605

EP

Caron, G., Delneste, Y., Roelandts, E., Duez, C., Bonnefoy, J. Y., Pestel, J., and Jeannin, P. (2001). Histamine polarizes human dendritic cells into Th2 cell-promoting effector dendritic cells. The Journal of Immunology, 167(7), 3682-3686. http://dx.doi.org/10.4049/jimmunol.167.7.3682

AC C

Casale, T. B., Cole, J., Beck, E., Vogelmeier, C. F., Willers, J., Lassen, C., ... and Wechsler, M. E. (2015). CYT003, a TLR9 agonist, in persistent allergic asthma–a randomized placebo‐ controlled Phase 2b study. Allergy, 70(9), 1160-1168. http://dx.doi.org/10.1111/all.12663 Castro, M., Zangrilli, J., Wechsler, M. E., Bateman, E. D., Brusselle, G. G., Bardin, P., ... and Korn, S. (2015). Reslizumab for inadequately controlled asthma with elevated blood eosinophil counts: results from two multicentre, parallel, double-blind, randomised, placebocontrolled, phase 3 trials. The Lancet Respiratory Medicine, 3(5), 355-366. http://doi.org/10.1016/S2213-2600(15)00042-9 Chanez, P., Bourdin, A., Vachier, I., Godard, P., Bousquet, J., and Vignola, A. M. (2004). Effects of inhaled corticosteroids on pathology in asthma and chronic obstructive pulmonary disease. Proceedings of the American Thoracic Society, 1(3), 184-190. http://dx.doi.org/10.1513/pats.200402-006ms

35

ACCEPTED MANUSCRIPT Chen, W., Sivaprasad, U., Gibson, A. M., Ericksen, M. B., Cunningham, C. M., Bass, S. A., ... and Hershey, G. K. K. (2013). IL-13 receptor α2 contributes to development of experimental allergic asthma. Journal of Allergy and Clinical Immunology, 132 (4), 951-958. http://dx.doi.org/10.1016/j.jaci.2013.04.016

RI PT

Chervinsky, P., Meltzer, E. O., Busse, W., Ohta, K., Bardin, P., Bredenbröker, D., and Bateman, E. D. (2015). Roflumilast for asthma: Safety findings from a pooled analysis of ten clinical studies. Pulmonary pharmacology and therapeutics, 35, S28-34. http://dx.doi.org/10.1016/j.pupt.2015.11.003 Chiba, Y., Matsusue, K., and Misawa, M. (2010). RhoA, a possible target for treatment of airway hyperresponsiveness in bronchial asthma. Journal of Pharmacological Sciences, 114(3), 239-247. http://dx.doi.org/10.1254/jphs.10r03cr

SC

Chung, K. F., Wenzel, S. E., Brozek, J. L., Bush, A., Castro, M., Sterk, P. J., ... and Teague, W. G. (2014). International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. European Respiratory Journal, 43 (2), 343-373. http://dx.doi.org/10.1183/09031936.00202013

M AN U

Clarke, D., Damera, G., Sukkar, M. B., and Tliba, O. (2009). Transcriptional regulation of cytokine function in airway smooth muscle cells. Pulmonary Pharmacology and Therapeutics, 22 (5), 436-445. http://dx.doi.org/10.1016/j.pupt.2009.04.003 Corren, J., Lemanske Jr, R. F., Hanania, N. A., Korenblat, P. E., Parsey, M. V., Arron, J. R., ... and Matthews, J. G. (2011). Lebrikizumab treatment in adults with asthma. New England Journal of Medicine, 365(12), 1088-1098. http://dx.doi.org/10.1056/nejmoa1106469

TE D

Corren, J., Weinstein, S., Janka, L., Zangrilli, J., and Garin, M. (2016). Phase 3 study of reslizumab in patients with poorly controlled asthma: effects across a broad range of eosinophil counts. Chest. http://doi.org/10.1016/j.chest.2016.03.018 Corry, D. B., and Kheradmand, F. (2009). Toward a comprehensive understanding of allergic lung disease. Transactions of the American Clinical and Climatological Association, 120, 33.

EP

Cox, L. S. (2009). How safe are the biologicals in treating asthma and rhinitis? Allergy, Asthma, and Clinical Immunology, 5 (1), 4. http://dx.doi.org/10.1186/1710-1492-5-4

AC C

Dahl, R. (2006). Systemic side effects of inhaled corticosteroids in patients with asthma. Respiratory Medicine, 100 (8), 1307-1317. http://dx.doi.org/10.1016/j.rmed.2005.11.020 Daines, M. O., Tabata, Y., Walker, B. A., Chen, W., Warrier, M. R., Basu, S., and Hershey, G. K. K. (2006). Level of expression of IL-13Rα2 impacts receptor distribution and IL-13 signaling. The Journal of Immunology, 176 (12), 7495-7501. http://dx.doi.org/10.4049/jimmunol.176.12.7495 del Giudice, M. M., Pezzulo, A., Capristo, C., Alterio, E., Caggiano, S., de Benedictis, D., and Capristo, A. F. (2009). Leukotriene modifiers in the treatment of asthma in children. Therapeutic Advances in Respiratory Disease, 3 (5), 245-251. http://dx.doi.org/10.1177/1753465809348014 Desai, D. and Brightling, C. (2009). Cytokine and anti‐cytokine therapy in asthma: Ready for the clinic? Clinical and Experimental Immunology, 158 (1), 10-19. http://dx.doi.org/10.1111/j.1365-2249.2009.03998.x

36

ACCEPTED MANUSCRIPT Dheda, K., Koegelenberg, C. F. N., Esmail, A., Irusen, E., Wechsler, M. E., Niven, R. M., ... and Chung, K. F. (2015). Recommendations for the use of bronchial thermoplasty in the management of severe asthma. South African Medical Journal, 105 (9), 726-732. http://dx.doi.org/10.7196/samjnew.8207.

RI PT

Diamant, Z., Sidharta, P. N., Singh, D., O'Connor, B. J., Zuiker, R., Leaker, B. R., ... and Dingemanse, J. (2014). Setipiprant, a selective CRTH2 antagonist, reduces allergen‐induced airway responses in allergic asthmatics. Clinical and Experimental Allergy, 44(8), 1044-1052. http://dx.doi.org/10.1111/cea.12357 Domínguez-Ortega, J., Phillips-Anglés, E., Barranco, P., and Quirce, S. (2015). Costeffectiveness of asthma therapy: A comprehensive review. Journal of Asthma, 52 (6), 529537. http://dx.doi.org/10.3109/02770903.2014.999283

SC

Dong, C. (2008). TH17 cells in development: an updated view of their molecular identity and genetic programming. Nature Reviews. Immunology, 8 (5), 337-348. http://dx.doi.org/10.1038/nri2295

M AN U

Dunford, P. J., O’Donnell, N., Riley, J. P., Williams, K. N., Karlsson, L., and Thurmond, R. L. (2006). The histamine H4 receptor mediates allergic airway inflammation by regulating the activation of CD4+ T cells. The Journal of Immunology, 176 (11), 7062-7070. http://dx.doi.org/10.4049/jimmunol.176.11.7062 Erin, E. M., Leaker, B. R., Nicholson, G. C., Tan, A. J., Green, L. M., Neighbour, H., … Hansel, T. T. (2006). The effects of a monoclonal antibody directed against tumor necrosis factor-α in asthma. American Journal of Respiratory and Critical Care Medicine, 174 (7), 753-762. http://dx.doi.org/10.1164/rccm.200601-072oc

TE D

Expert panel report 3 (EPR3): guidelines for the diagnosis and management of asthma. Bethesda, MD: National Heart, Lung, and Blood Institute, August 2007. (NIH publication no. 07-4051.) Available from: http://www.nhlbi.nih.gov/guidelines/asthma/asthgdln.pdf, last accessed May 8, 2016.

EP

Fabbri, L. M., Calverley, P. M., Izquierdo-Alonso, J. L., Bundschuh, D. S., Brose, M., Martinez, F. J., and Rabe, K. F. (2009). Roflumilast in moderate to severe chronic obstructive pulmonary disease treated with long acting bronchodilators: Two randomised clinical trials. The Lancet, 374 (9691), 695-703. http://dx.doi.org/10.1016/s0140-6736(09)61252-6

AC C

Fajt, M. L., Gelhaus, S. L., Freeman, B., Uvalle, C. E., Trudeau, J. B., Holguin, F., and Wenzel, S. E. (2013). Prostaglandin D 2 pathway upregulation: Relation to asthma severity, control, and T H 2 inflammation. Journal of Allergy and Clinical Immunology, 131 (6), 15041512. http://dx.doi.org/10.1016/j.jaci.2013.01.035 Fanta, C. (2009). Drug therapy. The New England Journal of Medicine, 360, 1002-14. http://dx.doi.org/10.1056/nejmra0804579 Feltis, B. N., Wignarajah, D., Reid, D. W., Ward, C., Harding, R., and Walters, E. H. (2007). Effects of inhaled fluticasone on angiogenesis and vascular endothelial growth factor in asthma. Thorax, 62 (4), 314-319. http://dx.doi.org/10.1136/thx.2006.069229 Fonseca, D. E., and Kline, J. N. (2009). Use of CpG oligonucleotides in treatment of asthma and allergic disease. Advanced Drug Delivery Reviews, 61 (3), 256-262. http://dx.doi.org/10.1016/j.addr.2008.12.007

37

ACCEPTED MANUSCRIPT Franciosi, L. G., Diamant, Z., Banner, K. H., Zuiker, R., Morelli, N., Kamerling, I. M., ... and Page, C. P. (2013). Efficacy and safety of RPL554, a dual PDE3 and PDE4 inhibitor, in healthy volunteers and in patients with asthma or chronic obstructive pulmonary disease: findings from four clinical trials. The Lancet Respiratory Medicine, 1(9), 714-727. http://dx.doi.org/10.1016/s2213-2600(13)70187-5

RI PT

Fuhst, R., Runge, F., Buschmann, J., Ernst, H., Praechter, C., Hansen, T., ... and Pohlmann, G. (2013). Toxicity profile of the GATA-3-specific DNAzyme hgd40 after inhalation exposure. Pulmonary Pharmacology and Therapeutics, 26 (2), 281-289. http://dx.doi.org/10.1016/j.pupt.2012.12.005 Fulkerson, P. C., and Rothenberg, M. E. (2013). Targeting eosinophils in allergy, inflammation and beyond. Nature Reviews. Drug Discovery, 12 (2), 117-129. http://dx.doi.org/10.1038/nrd3838

SC

Garcia, G., Taillé, C., Laveneziana, P., Bourdin, A., Chanez, P., and Humbert, M. (2013). Anti-interleukin-5 therapy in severe asthma. European Respiratory Review, 22 (129), 251257. http://dx.doi.org/10.1183/09059180.00004013

M AN U

Gauvreau, G. M., Boulet, L. P., Cockcroft, D. W., Baatjes, A., Cote, J., Deschesnes, F., ... and O'Byrne, P. M. (2008). Antisense therapy against CCR3 and the common beta chain attenuates allergen-induced eosinophilic responses. American Journal of Respiratory and Critical Care Medicine, 177 (9), 952-958. http://dx.doi.org/10.1164/rccm.200708-1251oc Gauvreau, G. M., Boulet, L. P., Schmid-Wirlitsch, C., Côté, J., Duong, M., Killian, K. J., ... and O'Byrne, P. M. (2011a). Roflumilast attenuates allergen-induced inflammation in mild asthmatic subjects. Respiratory Research, 12 (140), 10-1186. http://dx.doi.org/10.1186/14659921-12-140

TE D

Gauvreau, G. M., Pageau, R., Seguin, R., Carballo, D., Gauthier, J., D’anjou, H., ... and Killian, K. (2011b). Dose–response effects of TPI ASM8 in asthmatics after allergen. Allergy, 66 (9), 1242-1248. http://dx.doi.org/10.1111/j.1398-9995.2011.02638.x

EP

Global Asthma Report (2014). Auckland, New Zealand: Global Asthma Network, 2014. Available at: http://www.globalasthmareport.org/resources/Global_Asthma_Report_2014.pdf, last accessed on May 8, 2016.

AC C

Goh, F. G., and Midwood, K. S. (2012). Intrinsic danger: activation of Toll-like receptors in rheumatoid arthritis. Rheumatology, 51 (1), 7-23. http://dx.doi.org/10.1093/rheumatology/ker257 Gosens, R., Schaafsma, D., Bromhaar, M. M. G., Vrugt, B., Zaagsma, J., Meurs, H., and Nelemans, S. A. (2004). Growth factor-induced contraction of human bronchial smooth muscle is Rho-kinase-dependent. European Journal of Pharmacology, 494 (1), 73-76. Gupta, G. K., and Agrawal, D. K. (2010). CpG Oligodeoxynucleotides as TLR9 Agonists. BioDrugs, 24 (4), 225-235. http://dx.doi.org/10.2165/11536140-000000000-00000 Haldar, P., Brightling, C. E., Hargadon, B., Gupta, S., Monteiro, W., Sousa, A., Marshall, R. P., Bradding, P., Green, R. H., Wardlaw, A. J. and Pavord, I. D. (2009). Mepolizumab and exacerbations of refractory eosinophilic asthma. New England Journal of Medicine, 360 (10), 973-984. http://dx.doi.org/10.1056/nejmoa0808991

38

ACCEPTED MANUSCRIPT Haley, K. J., Lasky-Su, J., Manoli, S. E., Smith, L. A., Shahsafaei, A., Weiss, S. T. and Tantisira, K. (2011). RUNX transcription factors: association with pediatric asthma and modulated by maternal smoking. American Journal of Physiology-Lung Cellular and Molecular Physiology, 301 (5), L693-L701. http://dx.doi.org/10.1152/ajplung.00348.2010

RI PT

Hall, I. P., Fowler, A. V., Gupta, A., Tetzlaff, K., Nivens, M. C., Sarno, M., ... and Sutherland, E. R. (2015). Efficacy of BI 671800, an oral CRTH2 antagonist, in poorly controlled asthma as sole controller and in the presence of inhaled corticosteroid treatment. Pulmonary Pharmacology and Therapeutics, 32, 37-44. http://dx.doi.org/10.1016/j.pupt.2015.03.003 Hall, S., and Agrawal, D. K. (2014). Key mediators in the immunopathogenesis of allergic asthma. International Immunopharmacology, 23 (1), 316-329. http://dx.doi.org/10.1016/j.intimp.2014.05.034

SC

Hambly, N., and Nair, P. (2014). Monoclonal antibodies for the treatment of refractory asthma. Current Opinion in Pulmonary Medicine, 20 (1), 87-94. http://dx.doi.org/10.1097/mcp.0000000000000007

M AN U

Hanania, N. A., Noonan, M., Corren, J., Korenblat, P., Zheng, Y., Fischer, S. K., ... and Yen, K. (2015). Lebrikizumab in moderate-to-severe asthma: pooled data from two randomised placebo-controlled studies. Thorax, 70(8), 748-756. http://dx.doi.org/10.1136/thoraxjnl-2014206719 Hansbro, P. M., Kaiko, G. E., and Foster, P. S. (2011). Cytokine/anti‐cytokine therapy–novel treatments for asthma? British Journal of Pharmacology, 163 (1), 81-95. http://dx.doi.org/10.1111/j.1476-5381.2011.01219.x

TE D

Hartwig, C., Munder, A., Glage, S., Wedekind, D., Schenk, H., Seifert, R., and Neumann, D. (2015). The histamine H4‐receptor (H4R) regulates eosinophilic inflammation in ovalbumin‐ induced experimental allergic asthma in mice. European Journal of Immunology, 45 (4), 1129-1140. http://dx.doi.org/10.1002/eji.201445179

EP

Harvima, I. T., Levi-Schaffer, F., Draber, P., Friedman, S., Polakovicova, I., Gibbs, B. F., ... and Maurer, M. (2014). Molecular targets on mast cells and basophils for novel therapies. Journal of Allergy and Clinical Immunology, 134 (3), 530-544. http://dx.doi.org/10.1016/j.jaci.2014.03.007

AC C

Hatchwell, L., Collison, A., Girkin, J., Parsons, K., Li, J., Zhang, J., ... and Mattes, J. (2015). Toll-like receptor 7 governs interferon and inflammatory responses to rhinovirus and is suppressed by IL-5-induced lung eosinophilia. Thorax, 70(9), 854-861. http://dx.doi.org/10.1136/thoraxjnl-2014-205465 Henderson, W. R., Banerjee, E. R., and Chi, E. Y. (2005). Differential effects of (S)-and (R)enantiomers of albuterol in a mouse asthma model. Journal of Allergy and Clinical Immunology, 116 (2), 332-340. http://dx.doi.org/10.1016/j.jaci.2005.04.013 Hennessy, E. J., Parker, A. E., and O'Neill, L. A. (2010). Targeting Toll-like receptors: emerging therapeutics? Nature Reviews. Drug Discovery, 9 (4), 293-307. http://dx.doi.org/10.1038/nrd3203 Henry, P. J., Mann, T. S., and Goldie, R. G. (2005). A Rho kinase inhibitor, Y-27632 inhibits pulmonary eosinophilia, bronchoconstriction and airways hyperresponsiveness in allergic mice. Pulmonary Pharmacology and Therapeutics, 18 (1), 67-74. http://dx.doi.org/10.1016/j.pupt.2004.10.002 39

ACCEPTED MANUSCRIPT Hernández-Hernández, L., Sanz, C., García-Solaesa, V., Padrón, J., García-Sánchez, A., Dávila, I., ... and Lorente, F. (2012). Tryptase: Genetic and functional considerations. Allergologia et Immunopathologia, 40 (6), 385-389. http://dx.doi.org/10.1016/j.aller.2012.04.004 Holgate, S. T. (2008). Pathogenesis of asthma. Clinical and Experimental Allergy, 38 (6), 872-897. http://dx.doi.org/10.1111/j.1365-2222.2008.02971.x

RI PT

Holgate, S. T. (2012). Trials and tribulations in identifying new biologic treatments for asthma. Trends in Immunology, 33 (5), 238-246. http://dx.doi.org/10.1016/j.it.2012.02.003 Holgate, S. T., Noonan, M., Chanez, P., Busse, W., Dupont, L., Pavord, I., ... and Nelson, H. (2011). Efficacy and safety of etanercept in moderate-to-severe asthma: a randomised, controlled trial. European Respiratory Journal,37(6), 1352-1359. . http://doi.org/10.1183/09031936.00063510

M AN U

SC

Homburg, U., Renz, H., Timmer, W., Hohlfeld, J. M., Seitz, F., Lüer, K., ... and Turowska, A. (2015). Safety and tolerability of a novel inhaled GATA3 mRNA targeting DNAzyme in patients with TH2-driven asthma. Journal of Allergy and Clinical Immunology, 136 (3), 797. http://dx.doi.org/10.1016/j.jaci.2015.02.018 Israel, E., Banerjee, T. R., Fitzmaurice, G. M., Kotlov, T. V., LaHive, K., and LeBoff, M. S. (2001). Effects of inhaled glucocorticoids on bone density in premenopausal women. New England Journal of Medicine, 345(13), 941-947. http://dx.doi.org/10.1056/nejmoa002304

TE D

Jenner, R. G., Townsend, M. J., Jackson, I., Sun, K., Bouwman, R. D., Young, R. A., ... and Lord, G. M. (2009). The transcription factors T-bet and GATA-3 control alternative pathways of T-cell differentiation through a shared set of target genes. Proceedings of the National Academy of Sciences, 106 (42), 17876-17881. http://dx.doi.org/10.1073/pnas.0909357106 Jia, G., Erickson, R. W., Choy, D. F., Mosesova, S., Wu, L. C., Solberg, O. D., … and Arron, J. R. (2012). Periostin is a systemic biomarker of eosinophilic airway inflammation in asthmatic patients. Journal of Allergy and Clinical Immunology, 130 (3), 647-654. http://dx.doi.org/10.1016/j.jaci.2012.06.025

AC C

EP

Kaiko, G. E., Horvat, J. C., Beagley, K. W., and Hansbro, P. M. (2008). Immunological decision‐making: how does the immune system decide to mount a helper T‐cell response? Immunology, 123 (3), 326-338. http://dx.doi.org/10.1111/j.13652567.2007.02719.x Kallinich, T., Beier, K. C., Wahn, U., Stock, P., and Hamelmann, E. (2007). T-cell costimulatory molecules: their role in allergic immune reactions. European Respiratory Journal, 29 (6), 1246-1255. http://dx.doi.org/10.1183/09031936.00094306 Kato, M., Komamura, K., and Kitakaze, M. (2006). Tiotropium, a novel muscarinic M3 receptor antagonist, improved symptoms of chronic obstructive pulmonary disease complicated by chronic heart failure. Circulation Journal, 70 (12), 1658-1660. http://dx.doi.org/10.1253/circj.70.1658 Kauffman, H. F., and van der Heide, S. (2003). Exposure, sensitization, and mechanisms of fungus-induced asthma. Current Allergy and Asthma Reports, 3 (5), 430-437. http://dx.doi.org/10.1007/s11882-003-0080-z Kim, S. H., Hong, J. H., and Lee, Y. C. (2013). Ursolic acid, a potential PPARγ agonist, suppresses ovalbumin-induced airway inflammation and Penh by down-regulating IL-5, IL40

ACCEPTED MANUSCRIPT 13, and IL-17 in a mouse model of allergic asthma. European Journal of Pharmacology, 701(1), 131-143. http://dx.doi.org/10.1016/j.ejphar.2012.11.033 Kline, J. N., and Krieg, A. M. (2010). Toll-like receptor 9 activation with CpG oligodeoxynucleotides for asthma therapy. New Drugs and Targets for Asthma and COPD, 39, 95-99. http://dx.doi.org/10.1159/000320805

RI PT

Knudsen, T. B., Thomsen, S. F., Nolte, H., and Backer, V. (2009). A population-based clinical study of allergic and non-allergic asthma. Journal of Asthma,46(1), 91-94. http://dx.doi.org/10.1080/02770900802524657 Kobayashi, M., Kubo, S., Shiraki, K., Iwata, M., Hirano, Y., Ohtsu, Y., ... and Shimizu, Y. (2012). Therapeutic potential of ASP3258, a selective phosphodiesterase 4 inhibitor, on chronic eosinophilic airway inflammation. Pharmacology, 90 (3-4), 223-232. http://dx.doi.org/10.1159/000342380

M AN U

SC

Kolbeck, R., Kozhich, A., Koike, M., Peng, L., Andersson, C. K., Damschroder, M. M., ... and Coyle, A. J. (2010). MEDI-563, a humanized anti–IL-5 receptor α mAb with enhanced antibody-dependent cell-mediated cytotoxicity function. Journal of Allergy and Clinical Immunology, 125 (6), 1344-1353. http://dx.doi.org/10.1016/j.jaci.2010.04.004 Koopmans, T., Anaparti, V., Castro-Piedras, I., Yarova, P., Irechukwu, N., Nelson, C., ... and Wright, D. B. (2014). Ca2+ handling and sensitivity in airway smooth muscle: Emerging concepts for mechanistic understanding and therapeutic targeting. Pulmonary Pharmacology and Therapeutics, 29 (2), 108-120. http://dx.doi.org/10.1016/j.pupt.2014.05.001

TE D

Krug, N., Hohlfeld, J. M., Kirsten, A. M., Kornmann, O., Beeh, K. M., Kappeler, D., ... and Zeitvogel, J. (2015). Allergen-induced asthmatic responses modified by a GATA3-specific DNAzyme. New England Journal of Medicine, 372 (21), 1987-1995. http://dx.doi.org/10.1056/nejmoa1411776

EP

Kudlacz, E., Conklyn, M., Andresen, C., Whitney-Pickett, C., and Changelian, P. (2008). The JAK-3 inhibitor CP-690550 is a potent anti-inflammatory agent in a murine model of pulmonary eosinophilia. European Journal of Pharmacology, 582 (1), 154-161. http://dx.doi.org/10.1016/j.ejphar.2007.12.024 Lambrecht, B. N., and Hammad, H. (2015). The immunology of asthma. Nature Immunology, 16 (1), 45-56. http://dx.doi.org/10.1038/ni.3049

AC C

Lassen, C., Casale, T., Beeh, K. M., and Pavord, I. (2014). Exhaled nitric oxide and the response to Cyt003-Qbg10 in patients with persistent allergic asthma. American Journal of Respiratory and Critical Care Medicine, 189, A6682. Liu, Z., Liu, X., Sang, L., Liu, H., Xu, Q., and Liu, Z. (2015). Boswellic acid attenuates asthma phenotypes by downregulation of GATA3 via pSTAT6 inhibition in a murine model of asthma. International Journal of Clinical and Experimental Pathology, 8(1), 236–243. Lötvall, J., Akdis, C. A., Bacharier, L. B., Bjermer, L., Casale, T. B., Custovic, A., ... and Greenberger, P. A. (2011). Asthma endotypes: a new approach to classification of disease entities within the asthma syndrome. Journal of Allergy and Clinical Immunology, 127 (2), 355-360. http://dx.doi.org/10.1016/j.jaci.2010.11.037 Lukacs, N. W., Berlin, A. A., Franz-Bacon, K., Šášik, R., Sprague, L. J., Ly, T. W., ... and Bacon, K. B. (2008). CRTH2 antagonism significantly ameliorates airway hyperreactivity and downregulates inflammation-induced genes in a mouse model of airway 41

ACCEPTED MANUSCRIPT inflammation. American Journal of Physiology-Lung Cellular and Molecular Physiology, 295 (5), L767-L779. http://dx.doi.org/10.1152/ajplung.90351.2008 Maarsingh, H., Bossenga, B. E., Bos, I. S. T., Volders, H. H., Zaagsma, J., and Meurs, H. (2009). L-arginine deficiency causes airway hyperresponsiveness after the late asthmatic reaction. European Respiratory Journal, 34 (1), 191-199. http://dx.doi.org/10.1183/09031936.00105408

RI PT

Maarsingh, H., Zuidhof, A. B., Bos, I. S. T., van Duin, M., Boucher, J. L., Zaagsma, J., and Meurs, H. (2008). Arginase inhibition protects against allergen-induced airway obstruction, hyperresponsiveness, and inflammation. American Journal of Respiratory and Critical Care Medicine, 178 (6), 565-573. http://dx.doi.org/10.1164/rccm.200710-1588oc

SC

Maher, S. A., Birrell, M. A., Adcock, J. J., Wortley, M. A., Dubuis, E. D., Bonvini, S. J., ... and Belvisi, M. G. (2015). Prostaglandin D2 and the role of the DP1, DP2 and TP receptors in the control of airway reflex events. European Respiratory Journal, 45 (4), 1108-1118. http://dx.doi.org/10.1183/09031936.00061614

M AN U

Mahn, K., Ojo, O. O., Chadwick, G., Aaronson, P. I., Ward, J. P., and Lee, T. H. (2010). Ca2+ homeostasis and structural and functional remodelling of airway smooth muscle in asthma. Thorax, 65 (6), 547-552. http://dx.doi.org/10.1136/thx.2009.129296 Maiga, M., Lun, S., Guo, H., Winglee, K., Ammerman, N. C., and Bishai, W. R. (2012). Risk of tuberculosis reactivation with tofacitinib (CP-690550). Journal of Infectious Diseases, 205 (11), 1705-1708. http://dx.doi.org/10.1093/infdis/jis269 Markham, A. (2016). Reslizumab: First Global Approval. Drugs, 76(8), 907-911. . http://doi.org/10.1007/s40265-016-0583-2

TE D

Marques, R. H., Reis, F. G., Starling, C. M., Cabido, C., de Almeida-Reis, R., Dohlnikoff, M., … and Tibério, I. F. L. C. (2012). Inducible nitric oxide synthase inhibition attenuates physical stress-induced lung hyper-responsiveness and oxidative stress in animals with lung inflammation. Neuroimmunomodulation, 19 (3), 158-170. http://dx.doi.org/10.1159/000331264

EP

Mata, M., Martinez, I., Melero, J. A., Tenor, H., and Cortijo, J. (2013). Roflumilast inhibits respiratory syncytial virus infection in human differentiated bronchial epithelial cells. PloS ONE, 8 (7), e69670. http://dx.doi.org/10.1371/journal.pone.0069670

AC C

Matera, M. G., Page, C., and Cazzola, M. (2014). PDE inhibitors currently in early clinical trials for the treatment of asthma. Expert Opinion on Investigational Drugs, 23 (9), 12671275. http://dx.doi.org/10.1517/13543784.2014.921157 Matsunaga, Y., Inoue, H., Fukuyama, S., Yoshida, H., Moriwaki, A., Matsumoto, T., ... and Nakanishi, Y. (2011). Effects of a Janus kinase inhibitor, pyridone 6, on airway responses in a murine model of asthma. Biochemical and Biophysical Research Communications, 404 (1), 261-267. http://dx.doi.org/10.1016/j.bbrc.2010.11.104 Meng, L., He, X., Zhu, W., Yang, X., Jiang, C., Sun, Q., ... and Lu, S. (2011). TLR3 and TLR7 modulate IgE production in antigen induced pulmonary inflammation via influencing IL-4 expression in immune organs. PloS ONE, 6 (2), e17252-e17252. http://dx.doi.org/10.1371/journal.pone.0017252 Mestre-Ferrandiz, J., Sussex, J., and Towse, A. (2012). The R and D cost of a new medicine. London: Office of Health Economics. Available from: 42

ACCEPTED MANUSCRIPT www.fiercebiotech.com/press-releases/new-ohe-study-pharmaceutical-rd-costs-released, last accessed May 8, 2016. Mowbray, C. E., Bell, A. S., Clarke, N. P., Collins, M., Jones, R. M., Lane, C. A., ... and Selby, M. D. (2011). Challenges of drug discovery in novel target space. The discovery and evaluation of PF-3893787: a novel histamine H4 receptor antagonist. Bioorganic and Medicinal Chemistry Letters, 21 (21), 6596-6602. http://dx.doi.org/10.1016/j.bmcl.2011.07.125

RI PT

Murata, Y., Song, M., Kikuchi, H., Hisamichi, K., Xu, X. L., Greenspan, A., ... and Thurmond, R. L. (2015). Phase 2a, randomized, double‐blind, placebo‐controlled, multicenter, parallel‐group study of a H4R‐antagonist (JNJ‐39758979) in Japanese adults with moderate atopic dermatitis. The Journal of Dermatology, 42 (2), 129-139. http://dx.doi.org/10.1111/1346-8138.12726

SC

Murray, C. J., Vos, T., Lozano, R., Naghavi, M., Flaxman, A. D., Michaud, C., ... and Bridgett, L. (2013). Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. The Lancet, 380 (9859), 2197-2223. http://dx.doi.org/10.1016/s0140-6736(12)61689-4

M AN U

Nagashima, S., Hondo, T., Nagata, H., Ogiyama, T., Maeda, J., Hoshii, H., ... and Ohno, K. (2009). Novel 7H-pyrrolo [2, 3-d] pyrimidine derivatives as potent and orally active STAT6 inhibitors. Bioorganic and Medicinal Chemistry, 17 (19), 6926-6936. http://dx.doi.org/10.1016/j.bmc.2009.08.021 Nagumo, H., Sasaki, Y., Ono, Y., Okamoto, H., Seto, M., and Takuwa, Y. (2000). Rho kinase inhibitor HA-1077 prevents Rho-mediated myosin phosphatase inhibition in smooth muscle cells. American Journal of Physiology-Cell Physiology, 278 (1), C57-C65.

TE D

Nair, P., Pizzichini, M. M., Kjarsgaard, M., Inman, M. D., Efthimiadis, A., Pizzichini, E., Hargreave, F. E. and O'Byrne, P. M. (2009). Mepolizumab for prednisone-dependent asthma with sputum eosinophilia. New England Journal of Medicine, 360 (10), 985-993. http://dx.doi.org/10.1056/nejmoa0805435

AC C

EP

Neumann, D., Beermann, S., Burhenne, H., Glage, S., Hartwig, C., and Seifert, R. (2013). The dual H3/4R antagonist thioperamide does not fully mimic the effects of the ‘standard’H4R antagonist JNJ 7777120 in experimental murine asthma. NaunynSchmiedeberg's Archives of Pharmacology, 386 (11), 983-990. http://dx.doi.org/10.1007/s00210-013-0898-4 Newcomb, D. C., and Peebles, R. S. (2013). Th17-mediated inflammation in asthma. Current Opinion in Immunology, 25 (6), 755-760. http://dx.doi.org/10.1016/j.coi.2013.08.002 Nichols, H. L., Saffeddine, M., Theriot, B. S., Hegde, A., Polley, D., El-Mays, T., ... and DeFea, K. A. (2012). β-Arrestin-2 mediates the proinflammatory effects of proteinaseactivated receptor-2 in the airway. Proceedings of the National Academy of Sciences, 109 (41), 16660-16665. http://dx.doi.org/10.1073/pnas.1208881109 Nie, H., Zhang, G., Liu, M., Ding, X., Huang, Y., and Hu, S. (2013). Efficacy of theophylline plus salmeterol/fluticasone propionate combination therapy in patients with asthma. Respiratory Medicine, 107 (3), 347-354. http://dx.doi.org/10.1016/j.rmed.2012.12.004 Noonan, M., Korenblat, P., Mosesova, S., Scheerens, H., Arron, J. R., Zheng, Y., ... and Matthews, J. G. (2013). Dose-ranging study of lebrikizumab in asthmatic patients not 43

ACCEPTED MANUSCRIPT receiving inhaled steroids. Journal of Allergy and Clinical Immunology, 132 (3), 567-574. http://dx.doi.org/10.1016/j.jaci.2013.03.051 North, M. L., Grasemann, H., Khanna, N., Inman, M. D., Gauvreau, G. M., and Scott, J. A. (2013). Increased ornithine-derived polyamines cause airway hyperresponsiveness in a mouse model of asthma. American Journal of Respiratory Cell and Molecular Biology, 48 (6), 694702. http://dx.doi.org/10.1165/rcmb.2012-0323oc

RI PT

North, M. L., Khanna, N., Marsden, P. A., Grasemann, H., and Scott, J. A. (2009). Functionally important role for arginase 1 in the airway hyperresponsiveness of asthma. American Journal of Physiology-Lung Cellular and Molecular Physiology, 296 (6), L911-L920. http://dx.doi.org/10.1152/ajplung.00025.2009

SC

Nowak, R. M., Parker, J. M., Silverman, R. A., Rowe, B. H., Smithline, H., Khan, F., ... and Molfino, N. A. (2015). A randomized trial of benralizumab, an antiinterleukin 5 receptor α monoclonal antibody, after acute asthma. The American Journal of Emergency Medicine, 33 (1), 14-20. http://dx.doi.org/10.1016/j.ajem.2014.09.036

M AN U

Ortega, H. G., Liu, M. C., Pavord, I. D., Brusselle, G. G., FitzGerald, J. M., Chetta, A., ... and Chanez, P. (2014). Mepolizumab treatment in patients with severe eosinophilic asthma. New England Journal of Medicine, 371 (13), 1198-1207. http://dx.doi.org/10.1056/nejmoa1403290 Page, C. P. (2014). Phosphodiesterase Inhibitors for the Treatment of Asthma and Chronic Obstructive Pulmonary Disease. International Archives of Allergy and Immunology, 165 (3), 152-164. http://dx.doi.org/10.1159/000368800

TE D

Panter, G., Kuznik, A., and Jerala, R. (2009). Therapeutic applications of nucleic acids as ligands for Toll-like receptors. Current Opinion in Molecular Therapeutics, 11 (2), 133-145. Parikh, N., and Chakraborti, A. (2016). Phosphodiesterase 4 (PDE4) Inhibitors in the Treatment of COPD: Promising Drug Candidates and Future Directions. Current Medicinal Chemistry, 23(2), 129–141. http://dx.doi.org/10.2174/0929867323666151117121334

EP

Pavord, I. D., Korn, S., Howarth, P., Bleecker, E. R., Buhl, R., Keene, O. N., Hector, O. and Chanez, P. (2012). Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. The Lancet, 380 (9842), 651-659. http://dx.doi.org/10.1016/s0140-6736(12)60988-x

AC C

Pégorier, S., Wagner, L. A., Gleich, G. J., and Pretolani, M. (2006). Eosinophil-derived cationic proteins activate the synthesis of remodeling factors by airway epithelial cells. The Journal of Immunology, 177 (7), 4861-4869. http://dx.doi.org/10.4049/jimmunol.177.7.4861 Pelaia, G., Vatrella, A., Busceti, M. T., Gallelli, L., Calabrese, C., Terracciano, R., and Maselli, R. (2015). Cellular Mechanisms Underlying Eosinophilic and Neutrophilic Airway Inflammation in Asthma. Mediators of Inflammation, 2015, 879783-879783. http://dx.doi.org/10.1155/2015/879783 Pettipher, R., Vinall, S. L., Xue, L., Speight, G., Townsend, E. R., Gazi, L., ... and Hunter, M. G. (2012). Pharmacologic profile of OC000459, a potent, selective, and orally active D prostanoid receptor-2 antagonist that inhibits mast cell-dependent activation of T helper 2 lymphocytes and eosinophils. Journal of Pharmacology and Experimental Therapeutics, 340 (2), 473-482. http://dx.doi.org/10.1124/jpet.111.187203

44

ACCEPTED MANUSCRIPT Pham Van, L., Bardel, E., Gregoire, S., Vanoirbeek, J., Schneider, E., Dy, M., and Thieblemont, N. (2011). Treatment with the TLR7 agonist R848 induces regulatory T‐cell‐ mediated suppression of established asthma symptoms. European Journal of Immunology, 41 (7), 1992-1999. http://dx.doi.org/10.1002/eji.201040914

RI PT

Pigati, P. A., Righetti, R. F., Possa, S. S., Romanholo, B. S., Rodrigues, A. P., dos Santos, A. S., ... and Tibério, I. D. F. L. C. (2015). Y-27632 is associated with corticosteroid-potentiated control of pulmonary remodeling and inflammation in guinea pigs with chronic allergic inflammation. BMC Pulmonary Medicine, 15 (1), 85. http://dx.doi.org/10.1186/s12890-0150073-4 Piper, E., Brightling, C., Niven, R., Oh, C., Faggioni, R., Poon, K., ... and Molfino, N. A. (2013). A phase II placebo-controlled study of tralokinumab in moderate-to-severe asthma. European Respiratory Journal, 41 (2), 330-338. http://dx.doi.org/10.1183/09031936.00223411

SC

Polosa, R., and Thomson, N. C. (2013). Smoking and asthma: dangerous liaisons. European Respiratory Journal, 41 (3), 716-726. http://dx.doi.org/10.1183/09031936.00073312

M AN U

Portnoy, A., Kumar, S., Behm, D. J., Mahar, K. M., Noble, R. B., Throup, J. P., and Russ, S. F. (2013). Effects of urotensin II receptor antagonist, GSK1440115, in asthma. Frontiers in Pharmacology, 4, 54. http://dx.doi.org/10.3389/fphar.2013.00054 Possa, S. S., Charafeddine, H. T., Righetti, R. F., da Silva, P. A., Almeida-Reis, R., SaraivaRomanholo, B. M., ... and Tibério, I. D. F. L. C. (2012). Rho-kinase inhibition attenuates airway responsiveness, inflammation, matrix remodeling, and oxidative stress activation induced by chronic inflammation. American Journal of Physiology-Lung Cellular and Molecular Physiology, 303 (11), L939-L952. http://dx.doi.org/10.1152/ajplung.00034.2012

TE D

Pouliquen, I. J., Kornmann, O., Barton, S. V., Price, J. A., and Ortega, H. G. (2015). Characterization of the relationship between dose and blood eosinophil response following subcutaneous administration of mepolizumab. International Journal of Clinical Pharmacology and Therapeutics, 53(12), 1015–1027. http://doi.org/10.5414/CP202446

AC C

EP

Prado, C. M., Leick-Maldonado, E. A., Kasahara, D. I., Capelozzi, V. L., Martins, M. A., and Tiberio, I. F. (2005). Effects of acute and chronic nitric oxide inhibition in an experimental model of chronic pulmonary allergic inflammation in guinea pigs. American Journal of Physiology-Lung Cellular and Molecular Physiology, 289 (4), L677-L683. http://dx.doi.org/10.1152/ajplung.00010.2005 Prado, C. M., Righetti, R. F., da Silva Pigati, P. A., Possa, S. S. and dos Santos, A. S. A. (2014). New pharmacological targets for asthma drug development. Journal of Allergy and Therapy, 5, 170-172. http://dx.doi.org/10.4172/2155-6121.1000170 Prado, C. M., Yano, L., Rocha, G., Starling, C. M., Capelozzi, V. L., Leick-Maldonado, E. A., ... and Tibério, I. F. L. C. (2011). Effects of inducible nitric oxide synthase inhibition in bronchial vascular remodeling-induced by chronic allergic pulmonary inflammation. Experimental Lung Research, 37 (5), 259-268. http://dx.doi.org/10.3109/01902148.2010.538289 Raja, S.G. (2012). Evaluation of clinical efficacy of fasudil for the treatment of pulmonary arterial hypertension. Recent Patents on Cardiovascular Drug Discovery, 7 (2), 100-104. http://dx.doi.org/10.2174/157489012801227238

45

ACCEPTED MANUSCRIPT Reddel, H. K., Bateman, E. D., Becker, A., Boulet, L. P., Cruz, A. A., Drazen, J. M., ... and FitzGerald, J. M. (2015). A summary of the new GINA strategy: a roadmap to asthma control. European Respiratory Journal, ERJ-00853. http://dx.doi.org/10.1183/13993003.00853-2015

RI PT

Reher, T. M., Neumann, D., Buschauer, A., and Seifert, R. (2012). Incomplete activation of human eosinophils via the histamine H 4-receptor: evidence for ligand-specific receptor conformations. Biochemical Pharmacology, 84 (2), 192-203. http://dx.doi.org/10.1016/j.bcp.2012.04.004 Reis, F. G., Marques, R. H., Starling, C. M., Almeida-Reis, R., Vieira, R. P., Cabido, C. T., ... and Tibério, I. F. (2012). Stress amplifies lung tissue mechanics, inflammation and oxidative stress induced by chronic inflammation. Experimental Lung Research, 38 (7), 344-354. http://dx.doi.org/10.3109/01902148.2012.704484

SC

Ricciardolo, F. L., Sterk, P. J., Gaston, B. and Folkerts, G. (2004). Nitric oxide in health and disease of the respiratory system. Physiological Reviews, 84 (3), 731-765. http://dx.doi.org/10.1152/physrev.00034.2003

M AN U

Righetti, R. F., Pigati, P. A. da S., Possa, S. S., Habrum, F. C., Xisto, D. G., Antunes, M. A., … and Tibério, I. F. L. C. (2014). Effects of Rho-kinase inhibition in lung tissue with chronic inflammation. Respiratory Physiology and Neurobiology, 192, 134-146. http://dx.doi.org/10.1016/j.resp.2013.12.012 Rodrigo, G. J. and Castro-Rodriguez, J. A. (2005). Anticholinergics in the treatment of children and adults with acute asthma: a systematic review with meta-analysis. Thorax, 60 (9), 740-746. http://dx.doi.org/10.1136/thx.2005.047803

TE D

Roth, M. and Black, J. L. (2006). Transcription factors in asthma: are transcription factors a new target for asthma therapy? Current Drug Targets, 7 (5), 589-595. http://dx.doi.org/10.2174/138945006776818638

EP

Saag, K. G., Furst, D. E. and Barnes, P. J. (2011). Major side effects of inhaled glucocorticoids. In: Bochner BS, Wood RA, editors. UpToDate, 19. Available from: http://www.uptodate.com/contents/major-side-effects-of-inhaled-glucocorticoids, last accessed May 8, 2016

AC C

Salcedo, C. (2013). Is the H4 receptor a new drug target for allergies and asthma. Front Biosci, E5(1), 178–187. http://dx.doi.org/10.2741/e606 Schaafsma D, Bos IST, Zuidhof AB, Zaagsma J, Meurs H. Inhalation of the Rho-kinase inhibitor Y-27632 reverses allergen-induced airway hyperresponsiveness after the early and late asthmatic reaction. Respiratory Research. 2006;7(1):121. http://dx.doi.org/10.1186/14659921-7-121. Schaafsma, D., Bos, I. S. T., Zuidhof, A. B., Zaagsma, J. and Meurs, H. (2008). The inhaled Rho kinase inhibitor Y-27632 protects against allergen-induced acute bronchoconstriction, airway hyperresponsiveness, and inflammation. American Journal of Physiology-Lung Cellular and Molecular Physiology, 295 (1), L214-L219. http://dx.doi.org/10.1152/ajplung.00498.2007 Schieck, M., Schouten, J. P., Michel, S., Suttner, K., Toncheva, A. A., Gaertner, V. D., … Kabesch, M. (2016). Doublesex and mab-3 related transcription factor 1 (DMRT1) is a sexspecific genetic determinant of childhood-onset asthma and is expressed in testis and 46

ACCEPTED MANUSCRIPT macrophages. Journal of Allergy and http://dx.doi.org/10.1016/j.jaci.2015.12.1305

Clinical

Immunology.

(In

press)

Schmidlin, F., Amadesi, S., Dabbagh, K., Lewis, D. E., Knott, P., Bunnett, N. W., ... and Stevens, M. E. (2002). Protease-activated receptor 2 mediates eosinophil infiltration and hyperreactivity in allergic inflammation of the airway. The Journal of Immunology, 169 (9), 5315-5321. http://dx.doi.org/10.4049/jimmunol.169.9.5315

RI PT

Seifert, R., Strasser, A., Schneider, E. H., Neumann, D., Dove, S., and Buschauer, A. (2013). Molecular and cellular analysis of human histamine receptor subtypes. Trends in Pharmacological Sciences, 34 (1), 33-58. http://dx.doi.org/10.1016/j.tips.2012.11.001

SC

Sel, S., Wegmann, M., Dicke, T., Sel, S., Henke, W., Yildirim, A. Ö., ... and Garn, H. (2008). Effective prevention and therapy of experimental allergic asthma using a GATA-3–specific DNAzyme. Journal of Allergy and Clinical Immunology, 121 (4), 910-916. http://dx.doi.org/10.1016/j.jaci.2007.12.1175

M AN U

Senti, G., Johansen, P., Haug, S., Bull, C., Gottschaller, C., Müller, P., Pfister, T., Maurer, P., Bachmann, M. F., Graf, N. and Kündig, T. M. (2009). Use of A‐type CpG oligodeoxynucleotides as an adjuvant in allergen‐specific immunotherapy in humans: a phase I/IIa clinical trial. Clinical and Experimental Allergy, 39 (4), 562-570. http://dx.doi.org/10.1111/j.1365-2222.2008.03191.x Sharma, A., Menche, J., Huang, C. C., Ort, T., Zhou, X., Kitsak, M., … Barabasi, A.-L. (2015). A disease module in the interactome explains disease heterogeneity, drug response and captures novel pathways and genes in asthma. Human Molecular Genetics, 24 (11), 3005-3020. http://dx.doi.org/10.1093/hmg/ddv001

TE D

Shishodia, S., Majumdar, S., Banerjee, S., and Aggarwal, B. B. (2003). Ursolic acid inhibits nuclear factor-κB activation induced by carcinogenic agents through suppression of IκBα kinase and p65 phosphorylation correlation with down-regulation of cyclooxygenase 2, matrix metalloproteinase 9, and cyclin D1. Cancer Research, 63 (15), 4375-4383.

EP

Sidharta, P. N., Diamant, Z., and Dingemanse, J. (2014). Single‐and multiple‐dose tolerability and pharmacokinetics of the CRTH2 antagonist setipiprant in healthy male subjects. Fundamental and Clinical Pharmacology, 28(6), 690-699. http://dx.doi.org/10.1111/fcp.12079

AC C

Singh, D., Cadden, P., Hunter, M., Collins, L. P., Perkins, M., Pettipher, R., ... and O'Connor, B. (2012). Inhibition of the asthmatic allergen challenge response by the CRTH2 antagonist OC000459. European Respiratory Journal, 41 (1), 46-52. http://dx.doi.org/10.1183/09031936.00092111 Singh, D., Petavy, F., Macdonald, A. J., Lazaar, A. L., and O'Connor, B. J. (2010). The inhaled phosphodiesterase 4 inhibitor GSK256066 reduces allergen challenge responses in asthma. Respiratory Research, 11 (1), 26. http://dx.doi.org/10.1186/1465-9921-11-26 Singh, D., Richards, D., Knowles, R. G., Schwartz, S., Woodcock, A., Langley, S., and O'Connor, B. J. (2007). Selective inducible nitric oxide synthase inhibition has no effect on allergen challenge in asthma. American Journal of Respiratory and Critical Care Medicine, 176 (10), 988-993. http://dx.doi.org/10.1164/rccm.200704-588oc Somma, T., Cinci, L., Formicola, G., Pini, A., Thurmond, R., Ennis, M., ... and Masini, E. (2013). A selective antagonist of histamine H4 receptors prevents antigen‐induced airway 47

ACCEPTED MANUSCRIPT inflammation and bronchoconstriction in guinea pigs: involvement of lipocortin‐1. British Journal of Pharmacology, 170 (1), 200-213. http://dx.doi.org/10.1111/bph.12264 Souza, F. C. R., Gobbato, N. B., Maciel, R. G., Prado, C. M., Martins, M. A., Leick, E. A. and Tibério, I. F. C. (2013). Effects of corticosteroid, montelukast and iNOS inhibition on distal lung with chronic inflammation. Respiratory Physiology and Neurobiology, 185 (2), 435-445. http://dx.doi.org/10.1016/j.resp.2012.08.015

RI PT

Spears, M., Donnelly, I., Jolly, L., Brannigan, M., Ito, K., McSharry, C., … Thomson, N. C. (2009). Effect of low-dose theophylline plus beclometasone on lung function in smokers with asthma: a pilot study. European Respiratory Journal, 33 (5), 1010-1017. http://dx.doi.org/10.1183/09031936.00158208

SC

Stark, A. K., Sriskantharajah, S., Hessel, E. M., and Okkenhaug, K. (2015). PI3K inhibitors in inflammation, autoimmunity and cancer. Current Opinion in Pharmacology, 23, 82-91. http://dx.doi.org/10.1016/j.coph.2015.05.017

M AN U

Starling, C. M., Prado, C. M., Leick-Maldonado, E. A., Lancas, T., Reis, F. G., Aristoteles, L. R., ... and Tiberio, I. F. (2009). Inducible nitric oxide synthase inhibition attenuates lung tissue responsiveness and remodeling in a model of chronic pulmonary inflammation in guinea pigs. Respiratory Physiology and Neurobiology, 165 (2), 185-194. http://dx.doi.org/10.1016/j.resp.2008.11.011 Stinson, S. E., Amrani, Y., and Brightling, C. E. (2015). D prostanoid receptor 2 (chemoattractant receptor–homologous molecule expressed on T H 2 cells) protein expression in asthmatic patients and its effects on bronchial epithelial cells. Journal of Allergy and Clinical Immunology, 135 (2), 395-406. http://dx.doi.org/10.1016/j.jaci.2014.08.027

TE D

Stoll, M. L., Solomon, D. H., Batra, K. L., Simard, J. F., Karlson, E. W., Dellaripa, P. F., Weinblatt, M. E., Glass, R. and Shadick, N. A. (2009). TNFα inhibitors may improve asthma symptoms: a case series of 12 patients with rheumatoid arthritis and asthma. Journal of Clinical Rheumatology, 15 (4), 198-200. http://dx.doi.org/10.1097/rhu.0b013e3181a7ace9

EP

Sylvin, H., Dahlbäck, M., Van Der Ploeg, I. and Alving, K. (2002). The tryptase inhibitor APC‐366 reduces the acute airway response to allergen in pigs sensitized to Ascaris suum. Clinical and Experimental Allergy, 32 (6), 967-971. http://dx.doi.org/10.1046/j.13652222.2002.01239.x

AC C

Taki, F., Kume, H., Kobayashi, T., Ohta, H., Aratake, H. and Shimokata, K. (2007). Effects of Rho‐kinase inactivation on eosinophilia and hyper‐reactivity in murine airways by allergen challenges. Clinical and Experimental Allergy, 37 (4), 599-607. http://dx.doi.org/10.1111/j.1365-2222.2007.02693.x Tanday, S. (2015). Calcium-sensing receptors linked to development of asthma. The Lancet Respiratory Medicine, 3(6), 428. http://dx.doi.org/10.1016/s2213-2600(15)00193-9 Tesmer, L. A., Lundy, S. K., Sarkar, S., & Fox, D. A. (2008). Th17 cells in human disease. Immunological reviews, 223(1), 87-113. Thurmond, R. L., Chen, B., Dunford, P. J., Greenspan, A. J., Karlsson, L., La, D., ... and Xu, X. L. (2014). Clinical and preclinical characterization of the histamine H4 receptor antagonist JNJ-39758979. Journal of Pharmacology and Experimental Therapeutics, 349 (2), 176-184. http://dx.doi.org/10.1124/jpet.113.211714

48

ACCEPTED MANUSCRIPT To, T., Stanojevic, S., Moores, G., Gershon, A. S., Bateman, E. D., Cruz, A. A., and Boulet, L. P. (2012). Global asthma prevalence in adults: findings from the cross-sectional world health survey. BMC Public Health, 12 (1), 204. http://dx.doi.org/10.1186/1471-2458-12-204 Tobinick, E. L. (2009). The value of drug repositioning in the current pharmaceutical market. Drug News and Perspective, 22 (2), 119-125. http://dx.doi.org/10.1358/dnp.2009.22.2.1343228

RI PT

Tong, L., and Tergaonkar, V. (2014). Rho protein GTPases and their interactions with NFkappaB: crossroads of inflammation and matrix biology. Bioscience Reports, 34 (3), 283295. http://dx.doi.org/10.1042/bsr20140021 Trian, T., Burgess, J. K., Niimi, K., Moir, L. M., Ge, Q., Berger, P., ... and Oliver, B. G. (2011). β2-Agonist induced cAMP is decreased in asthmatic airway smooth muscle due to increased PDE4D. PloS ONE, 6 (5) e20000. http://dx.doi.org/10.1371/journal.pone.0020000

M AN U

SC

Troy, N. M., Hollams, E. M., Holt, P. G., and Bosco, A. (2016). Differential gene network analysis for the identification of asthma-associated therapeutic targets in allergen-specific Thelper memory responses. BMC Medical Genomics, 9 (1), 9. http://dx.doi.org/10.1186/s12920-016-0171-z Turowska, A., Librizzi, D., Baumgartl, N., Kuhlmann, J., Dicke, T., Merkel, O., ... and Garn, H. (2013). Biodistribution of the GATA-3-specific DNAzyme hgd40 after inhalative exposure in mice, rats and dogs. Toxicology and Applied Pharmacology, 272 (2), 365-372. http://dx.doi.org/10.1016/j.taap.2013.06.020 Vale, K. (2016). Targeting the JAK-STAT pathway in the treatment of 'Th2-high' severe asthma. Future Medicinal Chemistry, 8 (4), 405-419. http://dx.doi.org/10.4155/fmc.16.4

TE D

Vatrella, A., Fabozzi, I., Calabrese, C., Maselli, R., and Pelaia, G. (2013). Dupilumab: a novel treatment for asthma. Journal of Asthma and Allergy, 7, 123-130. http://dx.doi.org/10.2147/jaa.s52387

EP

Velat, G. J., Kimball, M. M., Mocco, J. D., and Hoh, B. L. (2011). Vasospasm after aneurysmal subarachnoid hemorrhage: review of randomized controlled trials and metaanalyses in the literature. World Neurosurgery, 76 (5), 446-454. http://dx.doi.org/10.1016/j.wneu.2011.02.030

AC C

Verini, M., Consilvio, N. P., Di Pillo, S., Cingolani, A., Spagnuolo, C., Rapino, D., ... and Chiarelli, F. (2010). FeNO as a marker of airways inflammation: the possible implications in childhood asthma management. Journal of Allergy, 2010, 1452-1453. http://dx.doi.org/10.1155/2010/691425 Virchow, J. C., Backer, V., Kuna, P., Prieto, L., Nolte, H., Villesen, H. H., ... and de Blay, F. (2016). Efficacy of a house dust mite sublingual allergen immunotherapy tablet in adults with allergic asthma: a randomized clinical trial. Jama, 315(16), 1715-1725. http://dx.doi.org/10.1001/jama.2016.3964 Vliagoftis, H., Schwingshackl, A., Milne, C. D., Duszyk, M., Hollenberg, M. D., Wallace, J. L., ... and Moqbel, R. (2000). Proteinase-activated receptor-2–mediated matrix metalloproteinase-9 release from airway epithelial cells. Journal of Allergy and Clinical Immunology, 106 (3), 537-545. http://dx.doi.org/10.1067/mai.2000.109058 Vonk, J. M., Postma, D. S., Maarsingh, H., Bruinenberg, M., Koppelman, G. H., and Meurs, H. (2010). Arginase 1 and arginase 2 variations associate with asthma, asthma severity and β2 49

ACCEPTED MANUSCRIPT agonist and steroid response. Pharmacogenetics and Genomics, 20 (3), 179-186. http://dx.doi.org/10.1097/fpc.0b013e328336c7fd Vroman, H., van den Blink, B., and Kool, M. (2015). Mode of dendritic cell activation: the decisive hand in Th2/Th17 cell differentiation. Implications in asthma severity? Immunobiology, 220 (2), 254-261. http://dx.doi.org/10.1016/j.imbio.2014.09.016

RI PT

Wang, L., Pirozzi, G., Sutherland, E. R., Graham, N., Evans, R., Teper, A., and Wenzel, S. E. (2015). Dupilumab improves lung function and reduces severe exacerbations in uncontrolled asthmatics with baseline eosinophil levels above and below 300 cells/µL. American Journal of Respiratory and Critical Care Medicine, 191, A6362. http://dx.doi.org/10.1183/13993003.congress-2015.oa289 Wenzel, S. E. (2012). Asthma phenotypes: the evolution from clinical to molecular approaches. Nature Medicine, 18 (5), 716-725. http://dx.doi.org/10.1038/nm.2678

SC

Wenzel, S. E. (2013a). Complex phenotypes in asthma: current definitions. Pulmonary Pharmacology and Therapeutics, 26 (6), 710-715. http://dx.doi.org/10.1016/j.pupt.2013.07.003

M AN U

Wenzel, S. E., Barnes, P. J., Bleecker, E. R., Bousquet, J., Busse, W., Dahlén, S. E., ... and Baker, J. (2009). A randomized, double-blind, placebo-controlled study of tumor necrosis factor-α blockade in severe persistent asthma. American Journal of Respiratory and Critical Care Medicine, 179(7), 549-558. http://doi.org/10.1164/rccm.200809-1512OC

TE D

Wenzel, S., Castro, M., Corren, J., Maspero, J., Wang, L., Zhang, B., ... and Eckert, L. (2016). Dupilumab efficacy and safety in adults with uncontrolled persistent asthma despite use of medium-to-high-dose inhaled corticosteroids plus a long-acting β 2 agonist: a randomised double-blind placebo-controlled pivotal phase 2b dose-ranging trial. The Lancet, (in press). http://doi.org/10.1016/S0140-6736(16)30307-5

EP

Wenzel, S. E., Hopkins, R., Saunders, M., Chantry, D., Anderson, L., Aitchison, R., ... and Spector, S. L. (2014). Safety and Efficacy of ARRY-502, a Potent, Selective, Oral CRTh2 Antagonist, In Patients With Mild To Moderate Th2-Driven Asthma. The Journal of Allergy and Clinical Immunology, 2(133), AB4. http://dx.doi.org/10.1016/j.jaci.2013.12.037

AC C

Wenzel, S., Ford, L., Pearlman, D., Spector, S., Sher, L., Skobieranda, F., ... and Pirozzi, G. (2013b). Dupilumab in persistent asthma with elevated eosinophil levels. New England Journal of Medicine, 368 (26), 2455-2466. http://dx.doi.org/10.1056/nejmoa1304048 Wenzel, S., Wilbraham, D., Fuller, R., Getz, E. B., and Longphre, M. (2007). Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. The Lancet, 370 (9596), 1422-1431. http://dx.doi.org/10.1016/s0140-6736(07)61600-6 Wilson, R. H., Maruoka, S., Whitehead, G. S., Foley, J. F., Flake, G. P., Sever, M. L., ... and Cook, D. N. (2012). The Toll-like receptor 5 ligand flagellin promotes asthma by priming allergic responses to indoor allergens. Nature Medicine, 18 (11), 1705-1710. http://dx.doi.org/10.1038/nm.2920 Woo, Y., Jeong, D., Chung, D. H., and Kim, H. Y. (2014). The roles of innate lymphoid cells in the development of asthma. Immune Network, 14 (4), 171-181. http://dx.doi.org/10.4110/in.2014.14.4.171

50

ACCEPTED MANUSCRIPT Xie, T., Luo, G. Y., Zhang, Y., Wang, X., Wang, X. Y., Wu, M., and Li, G. P. (2015). Rho‐ kinase inhibitor fasudil reduces allergic airway inflammation and mucus hypersecretion by regulating STAT6 and NFκB. Clinical and Experimental Allergy, 45 (12), 1812-1822. http://dx.doi.org/10.1111/cea.12606

RI PT

Yang, M., Rangasamy, D., Matthaei, K. I., Frew, A. J., Zimmmermann, N., Mahalingam, S., ... and Foster, P. S. (2006). Inhibition of arginase I activity by RNA interference attenuates IL-13-induced airways hyperresponsiveness. The Journal of Immunology, 177 (8), 55955603. http://dx.doi.org/10.4049/jimmunol.177.8.5595 Zeng, G., Chen, J., Liang, Q. H., You, W. H., Wu, H. J., and Xiong, X. G. (2012). Ursolic acid inhibits T-cell activation through modulating nuclear factor-κ B signaling. Chinese Journal of Integrative Medicine, 18, 34-39. http://dx.doi.org/10.1007/s11655-011-0858-0

SC

Zhang, M., Thurmond, R. L., and Dunford, P. J. (2007). The histamine H 4 receptor: a novel modulator of inflammatory and immune disorders. Pharmacology and Therapeutics, 113 (3), 594-606. http://dx.doi.org/10.1016/j.pharmthera.2006.11.008

AC C

EP

TE D

M AN U

Zhou, W. and Nie, X. (2015). Afzelin attenuates asthma phenotypes by downregulation of GATA3 in a murine model of asthma. Molecular Medicine Reports, 12 (1), 71-76. http://dx.doi.org/10.3892/mmr.2015.3391

51