1,020
Views
1
CrossRef citations to date
0
Altmetric
REVIEW

Novel Anti-Inflammatory Approaches to COPD

ORCID Icon, , &
Pages 1333-1352 | Received 27 Apr 2023, Accepted 20 Jun 2023, Published online: 29 Jun 2023

Abstract

Airway inflammation, driven by different types of inflammatory cells and mediators, plays a fundamental role in COPD and its progression. Neutrophils, eosinophils, macrophages, and CD4+ and CD8+ T lymphocytes are key players in this process, although the extent of their participation varies according to the patient’s endotype. Anti-inflammatory medications may modify the natural history and progression of COPD. However, since airway inflammation in COPD is relatively resistant to corticosteroid therapy, innovative pharmacological anti-inflammatory approaches are required. The heterogeneity of inflammatory cells and mediators in annethe different COPD endo-phenotypes requires the development of specific pharmacologic agents. Indeed, over the past two decades, several mechanisms that influence the influx and/or activity of inflammatory cells in the airways and lung parenchyma have been identified. Several of these molecules have been tested in vitro models and in vivo in laboratory animals, but only a few have been studied in humans. Although early studies have not been encouraging, useful information emerged suggesting that some of these agents may need to be further tested in specific subgroups of patients, hopefully leading to a more personalized approach to treating COPD.

The Unmet Need for New Anti-Inflammatory Therapies for COPD

An aberrant inflammatory response that involves both innate and adaptive immunity characterizes COPD.Citation1 However, determining the precise role of specific cell types in the evolution of COPD is challenging.Citation2 While chronic inflammation is basically characterised by neutrophils and macrophages,Citation3 20–40% of patients exhibit increased eosinophil numbers in blood and lung tissue.Citation4 Furthermore, neutrophilic inflammation is variably associated with concomitant eosinophilic inflammation in many patients.Citation5 T cells, B cells, dendritic cells (DCs) and epithelial cells are also involved in COPD.Citation5

Corticosteroids (CSs) are the most often utilized anti-inflammatory medications in the treatment of COPD.Citation6 ICSs are indicated in COPD patients when there is an overlap with asthma or frequent exacerbations associated with high blood eosinophil counts (BEC).Citation7 Furthermore, CSs may play a cardioprotective role, reduce the risk of lung cancer, and possibly improve survival.Citation7 However, ICSs have little to no impact on the underlying inflammation in most COPD patients,Citation8 particularly in those who continue to smoke.Citation9

The transcription of some anti-inflammatory genes is controlled by histone deacetylase (HDAC)2, which has been found to be downregulated by oxidative stress generated by cigarette smoke and activated neutrophils.Citation8,Citation9 Another factor is the reduced glucocorticoid receptor (GR) expression in airway neutrophils, but not blood neutrophils, which has been found to be associated with the poor clinical response to ICS therapy in COPD patients.Citation10 Moreover, neutrophils from patients with COPD also showed an increase in GRβ levels,Citation11 which is thought to be a factor in the development of corticosteroid resistance because it is capable of inhibiting HDAC2 promoter activity.Citation12 Furthermore, several studies have revealed a connection between long-term ICS treatment and the incidence of pneumonia in COPD patients, mainly in those with lower BECs.Citation13

Since CSs are less effective in neutrophil-driven pulmonary inflammation, the demand for therapies that act on this type of inflammation is increasing.Citation6,Citation14

Over the past two decades, research has focused on discovering new targets capable of inhibiting the recruitment or activation of inflammatory cells involved in COPD and the development of drugs capable of blocking the inflammatory mediators released by these cells.Citation6 A complex network of inflammatory mediators, including chemokines, growth factors and lipid mediators, produced by the structural and inflammatory cells in the lung, are implicated in COPD and play a potential role in its pathogenesis.Citation15,Citation16 Therefore, their inhibition is an important strategy to tamper the ongoing inflammatory process. Unfortunately, the results have often fallen short of expectations.

This narrative review aims to examine ongoing research to identify and develop possible new anti-inflammatory therapies for use in patients with COPD.

Search Strategy

A review of the literature published up to May 2023 was performed through the PubMed and Scopus databases to identify studies related to our stated objective of reviewing novel drugs under development for treating COPD. Thereafter, evaluation of the references of the selected articles identified other relevant publications. All authors participated in evaluation of the literature.

Inhibition of Recruitment and Activation of the Inflammatory Cells

Several small molecule inhibitors and biologic therapies are able, at least in experimental models, to prevent the recruitment and activation of the cellular components of inflammation.Citation6 Some of these have now been tested in COPD (). The development of other classes of drugs acting on targets that were considered potentially usefulCitation17 was later abandoned because when tested on humans they proved to be poorly effective and/or induced major adverse effects. This is the case with the adenosine A2a receptor agonists (regadenoson and UK432.097) due to unsatisfactory results and unwanted effects on the cardiovascular system,Citation18,Citation19 anti-CXCL8 (ABX-CXCL) mAbs, which only slightly improved dyspnoea in COPD patientsCitation20 probably because such drugs only block free CXCL8 and not the bound one, TNF-α inhibitors (infliximab and etanercept) that had no beneficial effects in patients with mild, moderate, or severe COPDCitation21,Citation22 or for the treatment of acute exacerbations of COPD (AECOPDs),Citation23 likely because other proinflammatory cytokines drive the inflammatory processCitation24 and, furthermore, COPD patients had a significantly increased incidence of airway tumours and lung infections caused by TNF-α antibodies,Citation22 and anti-IL-1β mAbs (canakinumab and MEDI8986) because a lack of efficacy at least with regard to impact on the risk of AECOPDs, lung function, and HRQoL.Citation25

Table 1 Therapies That Inhibit Recruitment and Activation of the Cellular Components of Inflammation in COPD

Small Molecule Inhibitors

The main classes of small molecule inhibitors investigated for COPD include PDE inhibitors, CXCR2 antagonists, p38 MAPK inhibitors, PI3K inhibitors, and selectin antagonists.

Phosphodiesterase Inhibitors

There are 21 PDE genes in the human genome, which are organized into PDE families (PDE1 to PDE11), with many subtypes in each family.Citation26 PDEs catalyse the hydrolysis of cAMP and cGMP, thus controlling the intracellular levels of these cyclic nucleotides, their signalling pathways, and, ultimately, their biological responses.Citation26 Almost all cell types implicated in the pathophysiology of COPD are modulated by cAMP, which also controls the tone of the airway smooth muscle via the β2-adrenoceptor (β2-AR)-soluble adenylyl cyclase (sAC)-cAMP signalling pathway.Citation27 cGMP promotes vascular smooth muscle relaxation via the NO-soluble guanylyl cyclase-cGMP pathwayCitation28 and is also implicated in eliciting bronchodilation in human small airways via the β2-AR-sAC-cAMP pathway.Citation29 PDE4, PDE7, and PDE8 are cAMP-specific PDEs, while PDE5, PDE6, and PDE9 are cGMP-specific PDEs, and PDE1, PDE2, PDE3, PDE10, and PDE11 hydrolyse both cAMP and cGMP.Citation30

PDE4 inhibitors are likely the most studied because PDE4 is the predominant PDE expressed in T-cells, eosinophils, neutrophils, monocytes, and macrophages, and therefore its inhibition may also have inhibitory effects upon both inflammatory and immune cells.Citation30 Nevertheless, only one drug in this class, roflumilast N-oxide, has been approved for treating severe COPD.Citation31 However, roflumilast is not widely used because of a narrow therapeutic window with a range of dose-limiting side effects in the gastrointestinal tract and the central nervous system.Citation32 The addition of roflumilast is only recommended for patients who continue to exacerbate despite triple therapy with LABA + LAMA + ICS therapy or for those with a BEC of <100 cells/μL, especially if they have chronic bronchitis and a FEV1 <50%.Citation33

Therefore, other PDE4 inhibitors have been developed to be administered by the inhaled route to potentially reduce these adverse effects.Citation34 Unfortunately, the development of most of the inhaled PDE4 inhibitors has been discontinued due to modest effects in patients with COPD.Citation35 This lack of beneficial effects may be because inhaled drugs are generally designed to be retained in the lung and have no effect on systemic inflammation.Citation36 However, adding the inhaled PDE4 inhibitor, tanimilast, to inhaled triple therapy reduced AECOPDs.Citation37 Tanimilast exerts specific immunomodulatory effects linked to a T2 endotype and CD141 overexpression in DCs which suggests that it may have complementary anti-inflammatory effects to ICSs.Citation38 Tanimilast is currently being evaluated in two Phase 3 studies that are investigating its efficacy and safety as an add-on to triple maintenance therapy in subjects with COPD and chronic bronchitis (NCT04636801 and NCT04636814).

PDE4 is divided into four subfamilies: PDE4A, PDE4B, PDE4C, and PDE4D. The genes in each subfamily can express between 3 and 11 proteins, resulting in at least 25 distinct PDE4 protein isoforms.Citation39 PDE4 isoforms are essential spatiotemporal regulators of cAMP signalling due to their distinct intracellular distribution patterns, dynamic activity regulation, and isoform-specific regulation.Citation40 PDE4 isoform selective inhibitors do not yet exist. Therefore, it will be important to create novel isoform specific PDE4 inhibitors also because this approach may help to reduce side effects.Citation39 However, due to the structural similarities and highly conserved sequences of the isoforms, designing PDE4 isoform selective inhibitors is difficult.Citation30

Agents capable of simultaneously inhibiting PDE3 and PDE4 are also under development.Citation41 This approach induces additional or synergistic anti-inflammatory and bronchodilator benefits compared to PDE3 or PDE4 inhibition aloneCitation42 (). Furthermore, dual PDE3/4 inhibitors have been shown to improve mucociliary clearance.Citation43 Ensifentrine, a first-in-class inhaled “bifunctional” dual PDE3/4 inhibitor, exhibits both bronchodilator and anti-inflammatory activities.Citation43 The two Phase 3 ENHANCE (Ensifentrine as a Novel inHAled Nebulized COPD thErapy)-1 and −2 clinical trials, not yet published in full, demonstrated substantial improvements in lung function, a clinically significant reduction of approximately 40% in AECOPD rate following 24 weeks of treatment with favourable safety results, although the improvement in symptoms and quality of life reached statistical significance only in ENHANCE-1.Citation44,Citation45 The reduction in exacerbations was associated with a numerical reduction in circulating inflammatory biomarkers (IL-6, IL-8 and CRP).Citation46 The fact that ensifentrine was able to reduce the frequency and risk of exacerbations in patients with eosinophils >150 cells/μLCitation44 is fascinating because it suggests that in many patients, there would be no need to use ICSs. In ENHANCE-1, a higher percentage of patients were also under regular treatment for COPD (about 69% of subjects received LAMA or LABA and about 20% received ICS with concomitant LAMA or LABA), while in ENHANCE-2, this percentage was lower (about 55% of subjects received LAMA or LABA and about 15% received ICS with concomitant LAMA or LABA). Therefore, because few patients in these studies were on dual bronchodilation or triple therapy, it remains to be established what additional clinical benefit this drug brings to current standard of care.

Figure 1 Effect of PDE inhibition in COPD. Combined inhibition of PDE3 and PDE4 provides additive and synergistic anti-inflammatory and bronchodilator effects when compared to PDE3 or PDE4 inhibition alone. It also improves mucociliary clearance. The primary PDE implicated in the activity of the given cell is shown in red.

Figure 1 Effect of PDE inhibition in COPD. Combined inhibition of PDE3 and PDE4 provides additive and synergistic anti-inflammatory and bronchodilator effects when compared to PDE3 or PDE4 inhibition alone. It also improves mucociliary clearance. The primary PDE implicated in the activity of the given cell is shown in red.

Dual acting PDE4/PDE5 inhibitors, PDE4/PDE7 inhibitors, and PDE4/PDE1 inhibitors are also being investigated. The creation of pan-PDE inhibitors that can block a variety of PDE isoforms is another novel pharmacological approach being pursued.Citation41 An intriguing alternative is to create hybrid compounds with two or more pharmacophores, such as dual PDE4 inhibitors/muscarinic antagonists and dual PDE4 inhibitors/β2-agonists.Citation41

CXCR2 Antagonists

Due to their role in leukocyte chemotaxis, chemokines have long been thought to be involved in the onset and amplification of inflammatory reactions. One of the chemokine receptors, CXCR2, is expressed on a range of cell types and organs, which raises the possibility that these receptors have a wide functional involvement in the pathophysiology of COPD.Citation47 CXCR2 antagonists decreased neutrophil infiltration and tissue damage in the airways in pre-clinical models of cigarette smoke-induced acute neutrophilic inflammation in the lungs.Citation48 Some neutrophil CXCR2 antagonists have been investigated, but this drug class has only shown modest effects in patients with COPD, possibly due to redundancy in the chemokine network.Citation6 Navarixin, a dual CXCR1 and CXCR2 inhibitor, reduced inflammation and delayed the onset of the first exacerbation but also decreased the absolute neutrophil count, leading to the withdrawal of 18% of patients.Citation49 Danirixin, a selective CXCR2 antagonist, improved symptoms but its long-term administration caused a high incidence of AECOPDs and pneumonia, suggesting an adverse effect on host responses to infection.Citation50 Furthermore, in a small pilot study, there was no discernible difference between danirixin and placebo in terms of the % change from baseline in NETs, as determined by the histone-elastase immunoassay.Citation51 Ladarixin is another dual CXCR1 and CXCR2 antagonist that has been tested in a Phase 1 clinical trial (NCT04854642). It improved lung function and reduced neutrophilic airway inflammation in a corticosteroid-resistant model of cigarette smoke-induced influenza-A infection exacerbation.Citation52

p38 Mitogen-Activated Protein Kinase Inhibitors

MAPKs significantly influence chronic inflammation. The p38 MAPK subgroup consists of four isoforms (α, β, γ, δ),Citation53 with p38α MAPK being suggested to have a significant impact on COPD.Citation26 It is increased in bronchial epithelial cells, macrophages and CD20+ and CD8+ lymphocytes in COPD lungs.Citation54 Several extracellular triggers cause the p38 MAPK pathway to become activated, leading to inflammatory gene transcription with increased cytokine and chemokine production, particularly interleukin 1β (IL-1β), CXCL8 (IL-8), and TNF-α, which are associated with the neutrophilic endotype of COPDCitation26 (). Therefore, inhibiting p38 MAPK may be an effective treatment for patients with COPD.Citation6,Citation53

Figure 2 The role of p38 MAPK in the pathobiology of COPD. At the level of alveolar macrophages and other inflammatory cells, airborne pollutants, cigarette smoke, and microbial pathogens activate p38 MAPK. The p38 signaling pathway leads to increased cytokine and chemokine production, particularly interleukin (IL)-1β, IL-8, and tumor necrosis factor-α (TNF-α), which are associated with the neutrophilic endotype of COPD. Therefore, inhibiting p38 MAPK may be an effective treatment for patients with COPD.

Figure 2 The role of p38 MAPK in the pathobiology of COPD. At the level of alveolar macrophages and other inflammatory cells, airborne pollutants, cigarette smoke, and microbial pathogens activate p38 MAPK. The p38 signaling pathway leads to increased cytokine and chemokine production, particularly interleukin (IL)-1β, IL-8, and tumor necrosis factor-α (TNF-α), which are associated with the neutrophilic endotype of COPD. Therefore, inhibiting p38 MAPK may be an effective treatment for patients with COPD.

Some p38 MAPK inhibitors have been evaluated in patients with COPD. Losmapimod, an oral dual p38α/β inhibitor was not effective in reducing the moderate/severe COPD exacerbation rate in patients with blood eosinophils ≤2% and frequent exacerbations.Citation54 AZD7624, another p38α/β inhibitor, reduced the production of inflammatory mediators by primary human bronchial epithelial cells stimulated with polyinosinic: polycytidylic acid and lipopolysaccharide (LPS)-stimulated bronchoalveolar lavage fluid (BALF) macrophages to a greater extent than budesonide.Citation55 It was also more effective than budesonide in reducing IL-6 expression in bronchial epithelial cells, but without impacting AECOPD.Citation56 The inhaled narrow spectrum (p38α/γ) inhibitor RV568 improved lung function in a small 14-day clinical trial in COPD patients.Citation57 Acumapimod, a p38α inhibitor, improved FEV1 compared to placebo in severe AECOPD.Citation58 It decreased the number of hospital readmissions due to COPD exacerbations.Citation59 CHF6297, a selective p38α inhibitor, has been evaluated for safety, tolerability, and pharmacokinetics in healthy participants and for its anti-inflammatory effects in patients with COPD (NCT02815488). However, the results of these studies have not yet been made public. A systematic review with meta-analysis that included 10 RCTs in patients with COPD concluded that p38 MAPK inhibition was safe when compared with placebo, however this approach at best only caused a post-bronchodilator improvement in forced vital capacity.Citation59

Oral administration of this class of drugs at safe doses is likely insufficient to induce significant clinical benefit.Citation60 However, inhaled p38 MAPK inhibitors may lead to a higher local drug concentrations in the lung. An alternative approach could be the development of selective inhibitors of the α-δ subgroups. However, it is now recognized that the transforming growth factor-activated kinase-1 and mixed-lineage kinase are hyperactivated due to p38 MAPK inhibitors’ blocking of the upstream MAPK kinases, which in turn causes the c-Jun N-terminal kinase to become overactive.Citation61 These observations suggest a need to develop drugs that target downstream substrates in this signalling cascade.

Use of narrow spectrum kinase inhibitors, a new class of pharmaceutical agents that simultaneously targets key kinases involved in both innate and adaptive immune cell signalling, such as p38 MAPK, spleen tyrosine kinase (Syk), and Src family kinases (SFK), which include Src and lymphocyte-specific protein tyrosine kinase, is an additional option.Citation62 PUR1800 is a novel dry powder iSPERSE formulation of RV1162, a narrow spectrum kinase inhibitor, targeting p38 MAPK, Src and Syk kinases. In contrast to fluticasone propionate, it reduced TNF-α-induced cytokine release from primary bronchial epithelial cells of healthy volunteers and COPD patients.Citation63 The preliminary pharmacokinetic data indicate that PUR1800 results in low and consistent systemic exposure in COPD patients when administered via oral inhalation.Citation64

Phosphoinositide 3-Kinase Inhibitors

PI3K is an enzyme that catalyses the production of phosphatidylinositol-3,4,5-triphosphate and is involved in the activation of macrophages and neutrophils, as well as the hyperphosphorylation and ubiquitination of HDAC2, thus influencing sensitivity of inflammatory cells to corticosteroids.Citation65 The function of this enzyme increases in patients with COPD.Citation65 Increased migratory speed and decreased directionality toward the cytokine gradient were seen with neutrophils from patients with COPD, and these changes were reversed by PI3Kδ inhibition.Citation66 Furthermore, PI3Kδ inhibition reduces pro-inflammatory cytokine secretion and improves survival rates following infection.Citation67

In a proof-of-concept trial, adding nemiralisib, an inhaled PI3Kδ inhibitor, to usual care was associated with a more effective recovery from AECOPDs and improved lung function but also resulted in cough.Citation68 However, in another trial, nemiralisib had no positive impact on lung function or relapses after an AECOPD.Citation69 Furthermore, this drug altered neutrophil migratory behaviour in stable patients with COPD, but not in those with AECOPD, despite improving lung function in the latter group.Citation70

CHF6523 is another inhaled PI3K inhibitor, currently in clinical development and has been investigated for safety and tolerability in patients with COPD.Citation6 AZD8154, a PI3Kγδ inhibitor, has been tested in healthy volunteers,Citation71 while RV1729, another PI3Kγδ inhibitor, is being investigated in patients with COPD (NCT02140346). GSK045, a selective PI3Kδ inhibitor, and ZSTK474, a pan PI3K inhibitor, are other potential drugs for inhibiting PI3K in COPD.Citation6

Selectin Antagonists

The selectin family consists of three members (E-, L-, and P-selectin). While L-selectin is constitutively expressed on circulating leukocytes, E- and P-selectin are expressed on the endothelium.Citation17 Selectins regulate inflammatory cell migration from the bloodstream into the lungs.Citation17 E-selectin, which is up-regulated on endothelial cells in the airways of COPD patients,Citation72 is required for the first attachment of neutrophilic granulocytes to endothelial cells in the airways.Citation73 Therefore, targeting these molecules may provide another approach to reducing inflammation in the lungs of patients with COPD.

Inhaled bimosiamose, a synthetic pan-selectin antagonist, decreased airway inflammation and improved lung function in patients with COPD, confirming the critical function of selectins in the movement of inflammatory cells.Citation74 Also, rivipansel, a pan-selectin antagonist, and uproleselan, a selective inhibitor of E-selectin, might be evaluated in the treatment of COPD.Citation6

Developing inhibitors of selectin-ligand interactions in vivo is tricky because extremely potent inhibitors could have undesirable effects on healing processes, and weak inhibitors may not sufficiently act on the pathological processes of serious inflammatory diseases.Citation75 It has been suggested that a clinically effective inhibitor is likely to inhibit at least two selectins.Citation75

Biologic Therapies

Anti-IL-17A mAbs, collectins, anti-IL-5 mAbs, anti-IL13/IL4 mAbs, anti-TSLP mAbs, and anti-IL-33 mAbs are among the biological therapies that have the potential to treat COPD.Citation24,Citation76

Anti-IL-17A mAbs

Along with IL-22, IL-17A and IL-17F are linked to an increase in neutrophils in the airways, which results in chronic inflammation and increased mucus production and smooth muscle mass in the airways.Citation76 To reduce neutrophil recruitment and airway inflammation, many anti-IL-17A and anti-IL-17 receptor A (IL-17RA) mAbs are being developed,Citation77 but CNTO 6785 is the only anti-IL-17A mAb that has been investigated in patients with COPD. It caused a small improvement in FEV1 but did not interfere with the other primary or secondary endpoints.Citation78 Furthermore, more AECOPDs appeared in the CNTO 6785 arm than in the placebo arm at week 16. Brodalumab, which is a mAb that targets IL-17RA, secukinumab, a fully human anti-IL-17A of the immunoglobulin (Ig)G isotype, and CCJM112, a novel fully human anti-IL-17A IgG mAb that binds with similar affinity to both human IL-17A and IL-17AF, have not yet been tested in patients with COPD.Citation76

Collectins

SP-A and SP-D are termed collectins because they contain collagen and are functional lectins.Citation79 By competing with LPS for CD14 binding on alveolar macrophages, SP-A and SP-D decrease TNF-α production.Citation79 Furthermore, they modify inflammatory cytokine production following macrophage activation by cytokines or pathogen-associated molecular patterns.Citation17 Pulmonary SP-D levels in COPD are lower than in smokers.Citation80 Supplementation with recombinant forms of SP-A and SP-D might correct collectin deficiencies in inflammatory respiratory disorders. The trimeric proteins SP-A and SP-D are available as small recombinant forms of human fragments (rfhSP-A and rfhSP-D).Citation78 AT-100 is an engineered version of the endogenous human protein rhSP-D. Local treatment of rfhSP-D reduces inflammation, ceramide production, and epithelial cell death caused by cigarette smoke in mice,Citation81 but it has not yet been investigated in patients with COPD.

Targeting IL-5

As already mentioned, a subgroup of patients with COPD has elevated blood and airway eosinophils and elevated sputum IL-5 concentrations.Citation82 There is a relationship between IL-5 levels and the quantity of eosinophils in the sputum of COPD patients.Citation83 Moreover, there is proof that virus-induced AECOPD causes a rise in soluble IL-5Rα.Citation84 Blocking IL-5 may likely prevent or decrease eosinophil-mediated inflammation due to the fast death of eosinophils in the absence of IL-5.Citation76 Mepolizumab, a mAb that blocks IL-5, and benralizumab, an anti-IL-5Rα mAb, lower the rate of moderate and severe exacerbations in the highly selected population of individuals with COPD and higher levels of blood eosinophilsCitation85,Citation86 (). However, substantial eosinophil clearance from the airways in many COPD patients does not result in appreciable clinical improvement.Citation87 This is probably because human lung-resident eosinophils are present independently of IL-5.Citation88 Nonetheless, both mepolizumab (NCT04133909 and NCT04075331) and benralizumab (NCT04053634 and NCT04098718) are still being investigated in patients having COPD exacerbations and higher levels of blood eosinophils.

Figure 3 Monoclonal antibodies targeting T2 cytokines in COPD.

Abbreviations: ICL2, group 2 innate lymphoid cells; IL-4Rα, interleukin-4 receptor subunit α; IL-5Rα, interleukin-5 receptor subunit α; IL-13Rα, interleukin-13 receptor subunit α1.
Figure 3 Monoclonal antibodies targeting T2 cytokines in COPD.

Targeting IL13/IL4

IL-4 and IL-13 are cytokines that promote eosinophilic recruitment, IgE production, mucus hypersecretion, and airway fibrosis and remodeling.Citation89 They signal through a common receptor, IL-4Rα, expressed by airway epithelial cellsCitation76 (). IL-4 and IL-13 can drive eosinophilic inflammation through the production of chemoattractants such CC-chemokine ligand 26 from airway epithelial cells.Citation89 Patients with eosinophilic COPD and concomitant emphysema had significant levels of sputum matrix metalloprotease-12 (MMP-12), which is produced by alveolar macrophages when IL-13 is present.Citation76 Furthermore, it has been demonstrated that individuals with stable COPD or during an AECOPD have greater levels of group 2 innate lymphoid cells, which can produce IL-13.Citation90

Lebrikizumab, a humanized mAb that binds to soluble IL-13 and inhibits activation of IL-4Rα and IL-13Rα1 heterodimers, has been examined in patients with COPD with a history of exacerbations despite using ICS and at least one long-acting bronchodilator inhaler drug (NCT02546700).Citation76 Although the complete results have not yet been made public, early data indicates that lebrikizimab did not affect the AECOPD rate compared to placebo. Consequently, the clinical development of lebrikizumab for the treatment of patients with COPD has been halted indefinitely. Nevertheless, since IL-13 directly affects airway contractility and is critical for mucus production and remodelingCitation24 and airflow limitation and mucus hypersecretion are commonly treatable features in COPD, it would be interesting to conduct an RCT with lebrikizimab in patients belonging to the GOLD Group B, i.e. with COPD that is symptomatic but only slightly exacerbating.

A pivotal 52-week study, in which 939 adults aged 40 to 80 years with moderate-to-severe COPD and T2 inflammation, as measured by BECs ≥300 cells/µL, received dupilumab (n = 468) or placebo (n = 471), added to standard-of-care inhaled therapy, has shown that this biologic, a mAb that binds to IL4Rα and inhibits both IL-4 and IL-13 signaling, cut exacerbations by 30% compared with placebo (p<0.001).Citation91 The annualized rate of moderate or severe exacerbations was 0.78 with dupilumab and 1.10 with placebo. It also caused an increase in FEV1 from baseline of 160 mL at 12 weeks compared with 77 mL for placebo (p<0.001), with the benefit versus placebo sustained through week 52. Furthermore, it improved patient-reported HRQoL as measured by SGRQ and reduced the severity of respiratory symptoms of COPD as measured by Evaluation Respiratory Symptoms: COPD (E-RS: COPD) Scale. Another phase 3 pivotal trial (NCT04456673) is also assessing the efficacy, safety, and tolerability of dupilumab in COPD patients with evidence of T2 inflammation, with data expected in 2024.

Targeting Thymic Stromal Lymphopoietin (TSLP)

TSLP, an IL-7-like cytokine, is an upstream epithelial alarmin that plays an important role in the regulation of T2 immunity.Citation76 TSLP operates on a variety of cells, including dendritic cells, T-cells, mast cells, innate lymphoid cells, and eosinophils, causing the production of a variety of interleukins, including IL-4, IL-5, and IL-13, which leads to airway eosinophilia and hyperresponsiveness. It is also capable of exerting multipotential pathogenic effects beyond T2 inflammationCitation76 ().

Figure 4 Targeting alarmins in COPD.

Abbreviations: DC, dendritic cell; EGFR, epidermal growth factor receptor; ILC2, group 2 innate lymphoid cells; IL-1RAP, IL-1 receptor accessory protein; IL-33red, reduced IL-33; IL-33ox, oxidated IL-33; MC, mast cell; RAGE, receptor for advanced glycation end products; ST2, suppression of tumorigenicity 2; TSLP, thymic stromal lymphopoietin; TSLPR, TSLP receptor.
Figure 4 Targeting alarmins in COPD.

TSLP is constitutively expressed in the airway smooth muscle of patients with COPD, where it probably interacts with and influences local immune cells.Citation92 In addition, viruses and T1 cytokines can induce COPD epithelial cells to overproduce TSLP,Citation93 suggesting that TSLP may play a role in AECOPD. However, it has been hypothesized that TSLP produced by DCs serves as a crucial molecular checkpoint to limit IL-1β-mediated effector responses through a negative feedback loop, potentially reducing the severity of the inflammatory response to injury.Citation94

Tezepelumab, a first-in-class human IgG2/λ mAb that binds to TSLP and inhibits its interaction with the TSLP receptor complex,Citation76 is currently under investigation as adjunctive therapy in preventing AECOPD (NCT04039113) and in reducing airway inflammation in patients with COPD (NCT05507242). Ecleralimab, an antibody fragment that is part of the G1/λ immunoglobulin isotype subclass and binds to TSLP, is being evaluated in a Phase 2 study in patients with COPD (NCT04882124).

Targeting IL-33

IL-33, a member of the IL-1 superfamily, is released in a reduced form (IL-33red) from damaged epithelial cells and acts as an alarmin to notify the immune system of harm, thereby promoting the inflammatory response.Citation95 It exerts its biologically pro-inflammatory effects by activating its receptor, a heterodimeric complex composed of the IL-1 receptor accessory protein and tumor suppressor protein 2 (ST2)Citation96 ().

IL-33 stimulates lung epithelial and endothelial cells to produce and release IL-6 and IL-8, resulting in an influx of neutrophils that damage lung tissue by producing elastases and proteases.Citation97 This, in turn, promotes pulmonary fibrosis and impairs lung function. Additionally, in non-atopic patients with COPD, IL-33 has been connected to the emergence of eosinophilic airway inflammation.Citation98

Itepekimab, a human IgG4 mAb targeting IL-33, added to the standard of care, did not significantly reduce the annualized rate of moderate-to-severe AECOPDs compared to placebo after 52 weeks of treatment.Citation99 However, it decreased the AECOPD rate and improved lung function in former COPD smokers. Itepekimab is under investigation in two Phase 3 trials to compare its efficacy to placebo on the annualized rate of mild to moderately severe AECOPDs in a 52-week placebo-controlled trial in former smokers with mild to moderately severe COPD (NCT04701983 and NCT04751487).

Astegolimab, an anti-ST2 mAb, did not significantly reduce the AECOPD rate but improved health status compared with placebo in patients with moderate-to-very severe COPD who were administered this drug subcutaneously in a 48-week RCT.Citation100 Two ongoing trials are evaluating the efficacy and safety of astegolimab compared with placebo in participants with COPD who are former or current smokers and have a history of frequent exacerbations (NCT05037929 and NCT05595642).

Tozorakimab (MEDI3506) is a human IgG1 mAb that prevents IL-33 signaling. It possesses a higher affinity for the reduced form (IL-33red) than soluble version of ST2 (sST2) and a rapid association rate comparable to that of sST2.Citation101 In primary human cells and in a humanized IL-33 murine model of airway inflammation, tozorakimab suppressed ST2-dependent inflammation.Citation100 It binds IL-33red, blocking oxidation and, consequently, the action of the oxidated form (IL-33ox) via receptor for advanced glycation end products/epidermal growth factor receptor.Citation102 Elimination of IL-33ox enhanced wound healing in vitro following epithelial injury. Tozorakimab is being investigated in several RCTs in patients with COPD. One of them (NCT04631016) will assess its efficacy and safety in patients with moderate to severe COPD and chronic bronchitis. Furthermore, two identical global Phase 3 studies (NCT05166889 and NCT05158387) are designed to evaluate the efficacy and safety of two different doses of tozorakimab administered for 52 weeks in patients with symptomatic COPD and a history of ≥2 moderate exacerbations or ≥1 severe AECOPD in the previous 12 months, despite receiving optimized treatment (triple therapy or dual therapy if triple therapy is not indicated or contraindicated) at a stable dose for at least 3 months before enrolment.

Targeting the Products of the Cellular Components of Inflammation

Several attempts have been made to develop therapies that target the products of the cellular components of inflammation. Some of these have been evaluated in humans ().

Table 2 Therapies That Antagonize the Products of the Cellular Components of Inflammation in COPD

Matrix Metalloproteinase Inhibitors

MMPs are proteolytic enzymes that break down basement membrane and ECM proteins.Citation103 Consequently, these enzymes control airway remodelling, a critical pathogenetic aspect of COPD. Furthermore, proteolytic damage in the lungs can lead to elastin loss and emphysema formation, which are linked to reduced lung function in COPD patients. MMPs, particularly those derived from macrophages, have been linked to the development of COPD in several human and animal studies. MMPs have been shown to influence macrophage activation rather than working directly to destroy elastin, promoting inflammation and the disease course.Citation104,Citation105 In addition to their activity on the ECM, MMPs can act on various other substrates.

In humans, there are 23 different MMPs.Citation103 MMP-9 and MMP-12 are those particularly involved in tissue remodelling and degradation of the ECM components and likely perpetuate and regulate lung inflammation when released by neutrophils and macrophages.Citation6 There is currently limited research investigating the use of MMP inhibitors in patients with COPD.Citation6 While V85546, a selective MMP-12 inhibitor, has completed Phase 1 clinical testing, AZD1236, a dual MMP-9/12 inhibitor, was ineffective in patients with moderate-to-severe COPD.Citation106 Andecaliximab, a recombinant chimeric IgG4 mAb, has completed a Phase 1 clinical trial and is now being investigated in patients with COPD (NCT02077465). FP-025, an inhibitor of MMP-12, is currently being tested for efficacy in patients with asthma (NCT03858686), but to date no studies have been described in patients with COPD.

A novel series of compounds with single-digit nanomolar inhibition against MMP-12 have been discovered using the high-throughput screening by nuclear magnetic resonance technique.Citation107 Animals exposed to porcine pancreatic elastase but given MMP-12 inhibitors (MMP408, or lead agents compound 25, and 26) showed a substantial reduction in emphysema-like pathologies.

Drug repurposing might be an appealing technique for identifying current medicines with unique MMP inhibitory action. For example, doxycycline, a broad-spectrum antibiotic, has been shown to reduce MMP12 activity in vitro.Citation108 Despite its safety profile, antibiotic medication repurposing requires close monitoring to prevent the possibility of antibiotic resistance.

Neutrophil Elastase Inhibitors

NE is stored in neutrophil azurophilic granules and is released when neutrophils are activated by exposure to cigarette smoke or inflammationCitation107 It causes alveolar expansion and disintegration by proteolyzing lung tissues. Furthermore, NE induces airway epithelial cells to express MUC5AC, causing secretory cell hyperplasia and mucin synthesis.Citation109 Finally, NE stimulates macrophages, causing them to release active MMPs, which cause lung damage and emphysema, and proinflammatory cytokines, which cause inflammation.Citation110

In individuals with COPD who have an overexpression of this enzyme, restoring the balance between NE and endogenous antiproteases by blocking NE might be a treatment option.

However, alvelestat, an orally available, potent, and selective inhibitor of human NE, did not induced changes in lung function or inflammation when added to budesonide/formoterol maintenance therapy in patients with COPD.Citation111 Furthermore, combined with tiotropium in patients with COPD, it failed to demonstrate a clinical benefit or an impact on biomarkers of tissue deterioration.Citation112 Nevertheless, a trial is investigating the effects of alvelestat on blood and sputum biomarkers in patients with PiZZ α1-antitrypsin deficiency (AATD) (NCT03636347).

Sivelestat is a NE inhibitor available only in Japan and South Korea to treat acute lung damage and acute respiratory distress syndrome in individuals with a systemic inflammatory response. Other NE inhibitors, such as BAY 85–8501, CHF6333, and lonodelestat, which could be useful in COPD, are currently in different stages of clinical development, but there is still no information on their effects on COPD.Citation6

Interestingly, polysulfated glycosaminoglycans (GAGs) are potent anti-elastase drugs with multiple anti-inflammatory properties.Citation113 In one double-blind, placebo-controlled pilot research, inhaling unfractionated heparin, the prototypical drug in this class, as a COPD treatment resulted in enhanced lung function, highlighting the potential of GAG therapy for the treatment of chronic lung disorders.Citation114

However, focusing solely on anti-NE activity does not appear to be sufficient to change the ongoing inflammatory state in the airways in a way that would alter the trajectory of clinical outcomes.Citation115 It has been suggested that employing combination therapy and/or multi-function drugs that have antiprotease and anti-inflammatory propertiesCitation115 or, alternatively developing an airway delivery system for these therapeutics, allowing activity not only on the free NE but also on the form bound to the extracellular vesicles found in the BALF of COPD patientsCitation116 may be a more successful strategy.

Dipeptidyl Peptidase 1 Inhibitors

DPP1 transforms the neutrophil serine protease pool, including NE, cathepsin G, proteinase 3, into catalytically active forms and activates proteases in other immune cell types, such as chymases in mast cells.Citation117,Citation118 Therefore, it may be a promising target for new drugs to treat COPD. GSK2793660, an irreversible DPP1 inhibitor, was stopped after a Phase 1 clinical study due to drug-related side effects and lack of impact on biomarkers.Citation119 This drug only decreased the activity of downstream serine proteases in 20% of the cases, although it suppressed most of DPP1. Brensocatib, an oral reversible DPP1 inhibitor, has been investigated for its effects in adult patients with non-cystic fibrosis bronchiectasis,Citation120 but its effects on COPD are unknown. Nevertheless, it has been proposed that limiting neutrophil protease activity before cell discharge into the circulation rather than at the inflammatory site, as well as blocking several inflammatory targets such as elastase, but also cathepsin G and proteinase-3, may be a crucial driver of success.Citation121

Granzyme B Inhibitors

When released from the cytoplasmic secretory granules of CD8+ T lymphocytes, granzyme B, which is a multifunctional serine protease and is also called α-1-antichymotrypsin (ACT), can start a cascade of DNA disruption that causes the apoptotic death of target cells, such as bronchial epithelial cells, which may lead to tissue damage and remodelling.Citation122 Furthermore, it increases inflammation by enhancing cytokine activity or sequestered growth factor release.Citation123 Thus, using targeted granzyme B inhibitors to treat COPD seems plausible. Serpina3n, a serine protease inhibitor that inhibits granzyme B,Citation124 an engineered extracellular granzyme B inhibitor in which the reactive center loop of human extracellular ACT is replaced with serpina3n,Citation125 VTI-1002, a first-in-class potent small-molecule inhibitor of granzyme B,Citation126 proteinase inhibitor 9 (PI-9), an endogenous GZMB inhibitor that can bind to GZMB in the cytosol of activated CD8+ T cells,Citation127 serine protease inhibitor 6 (SPI-6), the mouse homolog of PI-9Citation128 are potentially interesting molecules but have not yet been investigated on humans. Furthermore, cefpiramide, a third-generation semi-synthetic cephalosporin antibiotic, and mupirocin, produced by Pseudomonas fluorescens, have the propensity to inhibit the active site of granzyme BCitation129 and may therefore be of value in treating COPD.

α1-Antitrypsin Replacement Therapy

AAT modulates neutrophilic inflammation and inhibits proteases.Citation130 There is a proteolytic interplay between AAT, MMP and neutrophil elastase (NE) (). In patients with AATD, augmentation treatment with pure intravenous AAT is essential, although it reaches the lung in a somewhat inactive state.Citation130 New intravenous formulations could solve this problem. AAT Modified Process (AAT-MP), with improved AAT purity, is in a 3-year Phase 3 trial (NCT01983241). INBRX-101, a recombinant human AAT Fc fusion protein that extends the half-life of AAT in blood by enabling it to be recycled, is under Phase 1 study (NCT03815396).

Figure 5 Proteolytic interplay between α1-antitrypsin (AAT), matrix metalloproteinases (MMPs) and neutrophil elastase (NE). Several proteases, including neutrophil elastase (NE) and MMP-9 and MMP-12, are involved in COPD. Therefore, inhibiting a single enzyme with a NE inhibitor or a MMP inhibitor may not have a significant therapeutic impact. AAT inhibits NE and reduces macrophage MMP-12 synthesis.

Figure 5 Proteolytic interplay between α1-antitrypsin (AAT), matrix metalloproteinases (MMPs) and neutrophil elastase (NE). Several proteases, including neutrophil elastase (NE) and MMP-9 and MMP-12, are involved in COPD. Therefore, inhibiting a single enzyme with a NE inhibitor or a MMP inhibitor may not have a significant therapeutic impact. AAT inhibits NE and reduces macrophage MMP-12 synthesis.

The inhaled route for AAT is also of interest as it can act directly on the target organ when administered in this way.Citation130 Inhaled AAT administered for 50 weeks significantly reduced the number of symptomatic exacerbations of Anthonisen type I, with a propensity to improve FEV1 but without affecting the time to first exacerbation.Citation131 A trial is evaluating inhaled AAT in patients with AATD who have moderate or severe airflow limitation and have not experienced two or more moderate or one or more severe exacerbations of COPD during the past year (NCT04204252). Developing gene therapy to express AAT using various vector systemsCitation130 or administering AAT treatment via the transepidermal routeCitation132 are intriguing alternative approaches currently under investigation. A study is evaluating the safety and tolerability of subcutaneous AAT (NCT04722887).

Newer approaches that are predominantly in the preclinical phase tend to silence AAT production, enhance protein folding and secretion, or promote AAT degradation.Citation133 A folding corrector called VX-864 aims to increase Z-AAT secretion and activity. It is starting a Phase 2 study (NCT04474197) to show its safety and effectiveness in PiZZ patients.

Targeting Neutrophil Extracellular Traps

Great attention has been given to the dysregulation of NETs that play an essential role in the innate immune system against infections. NETs have been identified in the airways of patients with COPD with chronic neutrophilic inflammation.Citation134 NETs result from the suicidal death of neutrophils during the attack of a pathogen or lesion, with the release of decondensed chromatin entangled with antimicrobial peptides to trap and capture pathogens to be an effective mechanism against invading microorganisms thus controlling overwhelming infections.Citation135 However, an aberrant and massive NET formation can directly induce epithelial and endothelial cell death, impairing lung function and accelerating disease progression.Citation6,Citation136

Interfering with the NETopathic inflammation pathways could help develop innovative therapeutics for COPD.Citation135 Heparin, which interferes with neutrophil autophagy, suppresses histones, prevents platelets-histone interaction, and blocks high mobility group box-1, deoxyribonuclease, which acts on and cleaves DNA matrices, and reduces the infiltration of neutrophils hence playing role in inhibition/reduction of NETs, and hydroxychloroquine, which mediates MMPs-TIMPs interaction that helps in maintaining homeostasis of extracellular matrix, are potential anti-NET therapeutics.Citation6,Citation135 However, also protease inhibitors, CXCR2 antagonists, macrolides, and DPP1 inhibitors are drug classes that can specifically or non-specifically target NET formation,Citation137 although a pilot study did not find an effect of the reversible CXCR2 antagonist danirixin on NET formation.Citation49 Blockade of IL-1β and IL-17 could also be a valid strategy for interfering with the NETopathic inflammation pathways.Citation135

Conclusion

Finding therapies that can reduce COPD-related inflammation and prevent the disorder from becoming worse is crucial. Recent discoveries have resulted in developing novel drugs for several novel potential targets directly involved in the inflammatory process.Citation6,Citation14 To date, most of these new therapies are in preclinical or early clinical development and the results from clinical trials in patients with COPD are eagerly awaited.Citation138 However, the redundancy of the actions of signal-transmitting mediators involved in the complexity of the inflammatory response in COPD suggests that there will likely be several failures of the different approaches discussed above.Citation139

Since COPD is heterogeneous, with many endotypes and phenotypes reflecting different pathophysiological mechanisms, the endo/phenotypic characterization of the inflammatory profile of patients with COPD may allow the identification of a reasonably homogeneous population such that there is a high possibility that the disease being treated is driven by the specific target pathway of the drug being tested. This strategy involves defining more specific management approaches.Citation1 As recently highlighted by the Lancet commission, the new COPD therapies must be specific and target the exact biological pathways or endotypes responsible for disease manifestation to have the greatest impact and eventually abolish the disease.Citation1 Characterization of the heterogeneity of the inflammatory signature associated with COPD could pave the way for personalized medicine by identifying new and effective therapeutic approaches for COPD.Citation140 For this reason, many clinical trials are underway to characterize the impact of some novel molecules that act directly against the inflammatory process in treating distinct subgroups of COPD patients.Citation6

Nevertheless, other possible approaches, such as stem cell-based regenerative therapy and derivative productsCitation141 and modulators of cystic fibrosis transmembrane conductance regulator,Citation142 may indirectly modulate inflammation in COPD and do not require accurate endotypic precision. The scientific community eagerly awaits the results of this experimental and mainly clinical research in the hope of improving treatments for such a challenging disease.

Abbreviations

AAT, α1-antitrypsin, AATD, AAT deficiency; ACT, α-1-antichymotrypsin; AECOPD, acute exacerbations of COPD; AR, adrenoceptor; BALF, bronchoalveolar lavage fluid; BEC, blood eosinophil count; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; CS corticosteroid; COPD, chronic obstructive pulmonary disease; CXCR, CXC chemokine receptor; DC, dendritic cell; DPP1, dipeptidyl peptidase 1; ECM, extracellular matrix; FEV1, forced expiratory volume in the first second; GR, glucocorticoid receptor; HDAC. Histone deacetylase; HRQoL, health-related quality of life; ICS, inhaled corticosteroid; Ig, immunoglobulin; IL, interleukin; IL-4Rα, interleukin-4 receptor subunit α; IL-5Rα, interleukin-5 receptor subunit α; IL-17RA, IL-17 receptor A; IL-33ox, IL-33 oxidated form; IL-33red, IL-33 reduced form; LABA, long-acting β2-agonist; LAMA, long-acting muscarinic antagonist; LPS, lipopolysaccharide; mAb, monoclonal antibody; MAPK, mitogen-activated protein kinase; MMP, matrix metalloprotease; NE, neutrophil elastase; NETs, neutrophil extracellular traps; PDE, phosphodiesterase; PI3K, phosphoinositide 3-kinase; RCT, randomized controlled trial; Rfh, recombinant form of human fragments; sAC, soluble adenylyl cyclase; SP, surfactant protein; Syk, spleen tyrosine kinase; ST2, tumor suppressor protein 2; sST2, soluble version of ST2; T, type; TNF-α, tumor necrosis factor-α; TSLP, thymic stromal lymphopoietin.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

M.C. participated as a faculty member and advisor in scientific meetings and courses under the sponsorship of Abdi Ibrahim, Almirall, AstraZeneca, Boehringer Ingelheim, Chiesi Farmaceutici, Cipla, Edmond Pharma, GlaxoSmithKline, Glenmark, Lallemand, Mankind Pharma, Menarini Group, Mundipharma, Novartis, Pfizer, Sanofi, Teva, Verona Pharma, and Zambon and is or was a consultant to ABC Farmaceutici, AstraZeneca, Chiesi Farmaceutici, Edmond Pharma, Lallemand, Novartis, Ockham Biotech, Verona Pharma, and Zambon. N.A.H. received honoraria for serving as advisor or consultant for GSK, AstraZeneca, Sanofi, Regeneron, Boehringer Ingelheim, Verona, Amgen, Genentech, Novartis and Teva. His institution received research grant support of his behalf from GSK, Genentech, Sanofi, Teva, Novartis, and Astra Zeneca. C.P.P. has acted as a consultant to Eurodrug, Recipharm, Glycosynnovation and PrEP Biopharma. C.P.P. also holds equity in Verona Pharma. M.G.M. participated as a faculty member and advisor in scientific meetings and courses under the sponsorship of ABC Farmaceutici, Almirall, AstraZeneca, Chiesi Farmaceutici, GlaxoSmithKline, and Novartis and was a consultant to Chiesi Farmaceutici and GlaxoSmithKline. Her department was funded by GlaxoSmithKline and Novartis. The authors have no other 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 apart from those disclosed.

References

  • Stolz D, Mkorombindo T, Schumann DM, et al. Towards the elimination of chronic obstructive pulmonary disease: a lancet commission. Lancet. 2022;400(10356):921–972. doi:10.1016/S0140-6736(22)01273-9
  • Gamble E, Qiu Y, Wang D, et al. Variability of bronchial inflammation in chronic obstructive pulmonary disease: implications for study design. Eur Respir J. 2006;27(2):293–299. doi:10.1183/09031936.06.00027705
  • Deng F, Zhong S, Yu C, et al. Abnormal neutrophil polarization in chronic obstructive pulmonary disease and how cigarette smoke extracts attract neutrophils. Ann Transl Med. 2022;10(8):472. doi:10.21037/atm-22-1480
  • David B, Bafadhel M, Koenderman L, De Soyza A. Eosinophilic inflammation in COPD: from an inflammatory marker to a treatable trait. Thorax. 2021;76(2):188–195. doi:10.1136/thoraxjnl-2020-215167
  • Burgoyne RA, Fisher AJ, Borthwick LA. The role of epithelial damage in the pulmonary immune response. Cells. 2021;10(10):2763. doi:10.3390/cells10102763
  • Matera MG, Cazzola M, Page C. Prospects for COPD treatment. Curr Opin Pharmacol. 2021;56:74–84. doi:10.1016/j.coph.2020.11.003
  • Cazzola M, Ora J, Calzetta L, Rogliani P, Matera MG. Advances in inhaled corticosteroids for the treatment of chronic obstructive pulmonary disease: what is their value today? Expert Opin Pharmacother. 2022;23(8):917–927. doi:10.1080/14656566.2022.2076592
  • Barnes PJ. Glucocorticosteroids. Handb Exp Pharmacol. 2017;237:93–115. doi:10.1007/164_2016_62
  • Adcock IM, Bhatt SP, Balkissoon R, Wise RA. The use of inhaled corticosteroids for patients with COPD who continue to smoke cigarettes: an evaluation of current practice. Am J Med. 2022;135(3):302–312. doi:10.1016/j.amjmed.2021.09.006
  • Plumb J, Gaffey K, Kane B, et al. Reduced glucocorticoid receptor expression and function in airway neutrophils. Int Immunopharmacol. 2012;12(1):26–33. doi:10.1016/j.intimp.2011.10.006
  • Milara J, Lluch J, Almudever P, Freire J, Xiaozhong Q, Cortijo J. Roflumilast N-oxide reverses corticosteroid resistance in neutrophils from patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2014;134(2):314–322. doi:10.1016/j.jaci.2014.02.001
  • Li LB, Leung DY, Martin RJ, Goleva E. Inhibition of histone deacetylase 2 expression by elevated glucocorticoid receptor beta in steroid-resistant asthma. Am J Respir Crit Care Med. 2010;182(7):877–883. doi:10.1164/rccm.201001-0015OC
  • Pavord ID, Lettis S, Anzueto A, Barnes N. Blood eosinophil count and pneumonia risk in patients with chronic obstructive pulmonary disease: a patient-level meta-analysis. Lancet Respir Med. 2016;4(9):731–741. doi:10.1016/S2213-2600(16)30148-5
  • Matera MG, Calzetta L, Annibale R, Russo F, Cazzola M. Classes of drugs that target the cellular components of inflammation under clinical development for COPD. Expert Rev Clin Pharmacol. 2021;14(8):1015–1027. doi:10.1080/17512433.2021.1925537
  • Barnes PJ. Identifying molecular targets for new drug development for chronic obstructive pulmonary disease: what does the future hold? Semin Respir Crit Care Med. 2015;36(4):508–522. doi:10.1055/s-0035-1555611
  • Barnes PJ. Targeting cytokines to treat asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. 2018;18(7):454–466. doi:10.1038/s41577-018-0006-6
  • Cazzola M, Page CP, Calzetta L, Matera MG. Emerging anti-inflammatory strategies for COPD. Eur Respir J. 2012;40(3):724–741. doi:10.1183/09031936.00213711
  • Prenner BM, Bukofzer S, Behm S, Feaheny K, McNutt BE. A randomized, double-blind, placebo-controlled study assessing the safety and tolerability of regadenoson in subjects with asthma or chronic obstructive pulmonary disease. J Nucl Cardiol. 2012;19(4):681–692. doi:10.1007/s12350-012-9547-4
  • Haskó G, Linden J, Cronstein B, Pacher P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 2008;7(9):759–770. doi:10.1038/nrd2638
  • Mahler DA, Huang S, Tabrizi M, Bell GM. Efficacy and safety of a monoclonal antibody recognizing interleukin-8 in COPD: a pilot study. Chest. 2004;126(3):926–934. doi:10.1378/chest.126.3.926
  • van der Vaart H, Koëter GH, Postma DS, Kauffman HF, ten Hacken NH. First study of infliximab treatment in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2005;172(4):465–469. doi:10.1164/rccm.200501-147OC
  • Rennard SI, Fogarty C, Kelsen S, et al. The safety and efficacy of infliximab in moderate to severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;175(9):926–934. doi:10.1164/rccm.200607-995OC
  • Aaron SD, Vandemheen KL, Maltais F, et al. TNFα antagonists for acute exacerbations of COPD: a randomised double-blind controlled trial. Thorax. 2013;68(2):142–148. doi:10.1136/thoraxjnl-2012-202432
  • Cazzola M, Ora J, Cavalli F, Rogliani P, Matera MG. An overview of the safety and efficacy of monoclonal antibodies for the chronic obstructive pulmonary disease. Biologics. 2021;15:363–374. doi:10.2147/BTT.S295409
  • Rogliani P, Matera MG, Puxeddu E, et al. Emerging biological therapies for treating chronic obstructive pulmonary disease: a pairwise and network meta-analysis. Pulm Pharmacol Ther. 2018;50:28–37. doi:10.1016/j.pupt.2018.03.004
  • Maurice DH, Ke H, Ahmad F, Wang Y, Chung J, Manganiello VC. Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov. 2014;13(4):290–314. doi:10.1038/nrd4228
  • Jin SL, Goya S, Nakae S, et al. Phosphodiesterase 4B is essential for TH2-cell function and development of airway hyperresponsiveness in allergic asthma. J Allergy Clin Immunol. 2010;126(6):1252–9.e12. doi:10.1016/j.jaci.2010.08.014
  • Stasch JP, Pacher P, Evgenov OV. Soluble guanylate cyclase as an emerging therapeutic target in cardiopulmonary disease. Circulation. 2011;123(20):2263–2273. doi:10.1161/CIRCULATIONAHA.110.981738
  • Lam M, Bourke JE. A new pathway to airway relaxation: targeting the “other” cyclase in asthma. Am J Respir Cell Mol Biol. 2020;62(1):3–4. doi:10.1165/rcmb.2019-0274ED
  • Page CP. Phosphodiesterase inhibitors for the treatment of asthma and chronic obstructive pulmonary disease. Int Arch Allergy Immunol. 2014;165(3):152–164. doi:10.1159/000368800
  • Cazzola M, Calzetta L, Rogliani P, Matera MG. The discovery of roflumilast for the treatment of chronic obstructive pulmonary disease. Expert Opin Drug Discov. 2016;11(7):733–744. doi:10.1080/17460441.2016.1184642
  • Rogliani P, Calzetta L, Cazzola M, Matera MG. Drug safety evaluation of roflumilast for the treatment of COPD: a meta-analysis. Expert Opin Drug Saf. 2016;15(8):1133–1146. doi:10.1080/14740338.2016.1199683
  • Agustí A, Celli BR, Criner GJ, et al. Global Initiative for Chronic Obstructive Lung Disease 2023 report: GOLD executive summary. Am J Respir Crit Care Med. 2023;207(7):819–837. doi:10.1164/rccm.202301-0106PP
  • Matera MG, Rogliani P, Calzetta L, Cazzola M. Phosphodiesterase inhibitors for chronic obstructive pulmonary disease: what does the future hold? Drugs. 2014;74(17):1983–1992. doi:10.1007/s40265-014-0303-8
  • Matera MG, Ora J, Cavalli F, Rogliani P, Cazzola M. New avenues for phosphodiesterase inhibitors in asthma. J Exp Pharmacol. 2021;13:291–302. doi:10.2147/JEP.S242961
  • Phillips JE. Inhaled phosphodiesterase 4 (PDE4) inhibitors for inflammatory respiratory diseases. Front Pharmacol. 2020;11:259. doi:10.3389/fphar.2020.00259
  • Singh D, Emirova A, Francisco C, Santoro D, Govoni M, Nandeuil MA. Efficacy and safety of CHF6001, a novel inhaled PDE4 inhibitor in COPD: the PIONEER study. Respir Res. 2020;21(1):246. doi:10.1186/s12931-020-01512-y
  • Nguyen HO, Salvi V, Tiberio L, et al. The PDE4 inhibitor tanimilast shows distinct immunomodulatory properties associated with a type 2 endotype and CD141 upregulation. J Transl Med. 2022;20(1):203. doi:10.1186/s12967-022-03402-x
  • Li G, He D, Cai X, et al. Advances in the development of phosphodiesterase-4 inhibitors. Eur J Med Chem. 2023;250:115195. doi:10.1016/j.ejmech.2023.115195
  • Paes D, Hermans S, van den Hove D, Vanmierlo T, Prickaerts J, Carlier A. Computational investigation of the dynamic control of cAMP signaling by PDE4 isoform types. Biophys J. 2022;121(14):2693–2711. doi:10.1016/j.bpj.2022.06.019
  • Page C, Cazzola M. Bifunctional drugs for the treatment of respiratory diseases. Handb Exp Pharmacol. 2017;237:197–212. doi:10.1007/164_2016_69
  • Abbott-Banner KH, Page CP. Dual PDE3/4 and PDE4 inhibitors: novel treatments for COPD and other inflammatory airway diseases. Basic Clin Pharmacol Toxicol. 2014;114(5):365–376. doi:10.1111/bcpt.12209
  • Cazzola M, Calzetta L, Rogliani P, Matera MG. Ensifentrine (RPL554): an investigational PDE3/4 inhibitor for the treatment of COPD. Expert Opin Investig Drugs. 2019;28(10):827–833. doi:10.1080/13543784.2019.1661990
  • Barjaktarevic IZ, Rheault T, Bengtsson T, Rickard K. Ensifentrine, a novel dual phosphodiesterase (PDE) 3 and 4 inhibitor, significantly reduces annualized exacerbations and delays the time to first exacerbation in COPD: pooled sub-group analyses of ENHANCE-1 and ENHANCE-2 phase 3 trials. Am J Respir Crit Care Med. 2023;207:A5008. doi:10.1164/ajrccm-conference.2023.207.1_MeetingAbstracts.A5008
  • Sciurba F, Anzueto A, Rheault T, Bengtsson T, Rickard K. Ensifentrine, a novel dual phosphodiesterase (PDE) 3 and 4 inhibitor, improves lung function, symptoms, quality of life and reduces exacerbation rate and risk in patients with COPD: results from replicate phase 3 trials. Am J Respir Crit Care Med. 2023;207:A5005. doi:10.1164/ajrccm-conference.2023.207.1_MeetingAbstracts.A5005
  • Anzueto A, Rheault T, Bengtsson T, Rickard K. Treatment with ensifentrine, a dual PDE3 and PDE4 inhibitor, significantly reduced exacerbation rate and risk in subjects with COPD: sub-group results from the phase 3 trial, ENHANCE-2. Am J Respir Crit Care Med. 2023;207:A4998. doi:10.1164/ajrccm-conference.2023.207.1_MeetingAbstracts.A4998
  • Chapman RW, Phillips JE, Hipkin RW, Curran AK, Lundell D, Fine JS. CXCR2 antagonists for the treatment of pulmonary disease. Pharmacol Ther. 2009;121(1):55–68. doi:10.1016/j.pharmthera.2008.10.005
  • Thatcher TH, McHugh NA, Egan RW, et al. Role of CXCR2 in cigarette smoke-induced lung inflammation. Am J Physiol Lung Cell Mol Physiol. 2005;289(2):L322–L328. doi:10.1152/ajplung.00039.2005
  • Rennard SI, Dale DC, Donohue JF, et al. CXCR2 antagonist MK-7123. A phase 2 proof-of-concept trial for chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2015;191(9):1001–1011. doi:10.1164/rccm.201405-0992OC
  • Lazaar AL, Miller BE, Donald AC, et al. CXCR2 antagonist for patients with chronic obstructive pulmonary disease with chronic mucus hypersecretion: a phase 2b trial. Respir Res. 2020;21(1):149. doi:10.1186/s12931-020-01401-4
  • Keir HR, Richardson H, Fillmore C, et al. CXCL-8-dependent and -independent neutrophil activation in COPD: experiences from a pilot study of the CXCR2 antagonist danirixin. ERJ Open Res. 2020;6(4):00583–2020. doi:10.1183/23120541.00583-2020
  • Mattos MS, Ferrero MR, Kraemer L, et al. CXCR1 and CXCR2 inhibition by ladarixin improves neutrophil-dependent airway inflammation in mice. Front Immunol. 2020;11:566953. doi:10.3389/fimmu.2020.566953
  • Pelaia C, Vatrella A, Sciacqua A, Terracciano R, Pelaia G. Role of p38-mitogen-activated protein kinase in COPD: pathobiological implications and therapeutic perspectives. Expert Rev Respir Med. 2020;14(5):485–491. doi:10.1080/17476348.2020.1732821
  • Pascoe S, Costa M, Marks-Konczalik J, McKie E, Yang S, Scherbovsky PS. Biological effects of p38 MAPK inhibitor losmapimod does not translate to clinical benefits in COPD. Respir Med. 2017;130:20–26. doi:10.1016/j.rmed.2017.07.002
  • Higham A, Karur P, Jackson N, Cunoosamy DM, Jansson P, Singh D. Differential anti-inflammatory effects of budesonide and a p38 MAPK inhibitor AZD7624 on COPD pulmonary cells. Int J Chron Obstruct Pulmon Dis. 2018;13:1279–1288. doi:10.2147/COPD.S159936
  • Patel NR, Cunoosamy DM, Fagerås M, et al. The development of AZD7624 for prevention of exacerbations in COPD: a randomized controlled trial. Int J Chron Obstruct Pulmon Dis. 2018;13:1009–1019. doi:10.2147/COPD.S150576
  • Charron CE, Russell P, Ito K, et al. RV568, a narrow-spectrum kinase inhibitor with p38 MAPK-α and -γ selectivity, suppresses COPD inflammation. Eur Respir J. 2017;50(4):1700188. doi:10.1183/13993003.00188-2017
  • Strâmbu IR, Kobalava ZD, Magnusson BP, MacKinnon A, Parkin JM. Phase II study of single/repeated doses of acumapimod (BCT197) to treat acute exacerbations of COPD. COPD. 2019;16(5–6):344–353. doi:10.1080/15412555.2019.1682535
  • Wedzicha JA, MacKinnon A, Parkin JM. Effectiveness of acumapimod oral p38 inhibitor in the treatment of acute severe exacerbations of COPD: results of the AETHER phase II trial. Am J Respir Crit Care Med. 2018;197:A7710.
  • Yu H, Su X, Lei T, et al. Safety and efficacy of p38 mitogen-activated protein kinase inhibitors (MAPKIs) in COPD. Front Pharmacol. 2022;13:950035. doi:10.3389/fphar.2022.950035
  • Wang C, Zhou J, Wang J, et al. Progress in the mechanism and targeted drug therapy for COPD. Signal Transduct Target Ther. 2020;5(1):248. doi:10.1038/s41392-020-00345-x
  • Onions ST, Ito K, Charron CE, et al. Discovery of narrow spectrum kinase inhibitors: new therapeutic agents for the treatment of COPD and steroid-resistant asthma. J Med Chem. 2016;59(5):1727–1746. doi:10.1021/acs.jmedchem.5b01029
  • Curran A, Charron C, Russell P, Hava D. PUR1800 (RV1162), A novel narrow spectrum kinase inhibitor, but not fluticasone, reduces TNFa-induced cytokine release by primary bronchial epithelial cells from healthy volunteers and COPD patients. Eur Respir J. 2018;52:A5247. doi:10.1183/13993003.congress-2018.PA5247
  • Wasilewski M, Clayton R, Singh D, Perry J, Curran A. Safety and tolerability of PUR1800, an orally inhaled narrow spectrum kinase inhibitor, in patients with stable chronic obstructive pulmonary disease (COPD). J Allergy Clin Immunol. 2023;151(2 suppl):AB10. doi:10.1016/j.jaci.2022.12.062
  • Gupta V, Khan A, Higham A, et al. The effect of phosphatidylinositol-3 kinase inhibition on matrix metalloproteinase-9 and reactive oxygen species release from chronic obstructive pulmonary disease neutrophils. Int Immunopharmacol. 2016;35:155–162. doi:10.1016/j.intimp.2016.03.027
  • Sapey E, Stockley JA, Greenwood H, et al. Behavioral and structural differences in migrating peripheral neutrophils from patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2011;183(9):1176–1186. doi:10.1164/rccm.201008-1285OC
  • Stark AK, Chandra A, Chakraborty K, et al. PI3Kδ hyper-activation promotes development of B cells that exacerbate Streptococcus pneumoniae infection in an antibody-independent manner. Nat Commun. 2018;9(1):3174. doi:10.1038/s41467-018-05674-8
  • Cahn A, Hamblin JN, Robertson J, et al. An inhaled PI3Kδ inhibitor improves recovery in acutely exacerbating COPD patients: a randomized trial. Int J Chron Obstruct Pulmon Dis. 2021;16:1607–1619. doi:10.2147/COPD.S309129
  • Fahy WA, Homayoun-Valiani F, Cahn A, et al. Nemiralisib in patients with an acute exacerbation of COPD: placebo-controlled, dose-ranging study. Int J Chron Obstruct Pulmon Dis. 2021;16:1637–1646. doi:10.2147/COPD.S309320
  • Begg M, Hamblin JN, Jarvis E, et al. Exploring PI3Kδ molecular pathways in stable COPD and following an acute exacerbation, two randomized controlled trials. Int J Chron Obstruct Pulmon Dis. 2021;16:1621–1636. doi:10.2147/COPD.S309303
  • Sadiq MW, Asimus S, Belvisi MG, et al. Characterisation of pharmacokinetics, safety, and tolerability in a first-in-human study for AZD8154, a novel inhaled selective PI3Kγδ dual inhibitor targeting airway inflammatory disease. Br J Clin Pharmacol. 2022;88(1):260–270. doi:10.1111/bcp.14956
  • Di Stefano A, Maestrelli P, Roggeri A, et al. Upregulation of adhesion molecules in the bronchial mucosa of subjects with chronic obstructive bronchitis. Am J Respir Crit Care Med. 1994;149(3 Pt 1):803–810. doi:10.1164/ajrccm.149.3.7509705
  • Blidberg K, Palmberg L, James A, et al. Adhesion molecules in subjects with COPD and healthy non-smokers: a cross sectional parallel group study. Respir Res. 2013;14(1):47. doi:10.1186/1465-9921-14-47
  • Watz H, Bock D, Meyer M, et al. Inhaled pan-selectin antagonist Bimosiamose attenuates airway inflammation in COPD. Pulm Pharmacol Ther. 2013;26(2):265–270. doi:10.1016/j.pupt.2012.12.003
  • Tvaroška I, Selvaraj C, Koča J. Selectins - The two Dr. Jekyll and Mr. Hyde faces of adhesion molecules - A review. Molecules. 2020;25(12):2835. doi:10.3390/molecules25122835
  • Matera MG, Calzetta L, Cazzola M, Ora J, Rogliani P. Biologic therapies for chronic obstructive pulmonary disease. Expert Opin Biol Ther. 2023;23(2):163–173. doi:10.1080/14712598.2022.2160238
  • Higham A, Beech A, Wolosianka S, et al. Type 2 inflammation in eosinophilic chronic obstructive pulmonary disease. Allergy. 2021;76(6):1861–1864. doi:10.1111/all.14661
  • Eich A, Urban V, Jutel M, et al. A randomized, placebo-controlled phase 2 trial of CNTO 6785 in chronic obstructive pulmonary disease. COPD. 2017;14(5):476–483. doi:10.1080/15412555.2017.1335697
  • Murugaiah V, Tsolaki AG, Kishore U. Collectins: innate Immune pattern recognition molecules. Adv Exp Med Biol. 2020;1204:75–127. doi:10.1007/978-981-15-1580-4_4
  • Watson A, Madsen J, Clark HW. SP-A and SP-D: dual functioning immune molecules with antiviral and immunomodulatory properties. Front Immunol. 2021;11:622598. doi:10.3389/fimmu.2020.622598
  • Pilecki B, Wulf-Johansson H, Støttrup C, et al. Surfactant protein D deficiency aggravates cigarette smoke-induced lung inflammation by upregulation of ceramide synthesis. Front Immunol. 2018;9:3013. doi:10.3389/fimmu.2018.03013
  • Bafadhel M, Saha S, Siva R, et al. Sputum IL-5 concentration is associated with a sputum eosinophilia and attenuated by corticosteroid therapy in COPD. Respiration. 2009;78(3):256–262. doi:10.1159/000221902
  • Eltboli O, Mistry V, Barker B, Brightling CE. Relationship between blood and bronchial submucosal eosinophilia and reticular basement membrane thickening in chronic obstructive pulmonary disease. Respirology. 2015;20(4):667–670. doi:10.1111/resp.12475
  • Caramori G, Adcock IM, Di Stefano A, Chung KF. Cytokine inhibition in the treatment of COPD. Int J Chron Obstruct Pulmon Dis. 2014;9:397–412. doi:10.2147/COPD.S42544
  • Pavord ID, Chapman KR, Bafadhel M, et al. Mepolizumab for eosinophil-associated COPD: analysis of METREX and METREO. Int J Chron Obstruct Pulmon Dis. 2021;16:1755–1770. doi:10.2147/COPD.S294333
  • Criner GJ, Celli BR, Singh D, et al. Predicting response to benralizumab in chronic obstructive pulmonary disease: analyses of GALATHEA and TERRANOVA studies. Lancet Respir Med. 2020;8(2):158–170. doi:10.1016/S2213-2600(19)30338-8
  • Brightling CE, Bleecker ER, Panettieri RA, et al. Benralizumab for chronic obstructive pulmonary disease and sputum eosinophilia: a randomised, double-blind, placebo-controlled, phase 2a study. Lancet Respir Med. 2014;2(11):891–901. doi:10.1016/S2213-2600(14)70187-0
  • Mesnil C, Raulier S, Paulissen G, et al. Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest. 2016;126(9):3279–3295. doi:10.1172/JCI85664
  • Lange P, Ahmed E, Lahmar ZM, Martinez FJ, Bourdin A. Natural history, and mechanisms of COPD. Respirology. 2021;26(4):298–321. doi:10.1111/resp.14007
  • Sun J, Liu T, Yan Y, et al. The role of Th1/Th2 cytokines played in regulation of specific CD4+ Th1 cell conversion and activation during inflammatory reaction of chronic obstructive pulmonary disease. Scand J Immunol. 2018;88(1):e12674. doi:10.1111/sji.12674
  • Bhatt SP, Rabe KF, Hanania NA, et al. Dupilumab for COPD with type 2 inflammation indicated by eosinophil counts. N Engl J Med. 2023. doi:10.1056/NEJMoa2303951
  • Zhang K, Shan L, Rahman MS, Unruh H, Halayko AJ, Gounni AS. Constitutive and inducible thymic stromal lymphopoietin expression in human airway smooth muscle cells: role in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol. 2007;293(2):L375–L382. doi:10.1152/ajplung.00045.2007
  • Calvén J, Yudina Y, Hallgren O, et al. Viral stimuli trigger exaggerated thymic stromal lymphopoietin expression by chronic obstructive pulmonary disease epithelium: role of endosomal TLR3 and cytosolic RIG-I-like helicases. J Innate Immun. 2012;4(1):86–99. doi:10.1159/000329131
  • Elder MJ, Webster SJ, Williams DL, Gaston JS, Goodall JC. TSLP production by dendritic cells is modulated by IL-1β and components of the endoplasmic reticulum stress response. Eur J Immunol. 2016;46(2):455–463. doi:10.1002/eji.201545537
  • Cohen ES, Scott IC, Majithiya JB, et al. Oxidation of the alarmin IL-33 regulates ST2-dependent inflammation. Nat Commun. 2015;6(1):8327. doi:10.1038/ncomms9327
  • Cayrol C. IL-33, an Alarmin of the IL-1 Family involved in allergic and non allergic inflammation: focus on the mechanisms of regulation of its activity. Cells. 2021;11(1):107. doi:10.3390/cells11010107
  • Gabryelska A, Kuna P, Antczak A, Białasiewicz P, Panek M. IL-33 mediated inflammation in chronic respiratory diseases-understanding the role of the member of IL-1 superfamily. Front Immunol. 2019;10:692. doi:10.3389/fimmu.2019.00692
  • Tworek D, Majewski S, Szewczyk K, et al. The association between airway eosinophilic inflammation and IL-33 in stable non-atopic COPD. Respir Res. 2018;19(1):108. doi:10.1186/s12931-018-0807-y
  • Rabe KF, Celli BR, Wechsler ME, et al. Safety and efficacy of itepekimab in patients with moderate-to-severe COPD: a genetic association study and randomised, double-blind, phase 2a trial. Lancet Respir Med. 2021;9(11):1288–1298. doi:10.1016/S2213-2600(21)00167-3
  • Yousuf AJ, Mohammed S, Carr L, et al. Astegolimab, an anti-ST2, in chronic obstructive pulmonary disease (COPD-ST2OP): a phase 2a, placebo-controlled trial. Lancet Respir Med. 2022;10(5):469–477. doi:10.1016/S2213-2600(21)00556-7
  • Cazzola M, Rogliani P, Calzetta L, Matera MG. Tozorakimab. Anti-IL-33 monoclonal antibody, Treatment of COPD and asthma, Treatment of atopic dermatitis, Treatment of diabetic kidney disease. Drugs Future. 2023;48(2):101–109. doi:10.1358/dof.2023.48.2.3474010
  • Scott IC, England E, Rees DG, et al. Tozorakimab: a dual-pharmacology anti-IL-33 antibody that inhibits IL-33 signalling via ST2 and RAGE/EGFR to reduce inflammation and epithelial dysfunction. Eur Respir J. 2022;60:2467. doi:10.1183/13993003.congress-2022.246
  • Christopoulou ME, Papakonstantinou E, Stolz D. Matrix metalloproteinases in chronic obstructive pulmonary disease. Int J Mol Sci. 2023;24(4):3786. doi:10.3390/ijms24043786
  • Fingleton B. Matrix metalloproteinases as regulators of inflammatory processes. Biochim Biophys Acta Mol Cell Res. 2017;1864(11 Pt A):2036–2042. doi:10.1016/j.bbamcr.2017.05.010
  • Gharib SA, Manicone AM, Parks WC. Matrix metalloproteinases in emphysema. Matrix Biol. 2018;73:34–51. doi:10.1016/j.matbio.2018.01.018
  • Dahl R, Titlestad I, Lindqvist A, et al. Effects of an oral MMP-9 and −12 inhibitor, AZD1236, on biomarkers in moderate/severe COPD: a randomised controlled trial. Pulm Pharmacol Ther. 2012;25(2):169–177. doi:10.1016/j.pupt.2011.12.011
  • Baggio C, Velazquez JV, Fragai M, Nordgren TM, Pellecchia M. Therapeutic targeting of MMP-12 for the treatment of chronic obstructive pulmonary disease. J Med Chem. 2020;63(21):12911–12920. doi:10.1021/acs.jmedchem.0c01285
  • Li H, Ezra DG, Burton MJ, Bailly M. Doxycycline prevents matrix remodeling and contraction by trichiasis-derived conjunctival fibroblasts. Invest Ophthalmol Vis Sci. 2013;54(7):4675–4682. doi:10.1167/iovs.13-11787
  • Ham J, Kim J, Ko YG, Kim HY. The dynamic contribution of neutrophils in the chronic respiratory diseases. Allergy Asthma Immunol Res. 2022;14(4):361–378. doi:10.4168/aair.2022.14.4.361
  • Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD. Requirement for macrophage elastase for cigarette smoke-induced emphysema in mice. Science. 1997;277(5334):2002–2004. doi:10.1126/science.277.5334.2002
  • Kuna P, Jenkins M, O’Brien CD, Fahy WA. AZD9668, a neutrophil elastase inhibitor, plus ongoing budesonide/formoterol in patients with COPD. Respir Med. 2012;106(4):531–539. doi:10.1016/j.rmed.2011.10.020
  • Vogelmeier C, Aquino TO, O’Brien CD, Perrett J, Gunawardena KA. A randomised, placebo-controlled, dose-finding study of AZD9668, an oral inhibitor of neutrophil elastase, in patients with chronic obstructive pulmonary disease treated with tiotropium. COPD. 2012;9(2):111–120. doi:10.3109/15412555.2011.641803
  • Voynow JA, Zheng S, Kummarapurugu AB. Glycosaminoglycans as multifunctional anti-elastase and anti-inflammatory drugs in cystic fibrosis lung disease. Front Pharmacol. 2020;11:1011. doi:10.3389/fphar.2020.01011
  • Voynow JA, Shinbashi M. Neutrophil elastase and chronic lung disease. Biomolecules. 2021;11(8):1065. doi:10.3390/biom11081065
  • Shute JK, Calzetta L, Cardaci V, Di Toro S, Page CP, Cazzola M. Inhaled nebulised unfractionated heparin improves lung function in moderate to very severe COPD: a pilot study. Pulm Pharmacol Ther. 2018;48:88–96. doi:10.1016/j.pupt.2017.10.001
  • Margaroli C, Madison MC, Viera L, et al. An in vivo model for extracellular vesicle-induced emphysema. JCI Insight. 2022;7(4):e153560. doi:10.1172/jci.insight.153560
  • Crocetti L, Quinn MT, Schepetkin IA, Giovannoni MP. A patenting perspective on human neutrophil elastase (HNE) inhibitors (2014–2018) and their therapeutic applications. Expert Opin Ther Pat. 2019;29(7):555–578. doi:10.1080/13543776.2019.1630379
  • Palmér R, Mäenpää J, Jauhiainen A, et al. Dipeptidyl peptidase 1 inhibitor AZD7986 induces a sustained, exposure-dependent reduction in neutrophil elastase activity in healthy subjects. Clin Pharmacol Ther. 2018;104(6):1155–1164. doi:10.1002/cpt.1053
  • Miller BE, Mayer RJ, Goyal N, et al. Epithelial desquamation observed in a Phase I study of an oral cathepsin C inhibitor (GSK2793660). Br J Clin Pharmacol. 2017;83(12):2813–2820. doi:10.1111/bcp.13398
  • Chalmers JD, Haworth CS, Metersky ML, et al. Phase 2 trial of the DPP-1 inhibitor brensocatib in bronchiectasis. N Engl J Med. 2020;383(22):2127–2137. doi:10.1056/NEJMoa2021713
  • Long MB, Chalmers JD. Treating neutrophilic inflammation in airways diseases. Arch Bronconeumol. 2022;58(6):463–465. doi:10.1016/j.arbres.2021.11.003
  • Kim WD, Chi HS, Choe KH, Kim WS, Hogg JC, Sin DD. The role of granzyme B containing cells in the progression of chronic obstructive pulmonary disease. Tuberc Respir Dis. 2020;83(Supple 1):S25–S33. doi:10.4046/trd.2020.0089
  • Afonina IS, Tynan GA, Logue SE, et al. Granzyme B-dependent proteolysis acts as a switch to enhance the proinflammatory activity of IL-1α. Mol Cell. 2011;44(2):265–278. doi:10.1016/j.molcel.2011.07.037
  • Aslam MS, Yuan L. Serpina3n: potential drug and challenges, mini review. J Drug Target. 2020;28(4):368–378. doi:10.1080/1061186X.2019.1693576
  • Marcet-Palacios M, Ewen C, Pittman E, et al. Design and characterization of a novel human Granzyme B inhibitor. Protein Eng Des Sel. 2015;28(1):9–17. doi:10.1093/protein/gzu052
  • Shen Y, Zeglinski MR, Turner CT, et al. Topical small molecule granzyme B inhibitor improves remodeling in a murine model of impaired burn wound healing. Exp Mol Med. 2018;50(5):1–11. doi:10.1038/s12276-018-0095-0
  • Hirst CE, Buzza MS, Bird CH, et al. The intracellular granzyme B inhibitor, proteinase inhibitor 9, is up-regulated during accessory cell maturation and effector cell degranulation, and its overexpression enhances CTL potency. J Immunol. 2003;170(2):805–815. doi:10.4049/jimmunol.170.2.805
  • Du W, Mohammadpour H, O’Neill RE, et al. Serine protease inhibitor 6 protects alloreactive T cells from Granzyme B-mediated mitochondrial damage without affecting graft-versus-tumor effect. Oncoimmunology. 2017;7(3):e1397247. doi:10.1080/2162402X.2017.1397247
  • Ikram S, Ahmad J, Durdagi S. Screening of FDA approved drugs for finding potential inhibitors against Granzyme B as a potent drug-repurposing target. J Mol Graph Model. 2020;95:107462. doi:10.1016/j.jmgm.2019.107462
  • Cazzola M, Stolz D, Rogliani P, Matera MG. α1-Antitrypsin deficiency and chronic respiratory disorders. Eur Respir Rev. 2020;29(155):190073. doi:10.1183/16000617.0073-2019
  • Stolk J, Tov N, Chapman KR, et al. Efficacy and safety of inhaled α1-antitrypsin in patients with severe α1-antitrypsin deficiency and frequent exacerbations of COPD. Eur Respir J. 2019;54(5):1900673. doi:10.1183/13993003.00673-2019
  • Tumpara S, Martinez-Delgado B, Gomez-Mariano G, et al. The delivery of α1-antitrypsin therapy through transepidermal route: worthwhile to explore. Front Pharmacol. 2020;11:983. doi:10.3389/fphar.2020.00983
  • Remih K, Amzou S, Strnad P. Alpha1-antitrypsin deficiency: new therapies on the horizon. Curr Opin Pharmacol. 2021;59:149–156. doi:10.1016/j.coph.2021.06.001
  • Zou Y, Chen X, He B, et al. Neutrophil extracellular traps induced by cigarette smoke contribute to airway inflammation in mice. Exp Cell Res. 2020;389(1):111888. doi:10.1016/j.yexcr.2020.111888
  • Trivedi A, Khan MA, Bade G, Talwar A. Orchestration of neutrophil extracellular traps (NETs), a unique innate immune function during chronic obstructive pulmonary disease (COPD) development. Biomedicines. 2021;9(1):53. doi:10.3390/biomedicines9010053
  • Grabcanovic-Musija F, Obermayer A, Stoiber W, et al. Neutrophil extracellular trap (NET) formation characterises stable and exacerbated COPD and correlates with airflow limitation. Respir Res. 2015;16(1):59. doi:10.1186/s12931-015-0221-7
  • Keir HR, Chalmers JD. Neutrophil extracellular traps in chronic lung disease: implications for pathogenesis and therapy. Eur Respir Rev. 2022;31(163):210241. doi:10.1183/16000617.0241-2021
  • Tanner L, Single AB. Animal Models Reflecting chronic obstructive pulmonary disease and related respiratory disorders: translating pre-clinical data into clinical relevance. J Innate Immun. 2020;12(3):203–225. doi:10.1159/000502489
  • Segreti A, Stirpe E, Rogliani P, Cazzola M. Defining phenotypes in COPD: an aid to personalized healthcare. Mol Diagn Ther. 2014;18(4):381–388. doi:10.1007/s40291-014-0100-9
  • Xu X, Huang K, Dong F, et al. The heterogeneity of inflammatory response and emphysema in chronic obstructive pulmonary disease. Front Physiol. 2021;12:783396. doi:10.3389/fphys.2021.783396
  • Calzetta L, Aiello M, Frizzelli A, et al. Stem cell-based regenerative therapy and derived products in COPD: a systematic review and meta-analysis. Cells. 2022;11(11):1797. doi:10.3390/cells11111797
  • Martinez-Garcia MA, Sierra-Párraga JM, Quintana E, López-Campos JL. CFTR dysfunction and targeted therapies: a vision from non-cystic fibrosis bronchiectasis and COPD. J Cyst Fibros. 2022;21(5):741–744. doi:10.1016/j.jcf.2022.04.018