Review

1.

Introduction

2.

Asthma and allergic rhinitis: similarities and differences through guidelines

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

3.

Advances in asthma drug discovery: evaluating the potential of nasal cell sampling and beyond Luigino Calzetta, Paola Rogliani, Mario Cazzola† & Maria Gabriella Matera †

University of Rome Tor Vergata, Department of System Medicine, Rome, Italy

Nasal cells sampling and procedures for novel anti-inflammatory agents development

4.

Conclusion

5.

Expert opinion

Introduction: Inhaled corticosteroid anti-inflammatory therapy is effective at controlling disease symptoms of asthma, but a subset of patients remains symptomatic despite optimal treatment, creating a clear unmet medical need. Moreover, none of the currently available drugs for asthma are really disease-modifying or curative. Although murine models of asthma, based on transgenic and knockout animals, may offer an integrated pathophysiological system for studying the characteristics of airway inflammation and hyperresponsiveness, these alterations are noteworthily different compared with those observed in asthmatic patients. Since a clear functional and inflammatory relationship between the nasal mucosa and bronchial tissue in patients suffering from asthma and allergic rhinitis has been recognized, using preclinical models based on human nasal cells sampling might support a prompt and effective anti-inflammatory drug discovery in asthma. Areas covered: The authors provide a review, which discusses the potential role of nasal cell sampling and its application in advanced drug discovery for asthma. The contents range from the similarities and differences between asthma and allergic rhinitis up to artificial airway models based on sophisticated human lung-on-a-chip devices. Expert opinion: Nasal cell sampling and processing have reached a great potential in asthma drug discovery. The authors believe that models of asthma, which are based on human nasal cells, can provide valuable indications of proof of pharmacological and potential therapeutic efficacy in both preclinical and early clinical settings. Keywords: allergic rhinitis, artificial airways, asthma, inflammation, nasal cells Expert Opin. Drug Discov. (2014) 9(6):595-607

1.

Introduction

For the majority of asthmatic patients, inhaled corticosteroid anti-inflammatory therapy is effective at controlling disease symptoms, but a subset of patients remain symptomatic despite optimal treatment, creating a clear unmet medical need [1]. Moreover, none of the currently available treatments for asthma have long-term effects on airway inflammation or remodeling, and therefore, are not disease-modifying or curative [2]. Clearly, there is an urgent need for targeted, disease-modifying asthma treatments. However, the development of novel therapy for asthma has proved disappointing despite intense effort and investment. An over-reliance on animal models of allergy to define targets and expectations of efficacy, a fundamental disconnect between the directions of basic research and clinical research and the different features of asthma, bronchoconstriction, symptoms and exacerbations, which respond diversely to treatment, are three 10.1517/17460441.2014.909403 © 2014 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X All rights reserved: reproduction in whole or in part not permitted

595

L. Calzetta et al.

.

.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

.

.

.

None of the currently available drugs for asthma are really disease-modifying or curative and a subset of asthmatic patients remains symptomatic despite optimal treatment creating a clear unmet medical need. An overestimation of murine models explains why the use of animal models of asthma by pharmaceutical companies has been disappointing in term of the lack of generation of novel treatments for asthma. A substantial functional and inflammatory relationship between the nasal mucosa and bronchial tissue in patients suffering from asthma and allergic rhinitis has been recognized. Standardized and sophisticated models of asthma based on human nasal cell challenge techniques and sensitive biomarker assays permit to effectively investigate the efficacy of novel anti-inflammatory drugs for asthma in preclinical settings. Nasal cell sampling and processing can reach the greatest potential in asthma drug discovery when adequately implemented with passive sensitization of the airway smooth muscle collected from low airway.

This box summarizes key points contained in the article.

particular issues that contribute to the challenge of identifying new therapeutics [3]. Animal models based on transgenic and knockout mice may offer an integrated pathophysiological system in which to study the characteristics of inflammation and airway alterations that are similar, but not equal, to conditions of asthmatic patients [4,5]. Unfortunately, animal models fail to reproduce all of the features of human asthma since there are noteworthy differences between human and murine models toward TH2-type inflammatory pathways, the lung structure and its development. Mice fail also to develop spontaneous symptoms, longlasting bronchoconstriction and inflammatory alterations in airway wall behavior as observed in asthmatic patients [6,7]. Access to human airway tissue has allowed development of ex vivo and in vitro models of the human airway that can be used for mechanistic studies, target identification and validation and toxicological testing [6]. These models are mainly based on cultured cells obtained from human airway tissue collected from volunteers at bronchoscopy as small biopsy samples, from lung resections as surgical waste, or postmortem [6,8]. Evidently, collecting these respiratory cells from bronchi is much more invasive and less accessible compared with harvesting respiratory cells from nasal mucosa (Figure 1). Therefore, this review will focus on the potential role of nasal cell sampling and its application for advanced drug discovery in asthma, from similarities and differences between asthma and allergic rhinitis up to artificial airway models based on sophisticated human lung-on-a-chip devices. 596

Asthma and allergic rhinitis: similarities and differences through guidelines

2.

Article highlights.

The Allergic Rhinitis and its Impact on Asthma (ARIA) initiative, which aims to educate and implement evidence-based management of allergic rhinitis in conjunction with asthma, and the Global Initiative for Asthma (GINA), with the purpose of reducing asthma prevalence, morbidity and mortality, represent the two worldwide initiative that allow understanding the complementarities between asthma and allergic rhinitis [9,10]. The analogies between these disorders have been suggested from the launch of these initiatives (GINA in 1993 and ARIA in 2001), and the subsequent reports and guidelines have increasingly highlighted the strict association between allergic rhinitis and asthma. To date, the most recent updated ARIA and GINA reports agree that asthma and allergic rhinitis are common comorbidities. The majority of asthmatic patients have evidence of allergic rhinitis and a significant number of patients suffering from persistent rhinitis have asthma. Both rhinitis and asthma share the same epidemiological, pathological and physiologic characteristics and are also linked by common risk factors and therapeutic approach [11,12]. A recent population-based epidemiological survey has been conducted in agreement with definitions suggested by the European Position Paper on Rhinosinusitis and Nasal Polyps [13]. This study, aimed to investigate the prevalence of asthma and its association with chronic rhinosinusitis (CRS), has demonstrated that, although geographical variation in the prevalence of asthma (5.1 -- 16.8%) was observed across Europe, asthma was more common in young adults, women and smokers and a significant association of asthma with CRS has been detected (adjusted odds ratio: 3.5) at all ages. Furthermore, the association with asthma was stronger in patients reporting both CRS and allergic rhinitis (adjusted odds ratio: 11.9) [14]. Therefore, asthma has to be suspected in patients with allergic rhinitis and asthmatic patients should be evaluated for rhinitis and, consequently, the therapeutic plans should be considered together [15]. These findings are supported by the evidence that both asthma and allergic rhinitis can be considered to be inflammatory disorders of the airways and, thus, the concept of ‘one airway, one disease’ has been suggested, and even the term ‘allergic rhinobronchitis’ has been proposed, although it has been not generally accepted [16-18]. Nevertheless, in spite of the clear association between asthma and allergic rhinitis, some distinctive characteristics may be identified such as the pathophysiological mechanisms, the clinical features and the therapeutic approach. Mainly, the nasal obstruction is essentially due to hyperaemia in rhinitis, while the enhanced bronchial contraction in asthma is predominantly mediated by the airway smooth muscle (ASM) hyperresponsiveness [19].

Expert Opin. Drug Discov. (2014) 9(6)

Accuracy of specimens for asthma models

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

Advances in asthma drug discovery

Local anesthesia not required

Local (nasal) anesthesia required

Local (bronchial) anesthesia required

Very easy procedures

Easy procedures

Specialist procedure Bronchial biopsy

Nasal biopsy

Nasal scraping Nasal brushing

Filter paper strips Nasal lavage Invasivity

Figure 1. Invasivity of procedures for collecting samples from respiratory airways and relative accuracy of the obtained specimens for preclinical human-based models of asthma.

However, treating allergic rhinitis decreases asthma symptoms in asthmatic patients and vice versa. Intranasal administration of corticosteroids, cromones, leukotriene (LT) modifiers, anticholinergics, the allergen-specific immunotherapy and the anti-IgE therapy are effective in both conditions via reducing the bronchial hyperresponsiveness (BHR), protecting against exacerbations of asthma and improving symptoms of rhinitis [15,20-26]. Unfortunately, there are not solid evidences on the real effectiveness of intranasal corticosteroids in improving asthmatic symptoms and, effectively, some clinical trials have shown conflicting results [27,28]. On the other hand, others drugs such as H1-antagonists and b2-agonists are selectively effective for rhinitis and asthma, respectively [20,21,29]. Compared with other treatments, allergen-specific immunotherapy represents the only treatment working on the causes of allergy and, therefore, this therapeutic approach has been recommended by ARIA guidelines for treating allergic rhinitis, although its role in adult asthma is limited [11,30]. Allergenspecific immunotherapy showed the potential of preventing asthma in children with allergic rhino-conjunctivitis, but the modest benefits must be weighed against the risk of adverse events [11]. However, there are findings suggesting that allergen-specific injection immunotherapy is highly effective in IgE-mediated diseases, via inhibiting both early and late responses to allergen exposure. Immunotherapy increases allergen-specific IgG, blocks IgE-dependent histamine release from basophils and also IgE-mediated antigen presentation to T cells. Furthermore, immunotherapy modifies peripheral and mucosal TH2 responses to allergen in favor of TH1 responses [31]. In fact, allergen-specific immunotherapy might

improve seasonal asthma and inhibit seasonal increases in BHR [32]. In any case, the evidence-based medicine moves toward managing allergic rhinitis and asthma as different manifestations of a single airway disease, rather than as two separate diseases of the nose and the lung and, therefore, combining the treatment of both these upper and lower airway conditions seems to be desirable [15,33]. In fact, the 2010 revision of ARIA clearly displayed drugs recommended for improving asthma in patients with concomitant allergic rhinitis and asthma [34]. Altogether these findings support the effort for using experimental models based on nasal cells sampling for a prompt and effective anti-inflammatory drug discovery in asthma.

Nasal cells sampling and procedures for novel anti-inflammatory agents development

3.

The nose represents the part of respiratory system, which is most easily accessible for morphological and pathophysiological investigations of inflammatory response to various stimuli. Therefore, considering the aforementioned correlation between inflammatory response in bronchi and nasal mucosa, collecting nasal lining fluid, mucous, cells and mucosa may represent a suitable approach for advanced modeling to be applied in novel anti-inflammatory drug discovery in asthma. Standardized and reproducible samples of both nasal exudates and mucosa cells can be obtained in a relatively noninvasive manner such as nasal lavage, filter paper strips, nasal

Expert Opin. Drug Discov. (2014) 9(6)

597

L. Calzetta et al.

brushing and scraping and nasal biopsy. All these sampling techniques may be performed before and after nasal allergen challenge (NAC) for comparing the inflammatory response, particularly in cells collected from patients suffering from allergic rhinitis. Nasal allergen challenge Recent findings have demonstrated bronchial inflammation after NAC and nasal inflammation after segmental bronchial provocation. These evidences focus on the critical link between nasal and bronchial inflammation and permit to suggest NAC a suitable model for investigating inflammatory response in asthma through nasal provocation in patients with allergic rhinitis [35-37]. NAC is performed by using solutions containing extracts of the most relevant clinical allergens. Usually, 50 -- 100 µl solution containing allergen extracts is delivered into each nostril by means of a pump spray. Acoustic rhinometry is used for detecting positive NAC reaction when the nasal volume reduction is > 30% and, subsequently, a second NAC can be performed up to 7 days after the first one [35,36]. NAC immediately induces symptoms of sneezing and itching, followed by rhinorrhea and nasal blockage. The late-phase reaction (LPR) is maximal between 6 and 24 h and resolves within 1 -- 3 days [38,39]. NAC permits to investigate novel approaches to therapy, such as the immunomodulation induced by recombinant hypoallergenic recombinant allergens. In fact, there are findings suggesting that allergen-specific immunotherapy can improve the tolerance of NAC via modifying the cellular inflammatory response assessed in nasal biopsies and the IgG levels detected in nasal lavage [40]. Furthermore, NAC allowed investigating the sublingual route of allergen-specific immunotherapy as a suitable alternative to the inoculation route for immunomodulatory therapy [41,42].

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

3.1

Methodologies for nasal cells sampling and processing 3.2.1 Nasal lavage 3.2

Nasal lavage consists in the introduction of 2.5 -- 5.0 ml of 0.9% NaCl physiologic solution at 37 C into the nasal cavity and its recovery after 10 s. Usually, the recovery of fluid ranges between 65 and 90% [43]. In order to decongest the nose mucosa, the intranasal instillation of an a-agonist such as oxymetazoline, a method that does not affect the quantification of mediators or biomarkers, might be necessary [44]. After that, the nasal lavage is refrigerated at 4 C and centrifugated at 400 -- 1000 g for 5 -- 20 min. The supernatant can be stored at -20 to -80 C before being analyzed and the pellet has to be resuspended in 0.5 ml PBS containing 0.1% human serum albumin for cytometry and immunostaining of cells from the nasal lining fluid [39]. Mast cells, basophils and eosinophils are normally tissue-resident cells that can be found in nasal lavage as a 598

consequence of airway recruitment due to allergic process [39]. Nasal lavage performed in subject with allergic rhinitis evidenced detectable-concentration inflammatory biomarkers such as histamine, tryptase, prostanoids, prostaglandin D2, eosinophil cationic protein, eosinophil peroxidase cysteinyl LTs, LTC4, LTD4 and LTE4, and these inflammatory biomarkers were particularly elevated after consecutive NAC [39,45-49]. On the other hand, there are conflicting evidences concerning the relevance of cytokines and chemokines, such as such as IL-3, IL-4, IL-5 and eotaxin, regulated on activation, normal T cell expressed and secreted (RANTES) and granulocytemacrophage colony-stimulating factor (GM-CSF) discerning between naturally occurred disease and NAC [50-53]. However, the concentrations of inflammatory markers are usually significantly higher in nasal lavage fluid of patients suffering from allergic rhinitis compared with normal subjects as control [54]. The validity of nasal lavage fluid analysis for assessing novel anti-inflammatory therapy in allergic rhinobronchitis has been proved by the finding that the pretreatment with topical glucocorticosteroids inhibited the inflammatory mediator release in allergic rhinitis after NAC [55]. Filter paper strips Another effective technique for recovering cytokines from inflammatory mucosa in situ is represented by the use of filter paper strips. The strips are placed on nasal turbinates for 10 min in order to adsorb nasal secretion. After that, the strips have to be washed in Hepes buffer containing 0.3% human serum albumin [56]. This method may be performed consecutively at different time after NAC in order to quantify the inflammatory response assessing inflammatory mediators. The filter paper strip method has been validated for detecting chemokines and cytokines such as IL-1b, IL-5, IL-6, IL-8, macrophage inflammatory protein-1 a, RANTES and GM-CSF at different time [56,57]. Studies on the role of cytokines during LPR have established a significant correlation between decreases in cytokine levels and symptoms after topical administration of corticosteroids in patients with allergic rhinitis, even after a single dose pretreatment [52,58]. Compared with the nasal lavage, the nasal sampling by absorption into filter paper strips gives information on both intracellular and extracellular inflammatory pathways, since the cells adhering on the paper surface may lyse and release their intracellular contents [59]. Furthermore, the filter paper strip method prevents the dilution of secretions caused by nasal lavage and, therefore, allows collecting proteins at detectable concentrations, which might be below the detection limits after nasal lavage [39]. 3.2.2

Nasal brushing and scraping Nasal cytology may be performed on samples obtained from nasal brushing and scraping. Nasal brushing can be carried out without local anesthesia. A small brush made with nylon 3.2.3

Expert Opin. Drug Discov. (2014) 9(6)

Advances in asthma drug discovery

Table 1. Specific techniques for staining cytology and histology specimens from nasal cavity. Stained cells

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

Eosinophils Basophils Epithelial cells, nuclear and cytoplasmic changes Mast cells Neutrophils Eosinophils, neutrophils and basophils

Staining technique Hansel, Leishman and Randolph Wright and toluidine blue Papanicolaou Alcian yellow May--Grunwald Wright--Giemsa

strings is gently introduced into the nasal cavity under direct visual guidance and placed between the nasal septum and the inferior turbinate. The brush is slightly rotated while being removed and then it is washed with physiological buffer solution to tear out harvested cells [60]. Harvested cells are those floating in the nasal cavity and are mainly represented by epithelial cells. Therefore, the sample significantly reflects the cell pool involved in the barrier function of the mucous membrane [61]. Nasal scraping is performed by using a small steal or plastic curette and it allows to obtain well-preserved chunks of epithelial layer. This technique is useful for studying epithelial cells, goblet cells and granulocytes but not for cells located in deeper layer of mucosa. Also, nasal scraping does not require anesthesia and permit to obtain repeatable epithelial specimens from specific areas of nasal cavity [60]. Cells obtained with these methods can be used for morphological assays, biochemical evaluations and molecular investigations. Although nasal brushing and scraping allow evaluating the nasal inflammatory response, there are conflicting evidences on their role in adequately investigating the inflammatory processes that involve the whole thickness of the nasal mucosa. Effectively, nasal biopsy permits to study the cellular inflammatory response to challenge in all layers of the nasal mucosa [19]. Nevertheless, there are findings suggesting that well-performed nasal brushing harvests an adequate number of cells from the surface of the nasal mucosa to be used in lieu of nasal biopsies for detecting the increase of eosinophils and mast cells and for investigating the effectiveness of topical corticosteroids in patients with allergic rhinitis [62]. Furthermore, in addition to cell harvesting, nasal brushing can also provide for supernatant suitable for the quantification of pro-inflammatory cytokines [63]. Nasal biopsy Nasal biopsy is an invasive procedure that requires adequate local anesthesia achieved by applying a cotton wool plug soaked in 10% cocaine and 0.025% adrenaline below the inferior turbinate for 10 min [39,59]. It is a well-tolerated practice if it is carried out by a specialist. Although biopsy can be obtained from different locations of nasal mucosa, 3.2.4

the most common site for carrying out the biopsy is the undersurface of the inferior turbinate, just posterior to its anterior insertion to the lateral wall of the nose. This permits to standardize the sampling since significant differences in the histology of the nasal mucosa have been documented [39,64]. Biopsies including mucosal epithelium with basement membrane and submucosal tissue can be obtained under direct vision with nasal biopsy forceps [65]. The handling of small respiratory biopsies presents some specific issues such as tissue fixation, embedding and analysis that has been extensively reviewed by Jeffery and colleagues [66]. The main advantage of the nasal biopsy technique, compared with nasal brushing method, is the opportunity of studying the T and B lymphocyte response. In fact, lymphocytes tend to compartmentalize to tissue rather than migrate into the nasal lumen. Furthermore, nasal biopsy offers the opportunity to culture T cells directly from the nasal compartment and to successively investigate the inflammatory cellular response to various stimuli and the influence of novel anti-inflammatory medications on the activation and migration of lymphocytes [39,64]. While nasal brushing is suitable for studies looking at the effect of treatment on inflammatory cells within the epithelium or on epithelial cells themselves in studies involving children, large groups or multiple measures, the nasal biopsy provides a whole and comprehensive representation of the nasal mucosa and of the residential inflammatory cells. Therefore, the sampling method selected for a given study would depend on the number of requested samples and whether results obtained from epithelial layer are satisfactory [39]. 3.3

Cytology and histology evaluation Nasal cytology

3.3.1

Adequate specimens for nasal cytology may be obtained from nasal lavage, filter paper strips and nasal brushing and scraping. Cytological qualitative, quantitative and semiquantitative analysis permits to characterize the mucosal inflammatory response following specific allergenic challenge and to assay the effectiveness of treatments. In fact, the measurement of nasal cellular modifications reflects the inflammatory response of less-accessible regions of the respiratory system, such as the bronchial tree, which are involved as part of a common mucosal response in allergic and inflammatory disease [39]. As shown in Table 1, specific staining may evidence for different cells involved in inflammatory response of airways. In normal subjects, there are not significant difference in normal nasal cytology among infants, children and adults. Usually, both ciliated and goblet cells but not eosinophils and basophils are detected, although a limited number of neutrophils and bacteria may be identified [67,68]. In patients suffering from allergic rhinitis, nasal specimens contain significantly more total cells compared to healthy subjects as reported in Table 2. Although the number of basophilic cells usually correlates with nasal eosinophilia in allergic rhinitis, the nasal reactivity to NAC may also increase without a concomitant enhancement

Expert Opin. Drug Discov. (2014) 9(6)

599

L. Calzetta et al.

Table 2. Quantitative and semi-quantitative characteristics of cytology from healthy subjects and patients suffering from allergic rhinitis. Cells per HPF (103)

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

Normal

Semi-quantitative assay

Allergic rhinitis

Normal

Eosinophils

0

from 1 to >20

ND

Basophils Neutrophils

0 from 0 to 5

from 0.4 to >6 from 1 to >20

ND From ND to few scattered cells or small clumps

Allergic rhinitis From few scattered cells to large clumps of cells covering the entire field From few scattered cells to large number of cells From few scattered cells to large clumps of cells covering the entire field

HPF: High-power field; ND: Not detectable.

in the number of surface basophilic cells [39]. In any case, a significant increase in the number of basophilic cells of nasal lavage has been confirmed at LPR post-NAC in patients with allergic rhinitis, which has been prevented, together with the histamine release, by the pretreatment with topic administration of corticosteroids [69]. Both eosinophilic and basophilic scores are suitable for evaluating the anti-inflammatory effectiveness of novel compounds, as previously assessed by independent studies that have also ruled out the significance of neutrophils and goblet cells as cellular markers of LPR [70-72]. In addition, eosinophil and basophil influx may be used for investigating the effectiveness of long-term immunotherapy [73-75]. Nasal cytology might be also useful for validating innovative approaches to immunotherapy represented by newly designed adjuvants, such as monophosphoryl lipid A or bacteria-derived nucleotide immunostimulatory sequences, which improve the efficacy and safety of allergen immunotherapy, and for investigating alternative therapeutic strategies including the administration of allergen-derived peptides or modified recombinant allergen vaccines [31,76]. Nasal histology evaluation Immunohistochemistry of nasal biopsy allows an easy approach for amplifying the signal, identifying antibody binding to the tissue antigens and, thus, staining lymphocytes, mast cells and eosinophils. Since the image analysis is performed on two-dimensional sections, a bias in favor of higher cell counts compared with larger cells exists. However, nowadays both commercial and open-source automating cell-counter software exist that use digital imaging analysis in order to standardize the counting procedure and to normalize the image-derived data [77-80]. These softwares detect cell phenotypes in a high-throughput manner by simultaneously measuring the size, shape, intensity and texture. In situ hybridization (ISH) is a technique that enables the monitoring of gene expression in individual cells [81]. ISH is used with light microscope and it is sensitive for detecting mRNAs that accumulate in only a small population of cells within an organ [82]. ISH studies carried out for profiling cytokines of nasal biopsies evidenced an increasing of 3.3.2

600

mRNA for IL-4, IL-5, IL-13 and GM-CSF, suggesting an imbalance in T-cell cytokine production in favor of a TH2-type response in patients suffering from allergic rhinitis [83,84]. Radiolabeled RNA probes are preferred for amplifying the staining of genes that are low expressed, while nonradiolabeled probes are adequately sensitive for genes that are expressed at a high level. However, non-traditional methods of probe synthesis, quantification and detection of hybridization have been proposed for appreciably improving the detection levels of non-radioactive ISH. In fact, the use of both PCR-generated templates for the synthesis of probes and the antibody sandwich technique increase the sensitivity of detection, thus reducing substrate processing time, thereby maintaining tissue morphology and providing good signal localization of non-radioactive ISH in human airway tissue [85]. Nasal cells and artificial airways Recently, several models have been proposed for the production in vitro of artificial bronchial wall by using airway tissues from bronchial biopsies, lung resections during lobectomy and postmortem [6]. Considering the significant functional and immunological relationship in terms of infiltrating leucocytes, inflammatory mediators and response to NAC between nasal and bronchial tissues, nasal sampling might be suitable for modeling artificial airways for investigating novel anti-inflammatory strategies for asthma [17,86,87]. Nasal sampling may provide for epithelial cells, fibroblasts, endothelial cells, dendritic cells, mast cells, eosinophils, basophils, macrophages, T cells and natural killer cells depending on the method used for obtaining the specimens. Residential nasal cells collected by nasal lavage, filter paper strips technique, brushing and scraping and biopsy can be grown as primary cultures and used for developing artificial airways. Monolayer cultures of both epithelial cells and fibroblasts and pseudostratified cultures of epithelial cells containing also goblet and ciliated cells can be used as monocultures [88-90]. Air--liquid interface (ALI) technique permits to closely mimic in vitro the respiratory mucosa. In fact, the availability of a huge selection of permeable supports based on 3.4

Expert Opin. Drug Discov. (2014) 9(6)

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

Advances in asthma drug discovery

microporous membrane filters, customizable for pore size, pore density and chemical compatibility, allows modeling the three-dimensional structure of airway wall [91-93]. These models are based on co-cultures of fibroblasts and/or endothelial cells submerged at the liquid side of the system, which are interfaced with the respiratory epithelial cell layer at the air-media side through permeable supports [6,94,95]. Further complex co-cultured ALI models include also immune cells incorporated into the epithelial layer [6]. Recently several studies, carried out by ALI technique with epithelial cells isolated from patients suffering from CRS with and without nasal polyps, have permitted to investigate the relationship between the microenvironmental milieu and the host innate immunity response, the role of oxidative stress in inducing unfolding protein response and the detrimental role of interleukins in altering the sinonasal epithelial barrier [96-98]. Although these techniques are usually performed with primary respiratory cells, there are findings evidencing that experiments with ALI methods may be carried also employing immortalized cellular lines such as IMR-90 human fetal lung fibroblasts and the epithelial cell line 16HBE14o- [99,100]. Airway biopsies can be used itself as tissues explant models (TEMs) that are maintained in short-term culture for investigating the inflammatory response of respiratory mucosa [101-104], the regulatory inflammatory and immunomodulatory pathways in sinonasal mucosa of CRS patients with and without nasal polyps [105,106] and the ciliary dysfunction in subjects with CRS [107,108]. Although TEMs from nasal biopsy have the advantage of including almost all the components of the airways excluding ASM cells, the delivery of nutrients, the gas exchange and the absence of circulation within the tissue limit the length of time TEMs can be cultured for investigations of novel anti-inflammatory drugs [6]. New advances in human airway modeling, which potentially can use also cells from nasal sampling, are represented by the three-dimensional cell co-culture to organs-on-chips. This sophisticated technology requires microfabrication technologies from the microchip industry and microfluidics approaches to create cell-culture microenvironments that both support tissue differentiation and recapitulate the tissue--tissue interfaces, spatiotemporal chemical gradients and mechanical microenvironments of living organs [109]. Recently, the physiological functions and the three-dimensional microarchitecture of the whole human lung have been microengineered [109]. Human lung-on-a-chip devices constituted by airway epithelial cells and airway endothelial cells can be used for demonstrating the organ-specific features concerning airway closure and reopening, alveolar--capillary interface, surfactant production, lung inflammation and extrapulmonary absorption [110-112]. These systems may effectively mimic the human inflammatory response to cytokines and chemokines placed into the air channel and to human neutrophils that are introduced into the capillary channel. Therefore, lung-on-a-chip devices can be used to study absorption and effectiveness of novel anti-inflammatory drugs in preclinical studies [109,110].

4.

Conclusion

It has long been recognized that there is a substantial functional and inflammatory relationship between the nasal mucosa and bronchial tissue in patients suffering from asthma and allergic rhinitis [17,86,87]. The concept of considering asthma and allergic rhinitis as a single airway disease is in line with ARIA and GINA guidelines [11,12]. The evidence of an overestimation of murine models, even when based on transgenic and knockout mice, might explain why the use of animal models of asthma by pharmaceutical companies has been disappointing in term of the lack of generation of novel treatments for asthma [6,7,39]. This scenario offers the foundation for developing human-based models of airway that would permit to reproduce in vitro conditions of asthmatic airway. To date, several models of asthma based on cells collected from low airways are available, but a greater interest should be given also to models based on human nasal cell sampling. Effectively, nasal cells are easy to be collected and even the nasal biopsy, although being an invasive procedure, may be well tolerated when compared with small bronchial biopsy. Nasal harvesting provides for a wide range of samples such as nasal lavage, epithelial cells, connective cells, inflammatory cells and specimens of mucosa in toto. The choice of the needed specimen and, consequently, the technique for collecting cells and tissue depends on the number of samples requested for the studies and by the endpoints of the investigations to be carried out [39]. Approaching models of asthma with human nasal cells allow to investigate the effectiveness, the potency and the duration of action of novel anti-inflammatory drugs via NAC assay in subjects with allergic rhinitis, a condition that closely mimics the bronchial inflammatory response of asthmatic patients and that has been well standardized [35-39]. In addition to the classical studies based on cytological and histological evaluations of nasal cells and mucosa, to date very advanced and sophisticated models for artificial airways exist such as three-dimensional ALI and lung-on-chip models that, together with epithelium and endothelium, can incorporate also fibroblasts and inflammatory cells [91-93,109,110]. Intriguingly, the majority of cellular lines needed for these novel techniques can be obtained from nasal sampling, excluded for ASM cells. Concluding, nowadays the availability of well-standardized and sophisticated models of asthma based on human nasal cell challenge techniques, in association with the accessibility to sensitive biomarkers assays, permits to effectively investigate the efficacy of novel anti-inflammatory drugs for asthma in preclinical settings. 5.

Expert opinion

Traditionally, a drug discovery starts from the need to find new molecules able to act on a particular mechanism in a

Expert Opin. Drug Discov. (2014) 9(6)

601

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

L. Calzetta et al.

specific disease pathology to resolve adverse effects or to improve the effectiveness of medications. Strategies in discovering drugs have been modified over the years, from exploiting observations to rational drug design based on hormone/ neurotransmitter receptor interactions, to high-throughput screening and knowledge of the human genome. In the early days of drug discovery, pharmacology provided both target identification and validation through the modification of mechanisms and the development of animal models that respond to therapies based on these mechanisms [113]. Therefore, usually a target has to be validated before investigating candidate compounds, assuming that this will work in man if the candidate results to be safe enough to take forward into the clinic [114]. In recent years, it has become increasingly important to get as much as possible information on clinical efficacy already in the early phases of drug development. Overcoming the obstacle of preclinical experimentation, the investigated compound can enter in the clinical Phase I trial and be administered for the first time in humans to evaluate its safety and pharmacokinetic. Overall, the objectives that a clinical trial on chronic airway diseases has to reach are to achieve normal lung function, maintain normal quality of life, prevent and treat the exacerbations, prevent mortality, relieve symptoms, improve exercise tolerance, improve health status and prevent disease progression. In particular, early clinical studies of asthma should demonstrate a drug’s anti-inflammatory and/or disease-modifying properties but, unfortunately, studies in patients are logistically and economically hampered and usually require large numbers of participants during long-term observations. In contrast, several validated models for airway inflammation and responsiveness in asthma are available and represent useful tools for drug efficacy studies, either in preclinical or in clinical studies [115]. To date, several concerns have been raised against murine models of asthma and, moreover, the human-based models of asthma have been primarily developed on fluids, cells and tissues collected from low airways [4-7,39]. Unfortunately, techniques for obtaining specimens from low airways remain limited in their widespread clinical application due to their invasive nature. On the other hand, the findings assessed in this review evidence that collecting human nasal specimens such as mucous, cells and mucosa needs of procedures that are easy to be performed and relatively noninvasive. This permits to obtain an elevated number of samples that can be processed before and after NAC, especially in patients suffering from allergic rhinitis. In addition, human specimens from nasal cavity may be used for applying the most innovative OMICS technologies, such as genomic, proteomic and metabolomic investigations that might offer a great potential in the future [116,117]. In this background of technological innovation, artificial three-dimensional ALI models of airways and lungon-a-chip micro-devices expand the capabilities of nasal cell 602

culture, providing valuable low-cost alternatives to animal and clinical studies for asthma drug discovery [110]. Effectively, recently, nasal epithelial cells and ALI cultures have been employed as a tool in the preclinical development of siRNAbased therapeutics for asthma, suggesting that curettage sampling has potential in both novel investigations of disease biomarker expression and early drug development [118]. There are a few studies that compared the similarities and differences between nasal and bronchial epithelial cells, especially in the response to pro-inflammatory stimuli. Comer and colleagues suggested that nasal epithelial cells collected from patients suffering from chronic obstructive pulmonary disease (COPD) cannot substitute for in vitro bronchial epithelial cells in airway inflammation studies [119], whereas findings obtained by McDougall and colleagues in a mixed population including subjects with history of wheeze, allergic rhinitis, asthma and COPD confirmed that nasal epithelial cultures might constitute an accessible surrogate for studying lower airway inflammation [13]. In addition, although further differences exist in the extent of eosinophilic inflammation of reticular basement membrane thickness and of the epithelium shedding between bronchi and nasal in patients with allergic rhinitis, we believe that the main distinction in using models of asthma based on nasal cells instead of bronchial cells is that the former cannot provide for ASM cells [86]. In fact, ASM alterations are the more striking structural changes in asthma, which are believed to be the major determinants of BHR and the increase of ASM mass correlates with asthma severity [120,121]. Furthermore, in addition to its contractile properties, ASM may contribute to the pathogenesis of asthma by increased proliferation, and by the expression and secretion of pro-inflammatory cytokines and mediators such as RANTES, GM-CSF and monocyte chemoattractant proteins 1, 2 and 3 [122]. Therefore, adequate airway models of asthma should not exclude ASM cells. This evidence suggests the necessity of integrating the models based on nasal cells with experiments performed with bronchial tissue although, unfortunately, collecting numerous and large bronchial specimens from asthmatic patients is a matter. Nevertheless, in the past years, several in vitro and ex vivo studies have demonstrated that bronchial tissue, ASM and epithelial cells collected from subjects undergoing lobectomy and passively sensitized with serum derived from atopic asthmatic subjects with high serum IgE levels represent a suitable model of asthma [123-129]. This approach allows to reduce the problem of the availability of bronchial samples from asthmatic patients. For a satisfactory preclinical asthma drug discovery, novel compounds should be assessed also on passively sensitized precision-cut lung slices. In particular, videomicrometry of small airway slices (bronchioles < 2 mm diameter and < 1 mm thickness), processed without the complications related to the use of confounding agarose gel to inflate the lung or complex parenchymal sections that have numerous

Expert Opin. Drug Discov. (2014) 9(6)

Advances in asthma drug discovery

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

contracting elements, might be a proper method for investigating the influence of inflammation on ASM cells hyperreactivity and, vice versa, the role of ASM cells on the inflammatory response in passively sensitized bronchioles [130,131]. Finally, nowadays, nasal cell sampling and processing have a great potential in asthma drug discovery. Especially when adequately implemented with nasal challenge models and passive sensitization of ASM cells collected from low airway, models of asthma based on human nasal cells can provide valuable indications of proof of pharmacological and potential Bibliography

therapeutic efficacy in both preclinical and early clinical settings.

Declaration of interest The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Papers of special note have been highlighted as either of interest () or of considerable interest () to readers.

9.

ARIA (Allergic Rhinitis and its Impact on Asthma) initiative. 2014. Available from: http://www.whiar.org

1.

Walsh GM. Novel cytokine-directed therapies for asthma. Discov Med 2011;11(59):283-91

10.

GINA (Global Initiative for Asthma). 2014. Available from: http://www. ginasthma.org

2.

Barnes PJ. New therapies for asthma: is there any progress? Trends Pharmacol Sci 2010;31(7):335-43

11.

3.

Mullane K. The increasing challenge of discovering asthma drugs. Biochem Pharmacol 2011;82(6):586-99

Global Initiative for Asthma, GINA Report, Global Strategy for Asthma Management and Prevention (update). 2012. Available from: http://ginasthma.org/local/uploads/files/ GINA_Report_March13.pdf

4.

Shapiro SD. Animal models of asthma: pro: allergic avoidance of animal (model [s]) is not an option. Am J Respir Crit Care Med 2006;174(11):1171-3

5.

6.

.

7.

.

8.

Shapiro SD. The use of transgenic mice for modeling airways disease. Pulm Pharmacol Ther 2008;21(5):699-701 Swindle EJ, Davies DE. Artificial airways for the study of respiratory disease. Expert Rev Respir Med 2011;5(6):757-65 A schematic revision of the most accessible techniques for producing artificial airways from respiratory cells for the study of respiratory diseases. Wenzel S, Holgate ST. The mouse trap: it still yields few answers in asthma. Am J Respir Crit Care Med 2006;174(11):1173-6; discussion 76-8 An editorial that evidences the pro and cons of using animal models in for asthma, evidencing that murine models fail to adequately reproduce all of the features of human asthma. Djukanovic R, Wilson JW, Lai CK, et al. The safety aspects of fiberoptic bronchoscopy, bronchoalveolar lavage, and endobronchial biopsy in asthma. Am Rev Respir Dis 1991;143(4 Pt 1):772-7

12.

13.

Bousquet J, Khaltaev N, Cruz AA, et al. Allergic Rhinitis and its Impact on Asthma (ARIA) 2008 update (in collaboration with the World Health Organization, GA(2)LEN and AllerGen). Allergy 2008;63(Suppl 86):8-160 McDougall CM, Blaylock MG, Douglas JG, et al. Nasal epithelial cells as surrogates for bronchial epithelial cells in airway inflammation studies. Am J Respir Cell Mol Biol 2008;39(5):560-8

14.

Jarvis D, Newson R, Lotvall J, et al. Asthma in adults and its association with chronic rhinosinusitis: the GA2LEN survey in Europe. Allergy 2012;67(1):91-8

15.

Bousquet J, Van Cauwenberge P, Khaltaev N, et al. Allergic rhinitis and its impact on asthma. J Allergy Clin Immunol 2001;108(5 Suppl):S147-334

19.

Bentley AM, Jacobson MR, Cumberworth V, et al. Immunohistology of the nasal mucosa in seasonal allergic rhinitis: increases in activated eosinophils and epithelial mast cells. J Allergy Clin Immunol 1992;89(4):877-83

20.

Pauwels R. Influence of treatment on the nose and/or the lungs. Clin Exp Allergy 1998;28(Suppl 2):37-40

21.

Adams RJ, Fuhlbrigge AL, Finkelstein JA, Weiss ST. Intranasal steroids and the risk of emergency department visits for asthma. J Allergy Clin Immunol 2002;109(4):636-42

22.

Wilson AM, O’Byrne PM, Parameswaran K. Leukotriene receptor antagonists for allergic rhinitis: a systematic review and meta-analysis. Am J Med 2004;116(5):338-44

23.

Abramson MJ, Puy RM, Weiner JM. Allergen immunotherapy for asthma. Cochrane Database Syst Rev 2003(4):CD001186

24.

Vignola AM, Humbert M, Bousquet J, et al. Efficacy and tolerability of antiimmunoglobulin E therapy with omalizumab in patients with concomitant allergic asthma and persistent allergic rhinitis: SOLAR. Allergy 2004;59(7):709-17

25.

Kopp MV, Brauburger J, Riedinger F, et al. The effect of anti-IgE treatment on in vitro leukotriene release in children with seasonal allergic rhinitis. J Allergy Clin Immunol 2002;110(5):728-35

16.

Grossman J. One airway, one disease. Chest 1997;111(2 Suppl):11S-6S

17.

American Thoracic Society Workshop. Immunobiology of asthma and rhinitis. Pathogenic factors and therapeutic options. Am J Respir Crit Care Med 1999;160(5 Pt 1):1778-87

26.

Philip G, Nayak AS, Berger WE, et al. The effect of montelukast on rhinitis symptoms in patients with asthma and seasonal allergic rhinitis. Curr Med Res Opin 2004;20(10):1549-58

18.

Simons FE. Allergic rhinobronchitis: the asthma-allergic rhinitis link. J Allergy Clin Immunol 1999;104(3 Pt 1):534-40

27.

Taramarcaz P, Gibson PG. Intranasal corticosteroids for asthma control in people with coexisting asthma and

Expert Opin. Drug Discov. (2014) 9(6)

603

L. Calzetta et al.

28.

Dahl R, Nielsen LP, Kips J, et al. Intranasal and inhaled fluticasone propionate for pollen-induced rhinitis and asthma. Allergy 2005;60(7):875-81

29.

Dykewicz MS, Fineman S. Executive Summary of Joint Task Force Practice Parameters on Diagnosis and Management of Rhinitis. Ann Allergy Asthma Immunol 1998;81(5 Pt 2):463-8

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

30.

31.

32.

33.

34.

35.

.

36.

.

604

Passalacqua G, Durham SR; Global Allergy and Asthma European Network. Allergic rhinitis and its impact on asthma update: allergen immunotherapy. J Allergy Clin Immunol 2007;119(4):881-91 Till SJ, Francis JN, Nouri-Aria K, Durham SR. Mechanisms of immunotherapy. J Allergy Clin Immunol 2004;113(6):1025-34; quiz 35 Walker SM, Pajno GB, Lima MT, et al. Grass pollen immunotherapy for seasonal rhinitis and asthma: a randomized, controlled trial. J Allergy Clin Immunol 2001;107(1):87-93 Van Cauwenberge P. ARIA: impact of compliance. Clin Exp Allergy Rev 2005;5(1):1472-9733 Brozek JL, Bousquet J, Baena-Cagnani CE, et al. Allergic Rhinitis and its Impact on Asthma (ARIA) guidelines: 2010 revision. J Allergy Clin Immunol 2010;126(3):466-76 Braunstahl GJ, Overbeek SE, Kleinjan A, et al. Nasal allergen provocation induces adhesion molecule expression and tissue eosinophilia in upper and lower airways. J Allergy Clin Immunol 2001;107(3):469-76 A study that describes as nasal allergen provocation results in generalized airway inflammation through upregulation of adhesion molecules, confirming the inflammatory relationship between upper and lower airways. Serrano CD, Valero A, Bartra J, et al. Nasal and bronchial inflammation after nasal allergen challenge: assessment using noninvasive methods. J Investig Allergol Clin Immunol 2012;22(5):351-6 A study that proves that nasal allergen challenge induces clinical and inflammatory responses in both nose and bronchi that can be assessed using noninvasive methods, confirming the

J Allergy Clin Immunol 1993;92(6):878-90

inflammatory relationship between upper and lower airways.

rhinitis. Cochrane Database Syst Rev 2003(4):CD003570 37.

Braunstahl GJ, Kleinjan A, Overbeek SE, et al. Segmental bronchial provocation induces nasal inflammation in allergic rhinitis patients. Am J Respir Crit Care Med 2000;161(6):2051-7

38.

Varney VA, Jacobson MR, Sudderick RM, et al. Immunohistology of the nasal mucosa following allergeninduced rhinitis. Identification of activated T lymphocytes, eosinophils, and neutrophils. Am Rev Respir Dis 1992;146(1):170-6

39.

..

Howarth PH, Persson CG, Meltzer EO, et al. Objective monitoring of nasal airway inflammation in rhinitis. J Allergy Clin Immunol 2005;115(3 Suppl 1):S414-41 The different approaches to the monitoring of nasal airway inflammation and their practical applicability are assessed in this exaustive paper.

40.

Larche M. Immunoregulation by targeting T cells in the treatment of allergy and asthma. Curr Opin Immunol 2006;18(6):745-50

41.

Compalati E, Passalacqua G, Bonini M, Canonica GW. The efficacy of sublingual immunotherapy for house dust mites respiratory allergy: results of a GA2LEN meta-analysis. Allergy 2009;64(11):1570-9

42.

Hirsch T, Sahn M, Leupold W. Double-blind placebo-controlled study of sublingual immunotherapy with house dust mite extract (D.pt.) in children. Pediatr Allergy Immunol 1997;8(1):21-7

43.

Naclerio RM, Meier HL, Kagey-Sobotka A, et al. Mediator release after nasal airway challenge with allergen. Am Rev Respir Dis 1983;128(4):597-602

44.

Svensson C, Pipkorn U, Alkner U, et al. Topical vasoconstrictor (oxymetazoline) does not affect histamine-induced mucosal exudation of plasma in human nasal airways. Clin Exp Allergy 1992;22(3):411-16

45.

46.

Wilson SJ, Lau L, Howarth PH. Inflammatory mediators in naturally occurring rhinitis. Clin Exp Allergy 1998;28(2):220-7 Gosset P, Malaquin F, Delneste Y, et al. Interleukin-6 and interleukin-1 alpha production is associated with antigen-induced late nasal response.

Expert Opin. Drug Discov. (2014) 9(6)

47.

Howarth PH, Wilson S, Lau L, Rajakulasingam K. The nasal mast cell and rhinitis. Clin Exp Allergy 1991;21(Suppl 2):3-8

48.

Naclerio RM, Proud D, Togias AG, et al. Inflammatory mediators in late antigen-induced rhinitis. N Engl J Med 1985;313(2):65-70

49.

Knani J, Campbell A, Enander I, et al. Indirect evidence of nasal inflammation assessed by titration of inflammatory mediators and enumeration of cells in nasal secretions of patients with chronic rhinitis. J Allergy Clin Immunol 1992;90(6 Pt 1):880-9

50.

Linden M, Svensson C, Andersson M, et al. Circulating eosinophil/basophil progenitors and nasal mucosal cytokines in seasonal allergic rhinitis. Allergy 1999;54(3):212-19

51.

Benson M, Strannegard IL, Wennergren G, Strannegard O. Low levels of interferon-gamma in nasal fluid accompany raised levels of T-helper 2 cytokines in children with ongoing allergic rhinitis. Pediatr Allergy Immunol 2000;11(1):20-8

52.

Weido AJ, Reece LM, Alam R, et al. Intranasal fluticasone propionate inhibits recovery of chemokines and other cytokines in nasal secretions in allergen-induced rhinitis. Ann Allergy Asthma Immunol 1996;77(5):407-15

53.

Kuna P, Lazarovich M, Kaplan AP. Chemokines in seasonal allergic rhinitis. J Allergy Clin Immunol 1996;97(1 Pt 1):104-12

54.

Salib RJ, Lau LC, Howarth PH. Nasal lavage fluid concentrations of eotaxin-1 (CCL11) in naturally occurring allergic rhinitis: relationship to disease activity, nasal luminal eosinophil influx, and plasma protein exudation. Clin Exp Allergy 2005;35(8):995-1002

55.

Pipkorn U, Proud D, Lichtenstein LM, et al. Inhibition of mediator release in allergic rhinitis by pretreatment with topical glucocorticosteroids. N Engl J Med 1987;316(24):1506-10

56.

Alam R, Sim TC, Hilsmeier K, Grant JA. Development of a new technique for recovery of cytokines from inflammatory sites in situ. J Immunol Methods 1992;155(1):25-9

Advances in asthma drug discovery

57.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

58.

59.

.

Sim TC, Grant JA, Hilsmeier KA, et al. Proinflammatory cytokines in nasal secretions of allergic subjects after antigen challenge. Am J Respir Crit Care Med 1994;149(2 Pt 1):339-44 Linden M, Svensson C, Andersson E, et al. Immediate effect of topical budesonide on allergen challenge-induced nasal mucosal fluid levels of granulocytemacrophage colony-stimulating factor and interleukin-5. Am J Respir Crit Care Med 2000;162(5):1705-8 Erin EM, Neighbour H, Tan AJ, et al. Nasal testing for novel anti-inflammatory agents. Clin Exp Allergy 2005;35(8):981-5 A concise editorial that focus on the novel anti-inflammatory drug discovery by using nasal cells and related procedures.

..

research: application to studies of pathogenesis and the effects of treatment. Am J Respir Crit Care Med 2003;168(6 Pt 2):S1-17 An extensive papers that investigates the proper methods for the assessment of airway biopsies in clinical research: from tissue fixation to variability and quantification analysis.

67.

Cohen GA, MacPherson GA, Golembesky HE, et al. Normal nasal cytology in infancy. Ann Allergy 1985;54(2):112-14

68.

Ventura MT, Murgia N, Montuschi P, et al. Exhaled breath condensate, nasal eosinophil cationic protein level and nasal cytology during immunotherapy for cypress allergy. J Biol Regul Homeost Agents 2013;27(4):1083-9

69.

Bascom R, Wachs M, Naclerio RM, et al. Basophil influx occurs after nasal antigen challenge: effects of topical corticosteroid pretreatment. J Allergy Clin Immunol 1988;81(3):580-9

60.

Pipkorn U, Karlsson G. Methods for obtaining specimens from the nasal mucosa for morphological and biochemical analysis. Eur Respir J 1988;1(9):856-62

61.

Pipkorn U, Karlsson G, Enerback L. A brush method to harvest cells from the nasal mucosa for microscopic and biochemical analysis. J Immunol Methods 1988;112(1):37-42

71.

Jacobson MR, Juliusson S, Lowhagen O, et al. Effect of topical corticosteroids on seasonal increases in epithelial eosinophils and mast cells in allergic rhinitis: a comparison of nasal brush and biopsy methods. Clin Exp Allergy 1999;29(10):1347-55

72.

Meltzer EO, Orgel HA, Rogenes PR, Field EA. Nasal cytology in patients with allergic rhinitis: effects of intranasal fluticasone propionate. J Allergy Clin Immunol 1994;94(4):708-15

73.

Furin MJ, Norman PS, Creticos PS, et al. Immunotherapy decreases antigeninduced eosinophil cell migration into the nasal cavity. J Allergy Clin Immunol 1991;88(1):27-32

62.

63.

64.

van Benten IJ, van Drunen CM, Koevoet JL, et al. Reduced nasal IL-10 and enhanced TNFalpha responses during rhinovirus and RSV-induced upper respiratory tract infection in atopic and non-atopic infants. J Med Virol 2005;75(2):348-57 Stoop AE, Hameleers DM, v Run PE, et al. Lymphocytes and nonlymphoid cells in the nasal mucosa of patients with nasal polyps and of healthy subjects. J Allergy Clin Immunol 1989;84(5 Pt 1):734-41

65.

Fokkens WJ, Vroom TM, Gerritsma V, Rijntjes E. A biopsy method to obtain high quality specimens of nasal mucosa. Rhinology 1988;26(4):293-5

66.

Jeffery P, Holgate S, Wenzel S; Endobronchial Biopsy Workshop. Methods for the assessment of endobronchial biopsies in clinical

70.

Connell JT. Nasal disease: mechanisms and classification. Ann Allergy 1983;50(4):227-35 Meltzer EO. Clinical and antiinflammatory effects of intranasal budesonide aqueous pump spray in the treatment of perennial allergic rhinitis. Ann Allergy Asthma Immunol 1998;81(2):128-34

74.

Cengizlier R, Saraclar Y, Adalioglu C, Tuncer A. Changes in nasal metachromatic cells during allergen immunotherapy in children. Allergol Immunopathol (Madr) 1995;23(3):111-16

75.

Otsuka H, Mezawa A, Ohnishi M, et al. Changes in nasal metachromatic cells during allergen immunotherapy. Clin Exp Allergy 1991;21(1):115-19

76.

Francis JN, Durham SR. Adjuvants for allergen immunotherapy: experimental results and clinical perspectives.

Expert Opin. Drug Discov. (2014) 9(6)

Curr Opin Allergy Clin Immunol 2004;4(6):543-8 77.

Javanmard SH, Moeiny A. Quantitative immunohistochemistry by measuring chromogen signal strength using a C# written program. J Res Med Sci 2009;14(3):201-3

78.

Matkowskyj KA, Schonfeld D, Benya RV. Quantitative immunohistochemistry by measuring cumulative signal strength using commercially available software photoshop and matlab. J Histochem Cytochem 2000;48(2):303-12

79.

Kamentsky L, Jones TR, Fraser A, et al. Improved structure, function and compatibility for CellProfiler: modular high-throughput image analysis software. Bioinformatics 2011;27(8):1179-80

80.

Carpenter AE, Jones TR, Lamprecht MR, et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol 2006;7(10):R100

81.

Langdale JA. In situ hybridization. In: The maize handbook. Springer, New York; 1994. p. 165-80

82.

Martineau B, Taylor WC. Cell-specific photosynthetic gene expression in maize determined using cell separation techniques and hybridization in situ. Plant Physiol 1986;82(2):613-18

83.

Durham SR, Ying S, Varney VA, et al. Cytokine messenger RNA expression for IL-3, IL-4, IL-5, and granulocyte/ macrophage-colony-stimulating factor in the nasal mucosa after local allergen provocation: relationship to tissue eosinophilia. J Immunol 1992;148(8):2390-4

84.

Ghaffar O, Laberge S, Jacobson MR, et al. IL-13 mRNA and immunoreactivity in allergen-induced rhinitis: comparison with IL-4 expression and modulation by topical glucocorticoid therapy. Am J Respir Cell Mol Biol 1997;17(1):17-24

85.

Divjak M, Glare EM, Walters EH. Improvement of non-radioactive in situ hybridization in human airway tissues: use of PCR-generated templates for synthesis of probes and an antibody sandwich technique for detection of hybridization. J Histochem Cytochem 2002;50(4):541-8

605

L. Calzetta et al.

86.

87.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

88.

89.

90.

Chanez P, Vignola AM, Vic P, et al. Comparison between nasal and bronchial inflammation in asthmatic and control subjects. Am J Respir Crit Care Med 1999;159(2):588-95

97.

Wise SK, Laury AM, Katz EH, et al. Interleukin-4 and interleukin-13 compromise the sinonasal epithelial barrier and perturb intercellular junction protein expression. Int Allergy Rhinol 2014. [Epub ahead of print]

Gelfand EW. Inflammatory mediators in allergic rhinitis. J Allergy Clin Immunol 2004;114(5 Suppl):S135-8

98.

Pierrou S, Broberg P, O’Donnell RA, et al. Expression of genes involved in oxidative stress responses in airway epithelial cells of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2007;175(6):577-86

Yeh TH, Lee SY, Hsu WC. Expression of SPLUNC1 protein in nasal polyp epithelial cells in air-liquid interface culture treated with IL-13. Am J Rhinol Allergy 2010;24(1):17-20

99.

Choe MM, Sporn PH, Swartz MA. Extracellular matrix remodeling by dynamic strain in a three-dimensional tissue-engineered human airway wall model. Am J Respir Cell Mol Biol 2006;35(3):306-13

Gray TE, Guzman K, Davis CW, et al. Mucociliary differentiation of serially passaged normal human tracheobronchial epithelial cells. Am J Respir Cell Mol Biol 1996;14(1):104-12 Jain R, Pan J, Driscoll JA, et al. Temporal relationship between primary and motile ciliogenesis in airway epithelial cells. Am J Respir Cell Mol Biol 2010;43(6):731-9

100. Zhang S, Smartt H, Holgate ST, Roche WR. Growth factors secreted by bronchial epithelial cells control myofibroblast proliferation: an in vitro co-culture model of airway remodeling in asthma. Lab Invest 1999;79(4):395-405

91.

Choe MM, Tomei AA, Swartz MA. Physiological 3D tissue model of the airway wall and mucosa. Nat Protoc 2006;1(1):357-62

101. Jaffar Z, Roberts K, Pandit A, et al. 7 costimulation is required for IL-5 and IL-13 secretion by bronchial biopsy tissue of atopic asthmatic subjects in response to allergen stimulation. Am J Respir Cell Mol Biol 1999;20(1):153-62

92.

Wu X, Peters-Hall JR, Bose S, et al. Human bronchial epithelial cells differentiate to 3D glandular acini on basement membrane matrix. Am J Respir Cell Mol Biol 2011;44(6):914-21

102. Hidi R, Riches V, Al-Ali M, et al. Role of B7-CD28/CTLA-4 costimulation and NF-kappa B in allergen-induced T cell chemotaxis by IL-16 and RANTES. J Immunol 2000;164(1):412-18

93.

Klein SG, Serchi T, Hoffmann L, et al. An improved 3D tetraculture system mimicking the cellular organisation at the alveolar barrier to study the potential toxic effects of particles on the lung. Part Fibre Toxicol 2013;10(1):31

103. Dent G, Hosking LA, Lordan JL, et al. Differential roles of IL-16 and CD28/ B7 costimulation in the generation of T-lymphocyte chemotactic activity in the bronchial mucosa of mild and moderate asthmatic individuals. J Allergy Clin Immunol 2002;110(6):906-14

94.

95.

96.

606

Drumm K, Messner C, Kienast K. Reactive oxygen intermediate-release of fibre-exposed monocytes increases inflammatory cytokine-mRNA level, protein tyrosine kinase and NF-kappaB activity in co-cultured bronchial epithelial cells (BEAS-2B). Eur J Med Res 1999;4(7):257-63 Mio T, Liu XD, Adachi Y, et al. Human bronchial epithelial cells modulate collagen gel contraction by fibroblasts. Am J Physiol 1998;274(1 Pt 1):L119-26 Jeanson L, Kelly M, Coste A, et al. Oxidative stress induces unfolding protein response and inflammation in nasal polyposis. Allergy 2012;67(3):403-12

104. Vijayanand P, Durkin K, Hartmann G, et al. Chemokine receptor 4 plays a key role in T cell recruitment into the airways of asthmatic patients. J Immunol 2010;184(8):4568-74 105. Lu X, Zhang XH, Wang H, et al. Expression of osteopontin in chronic rhinosinusitis with and without nasal polyps. Allergy 2009;64(1):104-11 106. Patou J, Gevaert P, Van Zele T, et al. Staphylococcus aureus enterotoxin B, protein A, and lipoteichoic acid stimulations in nasal polyps. J Allergy Clin Immunol 2008;121(1):110-15 107. Chen B, Antunes MB, Claire SE, et al. Reversal of chronic rhinosinusitis-

Expert Opin. Drug Discov. (2014) 9(6)

associated sinonasal ciliary dysfunction. Am J Rhinol 2007;21(3):346-53 108. Chen B, Shaari J, Claire SE, et al. Altered sinonasal ciliary dynamics in chronic rhinosinusitis. Am J Rhinol 2006;20(3):325-9 109. Huh D, Hamilton GA, Ingber DE. From 3D cell culture to organs-on-chips. Trends Cell Biol 2011;21(12):745-54 .. A very interesting review that focuses on the most recent techniques for approaching three-dimensional micro-engineering lung-on-achip devices. 110. Huh D, Matthews BD, Mammoto A, et al. Reconstituting organ-level lung functions on a chip. Science 2010;328(5986):1662-8 111. Huh D, Fujioka H, Tung YC, et al. Acoustically detectable cellular-level lung injury induced by fluid mechanical stresses in microfluidic airway systems. Proc Natl Acad Sci USA 2007;104(48):18886-91 112. Nalayanda DD, Puleo C, Fulton WB, et al. An open-access microfluidic model for lung-specific functional studies at an air-liquid interface. Biomed Microdevices 2009;11(5):1081-9 113. Trist DG. Scientific process, pharmacology and drug discovery. Curr Opin Pharmacol 2011;11(5):528-33 114. Orloff J, Douglas F, Pinheiro J, et al. The future of drug development: advancing clinical trial design. Nat Rev Drug Discov 2009;8(12):949-57 115. Diamant Z, Boot D, Kamerling I, Bjermer L. Methods used in clinical development of novel anti-asthma therapies. Respir Med 2008;102(3):332-8 116. Lodrup Carlsen KC, Hedlin G, Bush A, et al. Assessment of problematic severe asthma in children. Eur Respir J 2011;37(2):432-40 117. Holgate ST. Stratified approaches to the treatment of asthma. Br J Clin Pharmacol 2013;76(2):277-91 118. Healey GD, Evans N, Hopkin JM, et al. Evaluation of nasal epithelium sampling as a tool in the preclinical development of siRNA-based therapeutics for asthma. J Cell Mol Med 2013;17(3):356-64 119. Comer DM, Elborn JS, Ennis M. Comparison of nasal and bronchial epithelial cells obtained from patients

Advances in asthma drug discovery

with COPD. PLoS One 2012;7(3):e32924

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Connecticut on 04/13/15 For personal use only.

120.

Mauad T, Dolhnikoff M. Pathologic similarities and differences between asthma and chronic obstructive pulmonary disease. Curr Opin Pulm Med 2008;14(1):31-8

124.

Calzetta L, Page CP, Spina D, et al. Effect of the mixed phosphodiesterase 3/4 inhibitor RPL554 on human isolated bronchial smooth muscle tone. J Pharmacol Exp Ther 2013;346(3):414-23

125.

Matera MG, Calzetta L, Passeri D, et al. Epithelial-smooth muscle cooperation is needed for brain natriuretic peptidedependent bronchorelaxant activity. Pulm Pharmacol Ther 2013;26(1):156-7 Calzetta L, Spina D, Cazzola M, et al. Pharmacological characterization of adenosine receptors on isolated human bronchi. Am J Respir Cell Mol Biol 2011;45(6):1222-31

121.

Carroll N, Elliot J, Morton A, James A. The structure of large and small airways in nonfatal and fatal asthma. Am Rev Respir Dis 1993;147(2):405-10

122.

Chung KF. Airway smooth muscle cells: contributing to and regulating airway mucosal inflammation? Eur Respir J 2000;15(5):961-8 An analysis of the contribution of airway smooth muscle (ASM) on the pathogenesis of asthma concerning the proliferation, the expression and secretion of pro-inflammatory cytokines and mediators.

126.

Calzetta L, Passeri D, Kanabar V, et al. Brain natriuretic peptide protects against hyperreactivity of human asthmatic airway smooth muscle via an epithelial cell dependent mechanism. Am J Respir Cell Mol Biol 2014;50(3):493-501 A study that proves the relevance of models based on ASM cells and passively sensitized airway epithelial cells for investigating the bronchial hyperresponsiveness in asthma and the cross-talk between epithelium and ASM.

128.

Matera MG, Calzetta L, Parascandolo V, et al. Relaxant effect of brain natriuretic peptide in nonsensitized and passively sensitized isolated human bronchi. Pulm Pharmacol Ther 2009;22(6):478-82

129.

Matera MG, Calzetta L, Peli A, et al. Immune sensitization of equine bronchus: glutathione, IL-1beta expression and tissue responsiveness. Respir Res 2005;6:104

130.

van Lunteren E, Moyer M. Auxotonic contractile responses of rat tracheal and

.

123.

.

127.

Matera MG, Calzetta L, Passeri D, et al. Epithelium integrity is crucial for the relaxant activity of brain natriuretic peptide in human isolated bronchi. Br J Pharmacol 2011;163(8):1740-54

Expert Opin. Drug Discov. (2014) 9(6)

..

bronchial airway smooth muscle. Pulm Pharmacol Ther 2001;14(6):443-53 A study that describes an alternative approach, compared with the classic precision-cut lung slices method, for studing small airways that is based on a videomicrometry technique that prevents the complications related to the use of confounding agarose gel to inflate the lung or complex parenchymal sections that have numerous contracting elements.

131. Wohlsen A, Uhlig S, Martin C. Immediate allergic response in small airways. Am J Respir Crit Care Med 2001;163(6):1462-9

Affiliation Luigino Calzetta1 PhD, Paola Rogliani2 MD, Mario Cazzola†2 MD, Maria Gabriella Matera3 MD † Author for correspondence 1 IRCCS, San Raffaele Pisana Hospital, Department of Pulmonary Rehabilitation, Rome, Italy 2 University of Rome Tor Vergata, Department of System Medicine, Via Montpellier 1, 00133 Rome, Italy Tel: +0039 06 2090 0631/0633; E-mail: [email protected] 3 Second University of Naples, Department of Experimental Medicine, Naples, Italy

607

Advances in asthma drug discovery: evaluating the potential of nasal cell sampling and beyond.

Inhaled corticosteroid anti-inflammatory therapy is effective at controlling disease symptoms of asthma, but a subset of patients remains symptomatic ...
400KB Sizes 0 Downloads 0 Views