Kinase inhibitors in the treatment of obstructive pulmonary diseases

https://doi.org/10.1016/j.coph.2020.03.005Get rights and content

Highlights

Chronic pulmonary diseases, including chronic obstructive pulmonary disease (COPD) and asthma, are major causes of death and reduced quality of life. Characteristic of chronic pulmonary disease is excessive lung inflammation that occurs in response to exposure to inhaled irritants, chemicals, and allergens. Chronic inflammation leads to remodeling of the airways that includes excess mucus secretion, proliferation of smooth muscle cells, increased deposition of extracellular matrix proteins and fibrosis. Protein kinases have been implicated in mediating inflammatory signals and airway remodeling associated with reduced lung function in chronic pulmonary disease. This review will highlight the role of protein kinases in the lung during chronic inflammation and examine opportunities to use protein kinase inhibitors for the treatment of chronic pulmonary diseases.

Introduction

Asthma and chronic obstructive pulmonary disease (COPD) are the two most common chronic pulmonary diseases, affecting approximately 300 million and 250 million individuals worldwide, respectively, and causing 250 000 and 3.1 million deaths annually, respectively [1]. Asthma and COPD share symptoms of cough and dyspnea, the presence of chronic airway inflammation driven by exposure to inhaled immune stimuli, airway wall thickening, and the contribution of cellular senescence of airway epithelium. But the two diseases differ in the segment of the bronchial tree affected, the nature of the inflammation, the inciting immune stimulants, and their long-term course.

In asthma, the entire airway is involved while in COPD, pathologic changes are most pronounced in the small airways. Inflammation in asthma is typically allergen-driven and IgE-driven, and involves TH2 lymphocytes, mast cells and eosinophils [2]. In COPD, inflammation is driven by inhaled toxins and irritants, including those in tobacco smoke, non-tobacco biomass smoke, air pollution, and inhaled endotoxin, and is characterized by TH1 and TH17 lymphocytes, CD8+ lymphocyte predominance, neutrophil infiltration, and macrophage activation [3••]. Inhaled therapy with short-acting and long-acting beta agonists and corticosteroids are effective in both diseases. However, airway obstruction in asthma is usually more reversible with beta agonist therapy and more responsive to corticosteroid therapy than in COPD [4]. Corticosteroids have been shown to reduce the rate of exacerbations, airway inflammation, and the rate of quality of life deterioration in COPD patients; however, an increased risk of pneumonia along with local and systemic adverse effects has also been observed [5]. Inhaled long-acting muscarinic agonists are more effective in COPD than asthma [6]. More recently a growing number of injectable biologics targeting specific inflammatory pathways have become available for treating subclasses of asthma, but do not appear to be effective in most patients with COPD. It should be noted that asthma and COPD are both heterogenous diseases with a substantial overlap between the two, including a defined asthma-COPD overlap syndrome [7].

The chronic release of inflammatory cytokines and growth factors from epithelial and immune cells enhances protein kinase signalling pathways and underlies the pathology of asthma and COPD. Several cytokines and growth factors are implicated in chronic pulmonary disease and include the interleukins (IL) IL-1β IL-4, IL-5, IL-6, IL-13, IL-17A, IL-27, IL-33, interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), epidermal growth factor (EGF), and platelet-derived growth factor (PDGF) [8,9, 10, 11, 12, 13]. Many protein kinases have been implicated in the signal transduction pathways linking inflammatory mediators and the hyper-contractility of airways, mucus hypersecretion, immune cell infiltration, and airway remodelling that leads to debilitating lung function observed in chronic pulmonary disease [14,15]. Figure 1 summarizes protein kinases that are involved in the pathology of asthma and COPD.

Inflammatory signals mediate the pathology of obstructive lung disease through the activation of protein kinase activity [16]. Cytokine receptors (CR) and receptor tyrosine kinases (RTK) at the plasma membrane communicate cytokine and growth factor signals through several mitogen-activated protein (MAP) kinases, phosphatidylinositide 3-kinases (PI3K), and Janus kinases (JAK). Inflammation causes increased adenosine generation, which via activation of G-protein coupled adenosine receptors (A2BAR) and downstream kinase signalling cascades, contributes to the pathologic changes in asthma and COPD [17]. Gene expression associated with inflammation and tissue remodelling is regulated predominantly by a subset of transcription factors that include nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), activator protein-1 (AP-1), cAMP response element-binding protein (CREB), and signal transducer and activator of transcription (STAT) proteins [18].

MAP kinases that respond to cellular stress, such as the c-Jun N-terminal kinases (JNK) and p38 MAP kinases, have long been implicated in mediating inflammatory signals relevant to lung disease [16,19]. However, to date, none of the JNK or p38 MAP kinase inhibitors have shown efficacy in clinical trials and are dose limiting due to unwanted toxicity and side effects. Targeted inhibition of pro-inflammatory p38 MAP kinase substrates such as mitogen-activated protein kinase activated protein kinase-2 (MAPKAPK-2 or MK2) may overcome toxicity issues associated with p38 inhibitors and treating chronic pulmonary disease [20]. MK2 is a mediator of inflammatory signals such as TNF-α, IL-6, and IL-1β so MK2 mediators might further reduce the inflammatory response [21]. Eynott et al. provided evidence that targeted inhibition of JNK could mitigate allergen-induced inflammation and proliferation of airway epithelial and smooth muscle cells [22]. While one phase II clinical trial evaluating the JNK inhibitor Tanzisertib for treating idiopathic pulmonary fibrosis ended due to lack of efficacy, no JNK inhibitors are currently being tested for asthma or COPD.

Mitogen activation of the extracellular signal-regulated kinases (ERK) MAP kinases in airway smooth muscle (ASM) cells stimulates cell proliferation and subsequent airway remodelling associated with asthma [23]. This was supported by studies that showed that serum from atopic asthma patients enhanced the expression of cyclin D1, which increases proliferation of human ASM cells [24]. Activated ERK signalling enhances cell proliferation by increasing the expression of cyclin D1.

There is evidence that protein kinases associated with deregulated angiogenesis contribute to the progression of chronic lung disease including COPD [25]. Angiogenic factors such as cytokines and growth factors that are released as a result of obstructive pulmonary disease progression leads to the stimulation of direct and indirect proangiogenic markers that generate and stabilize new blood vessels [26]. This microvascular dysfunction leads to re-modulation and inflammation of the bronchi. Recent studies suggest that targeted inhibition of inflammation-mediated activation of the receptor tyrosine kinases vascular endothelial growth factor receptor (VEGFR) and fibroblast growth factor receptor-2 (FGFR-2) reduces angiogenesis associated with lung remodelling in COPD [26]. Similarly, a small clinical study suggested that inhibition of PDGF and the related c-Kit receptor tyrosine kinase with Masitinib is effective in severe corticosteroid-dependent asthma [27].

In COPD patients, protein kinases play an important role in the loss of tissue architecture by enhancing degradation of the extracellular matrix by proteinases such as matrix metalloproteinases-9 (MMP-9) [28]. Proinflammatory triggers including allergens, cigarette smoke, bacterial lipopolysaccharides (LPS), interleukins (IL-17 and IL-1β), and other inflammatory signals that induce an oxidative stress response, activate MAP kinases and PI3K signalling, which enhances AP-1-mediated expression of MMP-9 [22,29,30,31]. Another key regulator of inflammatory signals is the NF-κB transcription factor. Targeted inhibition of the inhibitor of κB kinase (IKK) is a potential approach to prevent NF-κB translocation to the nucleus and inflammatory gene expression. The status of past and current clinical trials evaluating protein kinase inhibitors for treatment of asthma or COPD is listed in Table 1.

Section snippets

Epithelial cells

Inflammation-inducing chemicals and irritants disrupt airway epithelium and stimulate aberrant kinase signaling both within epithelial cells. Increased expression of the receptor tyrosine kinase EGFR, and its ligands have been reported in the epithelium of asthmatic airways [39,40]. Activation of epithelial EGFR can lead to corticosteroid-insensitivity and plays a key role in airway remodeling, mucus secretion, and inflammation. The EGFR-tyrosine kinase inhibitors, Erlotinib and Osimertinib,

Conclusions and future directions

Currently, obstructive lung disease treatments focus on counteracting episodes of bronchospasm and reducing allergic inflammation through corticosteroids, which, although such strategies have shown success, they neither cure nor prevent disease progression [59,60,61]. Kinase inhibitors could provide a more targeted therapeutic option to meet this unmet need.

The effectiveness of protein kinases inhibitors will likely depend on delivery methods that limit off-target effects. The ubiquitous

Credit author statement

Amy E. Defnet, Jeffery D. Hasday, and Paul Shapiro all contributed to the writing, review, and editing.

Conflicts of interest statement

Nothing declared.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgement

This work was supported by the National Institutes of Health [grant numbers AI126492 and T32GM066706].

References (67)

  • A.B. Pernis et al.

    Jak-stat signaling in asthma

    J Clin Invest

    (2002)
  • E.J. Hanan et al.

    Discovery of potent and selective pyrazolopyrimidine janus kinase 2 inhibitors

    J Med Chem

    (2012)
  • G. Hodge et al.

    Therapeutic targeting steroid resistant pro-inflammatory nk and nkt-like cells in chronic inflammatory lung disease

    Int J Mol Sci

    (2019)
  • F. Chen et al.

    Retinoic acid signaling is essential for airway smooth muscle homeostasis

    JCI Insight

    (2018)
  • Collaborators GBDCRD

    Global, regional, and national deaths, prevalence, disability-adjusted life years, and years lived with disability for chronic obstructive pulmonary disease and asthma, 1990-2015: a systematic analysis for the global burden of disease study 2015

    Lancet Respir Med

    (2017)
  • A. KleinJan

    Airway inflammation in asthma: key players beyond the th2 pathway

    Curr Opin Pulmonary Med

    (2016)
  • P.J. Barnes

    Targeting cytokines to treat asthma and chronic obstructive pulmonary disease

    Nat Rev Immunol

    (2018)
  • J.L. McCracken et al.

    Diagnosis and management of asthma in adults: a review

    JAMA

    (2017)
  • I.A. Yang et al.

    Use of inhaled corticosteroids in copd: improving efficacy

    Expert Rev Respir Med

    (2016)
  • C.F. Vogelmeier et al.

    Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 report. Gold executive summary

    Am J Respir Crit Care Med

    (2017)
  • A. Alshabanat et al.

    Asthma and copd overlap syndrome (acos): a systematic review and meta analysis

    PLoS One

    (2015)
  • B.N. Lambrecht et al.

    The cytokines of asthma

    Immunity

    (2019)
  • L. Cohn et al.

    T helper 1 cells and interferon gamma regulate allergic airway inflammation and mucus production

    J Exp Med

    (1999)
  • A. Bellini et al.

    Interleukin (il)-4, il-13, and il-17a differentially affect the profibrotic and proinflammatory functions of fibrocytes from asthmatic patients

    Mucosal Immunol

    (2012)
  • M. Yang et al.

    Emerging roles of pulmonary macrophages in driving the development of severe asthma

    J Leukocyte Biol

    (2012)
  • K.F. Chung

    Cytokines in chronic obstructive pulmonary disease

    Eur Respir J Suppl

    (2001)
  • K. Blease et al.

    Emerging treatments for asthma

    Expert Opin Emerg Drugs

    (2003)
  • M. Caruso et al.

    Adenosine and adenosine receptors: their contribution to airway inflammation and therapeutic potential in asthma

    Curr Med Chem

    (2009)
  • P.J. Barnes

    Transcription factors in airway diseases

    Lab Invest

    (2006)
  • S. Kumar et al.

    P38 map kinases: key signalling molecules as therapeutic targets for inflammatory diseases

    Nat Rev Drug Discov

    (2003)
  • R.K. Singh et al.

    Novel therapeutic potential of mitogen-activated protein kinase activated protein kinase 2 (mk2) in chronic airway inflammatory disorders

    Curr Drug Targets

    (2019)
  • R.K. Singh et al.

    Biological functions and role of mitogen-activated protein kinase activated protein kinase 2 (mk2) in inflammatory diseases

    Pharmacol Rep

    (2017)
  • P.R. Eynott et al.

    Allergen-induced inflammation and airway epithelial and smooth muscle cell proliferation: Role of jun n-terminal kinase

    Brit J Pharmacol

    (2003)
  • Cited by (16)

    • Constitutive immune activity promotes JNK- and FoxO-dependent remodeling of Drosophila airways

      2021, Cell Reports
      Citation Excerpt :

      A certain level of JNK/FoxO activation/deregulation caused by chronic input can remain inconspicuous over a long time, but an additional stressor, such as an infection, might cause activation above the threshold, leading to remodeling of the airways (Broide, 2008; Cheng et al., 2007; Hart et al., 1998; Holgate, 2007b). Taking into account this highly important role of the JNK/FoxO axis for disease-associated remodeling processes, utilizing these signaling systems as intervention targets in chronic lung diseases is an emerging but highly promising field (Al-Tamari et al., 2018; Defnet et al., 2020; Eurlings et al., 2017; van der Velden et al., 2016). Activation of epithelial Imd signaling in the fly induced substantial structural changes comprising epithelial hyper- and metaplasia that resemble those observed in human inflammatory lung diseases (Ganesan and Sajjan, 2013; Grainge and Davies, 2013).

    View all citing articles on Scopus
    View full text