738
Views
6
CrossRef citations to date
0
Altmetric
Review

Pathological Mechanism and Targeted Drugs of COPD

, , , , &
Pages 1565-1575 | Published online: 12 Jul 2022

Abstract

Chronic obstructive pulmonary disease (COPD) includes chronic bronchitis, emphysema, and small airway obstruction. Incompletely reversible airflow limitation, inflammation, excessive mucus secretion and bronchial mucosal epithelial lesions are the main pathological basis of the disease. The prevalence of COPD is increasingly worldwide, which has caused the burden on individuals and society. This paper summarizes the pathogenesis of COPD and clarifies the effect and mechanism of the latest targeted drugs for COPD. Besides, we focus on NOD-like receptor thermal protein domain associated protein 3 inflammasome (NLRP3 inflammasome). NLRP3 can promote production of interleukin-1β (IL-1β) and interleukin-18 (IL-18). NLRP3 is an important factor in the migratory aggregation of macrophages and neutrophils and the generation of oxidative stress. Inhibition of NLRP3 inflammasome indirectly blocks the inflammatory effects of IL-1β and IL-18, which may be regarded as an ideal target for COPD treatment.

Introduction

COPD is an incurable chronic lung disease, which is also complicated by pulmonary heart disease and respiratory failure in some individuals with a tremendous burden on individuals and society. At present, it is not clear whether the pathological mechanisms of COPD are mainly thought to be the result of genetic and environmental interactions. Moreover, smoking is considered to be the main environmental factor to trigger COPD.Citation1Citation3 Except for genetics, gender,Citation4 occupation,Citation4,Citation5 airway hyperresponsiveness,Citation6 lung growth and development,Citation3,Citation7 and infectionCitation8 also play an important role in the development of COPD. It is easily understood that gender may influence the history of smoking and the particulate environment in which the occupation is located. However, the status of lung growth and development determines the susceptibility to COPD, which seems to have a close relationship with genetics.Citation9,Citation10 We should focus on airway hyperreactivity, which is an independent predictor of COPD and can exist independently without asthma and bronchitis, suggesting that the inflammatory response in COPD is different from asthma.Citation6,Citation11 In addition to the above causes, inflammatory mechanisms, oxidative stress, and protease-antiprotease imbalance are also involved in the development of COPD. Various reasons for bronchial mucosal epithelial cell degeneration, necrosis, squamous metaplasia and recurrent injury-repair airway wall eventually lead to the occurrence of structural repeated remodeling of the airways and scar formation.Citation12,Citation13

Currently, there is still no specific treatment for COPD, and palliative regimens to improve airflow limitation are the mainstay methods. We generally do not advocate drug intervention for COPD in the stable period, but in the acute attack stage, antibiotics, inhaled corticosteroids, bronchodilators, and other medicines are widely used in clinical practice. However, the negative effects of these drugs should not be ignored. For example, frequent use of inhaled corticosteroids can cause side effects such as osteoporosis, immunosuppression and increased probability of infection, especially infection that promotes the recurrence of COPD,Citation8,Citation14 while bronchodilators, for example, the anticholinergic agents and β2 agonists commonly used in clinical practice have side effects such as heart rate disturbance, impact on vision, urinary retention, and metabolic disorders, which cannot be ignored.Citation15,Citation16 With the development of molecular biology, targeted drugs for treatment by blocking COPD development are gradually being developed. This paper not only reviews the pathogenesis of COPD and the pharmacological mechanisms of COPD-related targeted drugs but also elaborates the concerned contribution of NLRP3 to COPD and the effectiveness of NLRP3 inhibitors and related advances.

Pathogenesis of COPD

Oxidative Stress Response

Oxidative stress is involved in the development of several inflammatory conditions and is an important pathogenetic factor in COPD. Stimulation of patients with smoke or dust leads to lung cell damage. Excessive mucus secretion and accumulation of neutrophils produce a large amount of reactive oxygen species (ROS). Oxidative inactivation of antiproteases loses inactivation and the structure of lung tissue is destroyed due to ROS.Citation17 The aggregation of neutrophils also leads to activation of a large number of inflammatory factors to produce more ROS, and aggravates the oxidative stress response.Citation18,Citation19 The oxidative system involves the secretion of the airway epithelial mucus, and the stimulation of noxious gases such as cigarettesCitation20 generates oxidative stress, which causes a large accumulation of ROS and regulates the relevant mucus genes, such as Muc5bCitation21 and Mu5ac.Citation22 In addition, epidermal growth factorCitation23 is also involved in the production of mucus. The signaling pathway of this kind of factor often resides in the oxidants’ activation of airway cells, and then involves in COPD. Regarding the structural destruction of lung tissue caused by protease-antiprotease imbalance, due to α1-antitrypsinCitation24 is the most active one, its inactivation is the most critical one. And the large amount of oxidants released by noxious gases, oxidative stress also makes the antiprotease inactivated and finally the protease-antiprotease imbalance occurs. Oxidative stress also enhances the inflammatory response in the lung by regulating redox-sensitive transcription factors such as nuclear factor kappa-B (NF-κB) and activator protein 1 (AP-1), releasing amounts of cell factors such as IL-1β, tumor necrosis factor-α (TNF-α). In addition,Citation25 the accumulation of ROS decreases the activity of histone deacetylases (HADC) and increases the activity of histone acetyltransferase, which can lead to a further aggregation of inflammatory cells, especially neutrophils.Citation26 So, we always observe a large number of neutrophil infiltrates during COPD pathological sections. If COPD worsens, the excessive oxidative stress will cause inflammatory cells to generate large amounts of ROS after accumulating. Then, a systemic response will occur.Citation27 Nuclear factor E2 (Nrf2)Citation28 can regulate antioxidant genes. In oxidative stress response, Nrf2 will dissociate. It is then transported to the nucleus to activate the transcription of antioxidant genes. Patients with COPD go through a diminished self-protective mechanism due to reduction of Nrf2 in level, resulting in lower endogenous antioxidant production.Citation29,Citation30

Inflammatory Cells, Inflammatory Mediators Cell Factors

Neutrophils and macrophages play a significant role in the oxidative stress response in COPD and are involved in the remodeling of COPD’s airway. NeutrophilsCitation7 accumulate in large numbers in the airways of COPD patients under oxidative stress, and this cell can secrete serine proteases,Citation31 including matrix metalloproteinase (MMP) and neutrophil elastase (NE). And MMP is significantly increased in patients with emphysema, and goes through the extracellular matrix of the lung destroyed by serine proteases, leading to the remodeling of the airway.Citation32 Neutrophils are sensitive to the infection response. When COPD patients are stimulated by infection, neutrophils will leave the circulation to aggregate in the lungs and protect the cells and surrounding tissues by phagocytosing the infectious agent to form proteases and bactericidal proteins, and produce ROS, which is the necessary mechanism to protect the body from free radical damage and the inductor of oxidative stress. In addition, the accuracy of neutrophil migratory aggregation is affected by physical fitness. Therefore, COPD is more common in the elderly population, which is associated with the expression of phosphatidylinositol 3-kinase (PI3K).Citation33 The accuracy of neutrophil migration can be improved by inhibiting type I PI3K-δ or PI3K-γ.Citation34

Activation of macrophagesCitation35 can regulate the beginning and the end of multiple inflammation. For COPD patients, the combination of Interferon-γ (IFN-γ) secreted by Th1, CD8+ cells, and B cells with the IFN-γ receptor will trigger a series of signaling cascades that lead to the activation and differentiation of M1 macrophages, which thenCitation36 produce a large number of cytokines such as TNF-α, IL-1β, interleukin-6 (IL-6) depending on the tissue site. M2 macrophages, on the other hand, are activated by a variety of cell factors (interleukin-4 (IL-4), interleukin-10 (IL-10) and interleukin-13 (IL-13) et al), and help the remodeling of airway by remodeling and repairing damaged tissues.Citation37 The inflammatory factor IL-6, produced by neutrophils and macrophages, can induce the production of elastase and oxygen radicals, which will increase the permeability of pulmonary vascular and aggravate the destruction of lung tissue.Citation38,Citation39 TNF-α, on the other hand, can modulate endothelial adhesion molecules, which will make polymorphonuclear leukocytes accumulate, and then release large amounts of elastase and ROS’s destroyed alveolar epithelium. And during the progression of COPD, TNF-α will generate an inflammatory cascadeCitation40 with IL-1β. In COPD patients, macrophages and neutrophils, entering the airways and upregulating chemokines such as monocyte chemotactic proteins (MCP-1, CCL-2),Citation41 and releasing large amounts of inflammatory factors, all indicate their contribution to the development of COPD.

Access Mechanism

NF-κB Access

NF-κB plays a role in systemic inflammation, such as rheumatoid arthritis and bronchial asthma. Activation of NF-kB is achieved by activating protein inhibitor kappa B (IkB)Citation42 to make ubiquitination of IkB. Because of IkB ubiquitination, NF-kB is released from the NF-kB/IkB complex, activates, exposes the nuclear localization domain, forms a p50/RelA dimer, and binds to target genes via the p50 subunit, thereby initiating the expression of target genes, such as TNF-α and IL-1, causing inflammatory responses.Citation43 In addition, the inhalation of mixtures, such as ozone, cigarette smoke and so on, leads to the migration of inflammatory cells such as neutrophils into the lungs, and generates ROS that is also a kind of factor in the activation of (NF-κB).Citation44 It will promote helper T cell type 1 (Th1)Citation45 to produce cell factors such as TNF-α, IL-1, IL-6, and so on which promote the maturation of resting monocytes into mature dendritic cells, which then provides autoantigens to self-reactive T lymphocytes, causing them to move to target tissues and the destruction of inflammation and lung tissue. All of these suggest that activation of the NF-κB pathway drives the release of inflammatory factors, leads to further enhancement of the oxidative stress response and exacerbates lung injury in patients. In respiratory tests in COPD patients, it is confirmed that the NF-κB expression is higher than in normal people. And in the case of smoking patients, the NF-κB expression is even higher. In addition, factors such as MCP-1,Citation46 IL-6, and CXCL-5Citation47 are released in large amounts during COPD exacerbations, and hypomethylation of NF-κB-mediated pathway gene DNA has also been observed to contribute to COPD exacerbation.Citation48

MAPK

Mitogen-activated protein kinase (MAPK)Citation49 is an important signaling pathway that transmits signals from the cell membrane to the nucleus. The pathway can be activated by the stimulation of cytokines, neurotransmitters, serine proteases, and oxidative stress to participate in stress adaptation and inflammatory responses.Citation50 In COPD, mitogen-activated protein kinase (p38MAPK)Citation51 plays an important role. The release of inflammatory factors caused by various environmental and genetic factors can lead to the activation of p38MAPK, while IL-8 and TNF-α, which are key factors associated with the development of COPD, are regulated by p38MAPK. The excessive release of these inflammatory factors eventually leads to aggregation of neutrophils, secretion of serine proteases, and destruction of lung structures. And all of p38MAPK isoforms (α, β, δ, γ)Citation52 in COPD patients occur with high expression and mediate lung inflammationCitation53 together. In addition, IL-8 and TNF-α, which are regulated by p38MAPK, appear to mediate glucocorticoid insensitivity in COPD patients. And these factors impair the function of glucocorticoid receptor (GR) by phosphorylating the GR, while the anti-inflammatory effects of glucocorticoids are exerted by GR. The endogenous p38MAPK antagonist MAPK-phosphatase-1 (MKP-1)Citation54 may be central to the reversal of glucocorticoid insensitivity in COPD patients. And it is found that glucocorticoid insensitivity could be reversed by blocking p38MAPK-α, γ and thus upregulating MKP-1.Citation55

PI3K/Akt

The PI3K/AktCitation56 pathway plays an important role in inhibiting cell proliferation and apoptosis. The activation of this pathway is mainly related to tyrosine kinase and G protein-coupled receptors.Citation57 Upon receiving the signal, the p85Citation58 regulatory subunit in PI3K aggregates to the plasma membrane site. The p110 and p85Citation59 subunit converts the substrate phosphatidylinositol 2 phosphate (PIP2) to phosphatidylinositol 3 phosphate (PIP3). And PIP3 binds to the N-terminal end of protein kinase B (Akt) and translocates to the cell membrane for activation.Citation60 It is an important mechanism of airway remodeling in COPDCitation33 that regulates the PI3K/Akt pathways and promote apoptosis through the tumor suppressor gene encoded by chromosome 10 negatively. In COPD patients, low expression of Nrf2 is closely associated with the oxidative stress response and the release of inflammatory factors and Nrf2 is a PI3K/Akt downstream signaling target.Citation61 In addition, when neutrophil migration is influenced by PI3K expression the persistent lung inflammation triggers neutrophil aggregation by activating the PI3K/Akt pathways. And the aggregation of associated cells generates large amounts of ROS to stimulate further exacerbation of oxidative stress. The activation of the PI3K/Akt pathways downregulates HADC activity, leading to the activation of inflammatory gene. Thus, inactivation of HADC is associated with glucocorticoid insensitivity.Citation62

Molecularly Targeted Drugs for COPD

As the pathogenesis of COPD has become better understood, research on molecularly targeted drugs has increased. Here, we will describe most (we only list those that have been registered in clinical trials and published results) of the targeted drugs used in COPD treatment and introduce the mechanisms of these drugs.

Antioxidants

Oxidative stress is an important mechanism in the development of COPD. The application of antioxidants is feasible in COPD patients, because the significantly elevated level of airway oxidative stress markers (eg, H2O2) and has been validated in in vivo experiments.Citation63 It is proved that N-acetylcysteine (NAC) can control COPD patients’ airway function in clinical trials.Citation64,Citation65 However, data from various studies are inconsistent, possibly due to oral administration affecting bioavailability. Other trials, such as superoxide dismutase, have a good anti-inflammatory effect achieved in animal studies, but still lack more clinical evidence.

Sulforaphane can attenuate oxidative stress by activating Nrf2 and regulating reactive nitrogen and ROS.Citation66 However, in a 4-week clinical trial,Citation67 no evidence was found to improve clinical outcomes in COPD (including Nrf2 expression, levels of relevant inflammatory markers). Therefore, the ability of turnip-sulfur as a therapeutic agent for COPD lacks the support of clinical evidence. The drugs are shown in .

Table 1 Antioxidants

Cytokine-Targeted Drugs

Numerous cytokines are involved in the development of COPD. High expression in COPD patients is also an important factor in activating various inflammatory signaling pathways, and oxidative stress responses. TNF-α plays an important role in interstitial lung disease. As a TNF-α targeting agent, Infliximab has been shown in animal studiesCitation68 to prevent smoke-induced emphysema in rats, and reduce the percentage of neutrophils and the level of IL-8 and TNF-α in rats. However, clinical trialsCitation69,Citation70 had less favorable outcomes and did not find an improvement in clinical outcomes in COPD patients. It has even been reported that it is possible to increase in the incidence of lung malignancies in COPD patients. Also, as an antagonist of TNF-α, the outcomes reported by etanerceptCitation71 were slightly better, but still not as effective as those of inhaled corticosteroids. Due to the lack of clinical evidence, whether TNF-α antagonists can be used as a treatment for COPD remains to be discussed.

IL-5 is in high expression in COPD patients. It seems feasible to inhibit IL-5 to achieve suppression of inflammatory and oxidative stress responses. Although mepolizumab is a target drug for IL-5, clinical evidence points to COPD with increased eosinophilia, probably because of the close association between IL-5 and the aggregation and differentiation of eosinophil.Citation72 Clinical trials have also demonstrated a reduction in COPD and exacerbation and a more pronounced reduction in eosinophil percentage than with placebo.Citation73 However, the FDA has not approved mepolizumab for COPD and trials related to it still have to be planned. Benralizumab, also an IL-5 antagonist, is approved for asthma with eosinophilia. However, clinical trials reported that benralizumab did not reduce the frequency of exacerbations in moderate-to-severe COPD.Citation74,Citation75 So, there is still a lack of more evidence to support it.

MK-7123 is a CXCR2 inhibitor, which can decrease neutrophil chemotaxis to reduce inflammatory manifestations in COPD patients. And in a clinical trial of 616 patients,Citation76 it was shown that MK-7123 at a 50 mg dose was effective in improving lung function and reducing lung inflammation in patients. Those drugs are shown in .

Table 2 Cytokine-Targeted Drugs

Enzyme Inhibitors

Protease-antiprotease imbalance is the main cause of lung damage in COPD patients. Protection against lung tissue damage and suppression of inflammation by inhibited NE seem feasible; however, clinical evidenceCitation77,Citation78 seems unsatisfactory, AZD9668, an NE inhibitor, failed to improve the lung function and the airway structure in patients. MMP inhibitors are one of the targets of anticancer drugs, but the clinical value of MMP inhibition for COPD remains unknown. MMP-9 and MMP-12 are significantly associated with airway inflammatory damage, both enzymes have enhanced activity in COPD patients, and their inhibition has been found to achieve better anti-inflammatory efficacy in animal model species of COPD,Citation79 however, no valuable outcomes have been reported in clinical trials.Citation80

Phosphodiesterases-3 and phosphodiesterases-4 (PED3 and PED4) are involved in the development of COPD, and they regulate cellular activity by hydrolyzing intracellular cAMP and cGMP.Citation81 PED3 is widely distributed in T lymphocytes, and lymphocyte function can be regulated by inhibition of PED3; inhibiting PED4 can reduce IL-4 and 5 gene expression in TH2 cells, decreases levels of inflammatory factors and has a synergistic effect with PED3 inhibitors in T cells.Citation82 Clinical evidenceCitation83,Citation84 also indicated that PED3/4 inhibitors (RPL-5 and roflumilast) both improve the lung function in COPD patients. However, when these patients simultaneously use standard bronchodilators, the function of RPL-554 and roflumilast benefits little. So, it did not justify the clinical efficacy in RPL-554 and roflumilast. Besides, roflumilast also reported side effects of severe gastrointestinal reactions (diarrhea and nausea)Citation85 and headache. Those drugs are shown in .

Table 3 Enzyme Inhibitors

Signaling Pathway Inhibitors

In COPD patients, activation of related pathways can enhance oxidative stress and cytokines, chemokines were massive release, which can further COPD development. Vitro trialsCitation86 demonstrated that verproside can achieve inflammation suppression by blocking the TNF-α/NF-κB pathway, but no clinical trials related to verproside have been seen, and no clinical evidence has been found for other inhibitors related to the NF-κB pathway.

p38MAPK inhibitors have recently received wide attention and have shown beneficial anti-inflammatory effects in smoke-induced pneumonia models,Citation87 in addition, inhibiting p38MAPK also reduces the production of associated cytokines by macrophages, whose aggregation is an important cell for oxidative stress and inflammatory factor release in COPD patientsCitation88 Currently, some clinical evidenceCitation89,Citation90 also confirmed that p38MAPK inhibitors (PH-797804, SB-681323) improve lung function and inflammatory factor levels (TNF-α) in COPD patients and perform well in hormone-insensitive classes of patients.

Inhibited PI3K can activate Nrf2, improve HDAC activity, modulate oxidative stress and improve inhaled corticosteroids resistance.Citation61,Citation62 In clinical trials,Citation91Citation94 the PI3K inhibitor GSK2269557 can improve lung function and related inflammatory factor levels (IL-8, IL-6) in COPD patients, but it has also been reported that it cannot change the clinical outcome. In addition, some studiesCitation95 pointed out that excessive PI3K inhibition may lead to immunosuppression. Nrf2, as a downstream target of PI3K, although there is no evidence of clinical effectiveness at this time. It remains to be validated and developed the medicine of downstream target of PI3K. Those drugs are shown in .

Table 4 Signaling Pathway Inhibitors

Relation of NLRP3 and Inflammatory Factors/Cells

Studies on NLRP3 in COPD, animal and in vitro experiments demonstrated that reducing NLRP3 expression can improve lung inflammation, inflammatory factor levels, and immune system function in COPD patients and so on, which also appears to improve glucocorticoid-insensitive classes of airway disease.

NLRP3 was demonstrated in mice experimentsCitation96 that Chlamydia and Haemophilus can increase NLR3, IL-1β responses and develop drug-resistant neutrophil inflammation. This experimental blockade of airway inflammation was made possible by NLRP3 inhibitor (MCC950), airway inflammation in asthma and the degree of glucocorticoid resistance were all related with IL-1β and NLRP3 expression. In in vitro models, NLRP3 expression is similarly elevated in models of COPD and exacerbated COPD, and IL-1β is similarly elevated.Citation97 In addition, experiments by YangCitation98 et al verified that in a tobacco-made COPD mouse model, knockdown of NLRP3 caused mice to lose evidence of lung inflammation and did not show pathological damage, while in NLRP3 knockdown mice, IL-1β, IL-18, macrophage, neutrophil, and lymphocyte levels were significantly lower than in COPD model mice. Although also affected by tobacco, NLRP3 knockdown significantly alleviated lung inflammation in mice, providing us with evidence that NLRP3 is a target for COPD treatment.

Relation of NLRP3 and Oxidative Stress

Research pointedCitation99 that NLRP3 deficiency can reduce oxidative stress and scavenge damaged mitochondria, and it seems to play an important role in neuroinflammation. After activation of NLRP3 inflammasome, the release of IL-1 β, IL-18 and other inflammatory factors will activate the polymorphonuclear neutrophils, produce a large number of ROS, and initiate the inflammatory response.Citation100 Furthermore, studiesCitation101 suggest that in smog-induced responses, activation of NLRP3 can induce apoptosis through the p53-Bax mitochondrial pathway. These results demonstrate the importance of NLRP3 inflammasome in the cell injury and apoptosis.

Conclusions

The pathogenesis of COPD is complex, mainly related to oxidative stress, inflammatory factors and over-expression or activation of signaling pathways. We found that these factors often co-exist, and it is difficult for us to achieve treatment of COPD through a single target among these interacting factors. Although some targeted drugs have achieved therapeutic efficacy, there are unknown consequences for the interaction of inflammatory factors and signaling pathways that inhibit only a single pathway. Therefore, most targeted drugs are still at a hypothetical stage in clinical practice. We need to reexamine the mechanisms of COPD and study each targeted pathway in depth to assess the safety of new targets through extensive in vitro and animal experiments. NLRP3 is highly correlated with the development of COPD and achieves therapeutic effects in COPD by controlling inflammation to inhibit the production of inflammatory factors by blocking the activation of related pathways. It is also effective in hormonic tolerant patients. Inhibited NLRP3 has not been reported any adverse effects in animal studies. Therefore, we believe that the targeted drugs have implications for continued development in the treatment of COPD.

Disclosure

The authors report no conflicts of interest in this work.

References

  • Rao W, Wang S, Duleba M, et al. Regenerative metaplastic clones in COPD lung drive inflammation and fibrosis. Cell. 2020;181(4):848–864. doi:10.1016/j.cell.2020.03.047
  • 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
  • Barnes PJ, Burney PG, Silverman EK, et al. Chronic obstructive pulmonary disease. Nat Rev Dis Primers. 2015;1:15076. doi:10.1038/nrdp.2015.76
  • Raherison C, Girodet PO. Epidemiology of COPD. Eur Respir Rev. 2009;18(114):213–221. doi:10.1183/09059180.00003609
  • Salvi SS, Barnes PJ. Chronic obstructive pulmonary disease in non-smokers. Lancet. 2009;374(9691):733–743. doi:10.1016/S0140-6736(09)61303-9
  • Kume H, Hojo M, Hashimoto N. Eosinophil inflammation and hyperresponsiveness in the airways as phenotypes of COPD, and usefulness of inhaled glucocorticosteroids. Front Pharmacol. 2019;10:765. doi:10.3389/fphar.2019.00765
  • Benjamin JT, Plosa EJ, Sucre JM, et al. Neutrophilic inflammation during lung development disrupts elastin assembly and predisposes adult mice to COPD. J Clin Invest. 2021;131(1):e139481. doi:10.1172/JCI139481
  • Linden D, Guo-Parke H, Coyle PV, et al. Respiratory viral infection: a potential “missing link” in the pathogenesis of COPD. Eur Respir Rev. 2019;28(151):180063. doi:10.1183/16000617.0063-2018
  • Huang X, Mu X, Deng L, et al. The etiologic origins for chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2019;14:1139–1158. doi:10.2147/COPD.S203215
  • Nasri A, Foisset F, Ahmed E, et al. Roles of mesenchymal cells in the lung: from lung development to chronic obstructive pulmonary disease. Cells. 2021;10(12):3467. doi:10.3390/cells10123467
  • Kim HT, Yin W, Nakamichi Y, et al. WNT/RYK signaling restricts goblet cell differentiation during lung development and repair. Proc Natl Acad Sci U S A. 2019;116(51):25697–25706. doi:10.1073/pnas.1911071116
  • Berg K, Wright JL. The pathology of chronic obstructive pulmonary disease: progress in the 20th and 21st centuries. Arch Pathol Lab Med. 2016;140(12):1423–1428. doi:10.5858/arpa.2015-0455-RS
  • Bagdonas E, Raudoniute J, Bruzauskaite I, Aldonyte R. Novel aspects of pathogenesis and regeneration mechanisms in COPD. Int J Chron Obstruct Pulmon Dis. 2015;10:995–1013. doi:10.2147/COPD.S82518
  • Agusti A, Fabbri LM, Singh D, et al. Inhaled corticosteroids in COPD: friend or foe? Eur Respir J. 2018;52(6):1801219. doi:10.1183/13993003.01219-2018
  • Gupta P, O’Mahony MS. Potential adverse effects of bronchodilators in the treatment of airways obstruction in older people: recommendations for prescribing. Drugs Aging. 2008;25(5):415–443. doi:10.2165/00002512-200825050-00005
  • Tanabe N, Sato S, Muro S, et al. Regional lung deflation with increased airway volume underlies the functional response to bronchodilators in chronic obstructive pulmonary disease. Physiol Rep. 2019;7(24):e14330. doi:10.14814/phy2.14330
  • Chang RM, Kauffman RJ, Kwon Y. Understanding the paradigm shift to computational social science in the presence of big data. Decis Support Syst. 2014;63:67–80. doi:10.1016/j.dss.2013.08.008
  • Dang X, He B, Ning Q, et al. Alantolactone suppresses inflammation, apoptosis and oxidative stress in cigarette smoke-induced human bronchial epithelial cells through activation of Nrf2/HO-1 and inhibition of the NF-κB pathways. Respir Res. 2020;21(1):95. doi:10.1186/s12931-020-01358-4
  • Zhang MY, Jiang YX, Yang YC, et al. Cigarette smoke extract induces pyroptosis in human bronchial epithelial cells through the ROS/NLRP3/caspase-1 pathway. Life Sci. 2021;269:119090. doi:10.1016/j.lfs.2021.119090
  • Shih YM, Chang YJ, Cooke MS, et al. Alkylating and oxidative stresses in smoking and non-smoking patients with COPD: implications for lung carcinogenesis. Free Radic Biol Med. 2021;164:99–106. doi:10.1016/j.freeradbiomed.2020.12.442
  • Roy MG, Livraghi-Butrico A, Fletcher AA, et al. Muc5b is required for airway defence. Nature. 2014;505(7483):412–416. doi:10.1038/nature12807
  • Samsuzzaman M, Uddin MS, Shah MA, Mathew B. Natural inhibitors on airway mucin: molecular insight into the therapeutic potential targeting MUC5AC expression and production. Life Sci. 2019;231:116485. doi:10.1016/j.lfs.2019.05.041
  • Wu YF, Li ZY, Dong LL, et al. Inactivation of MTOR promotes autophagy-mediated epithelial injury in particulate matter-induced airway inflammation. Autophagy. 2020;16(3):435–450. doi:10.1080/15548627.2019.1628536
  • Geramizadeh B, Jowkar Z, Karami L, Masoumpour M, Mehrabi S, Ghayoumi MA. Alpha-1 antitrypsin deficiency in Iranian patients with chronic obstructive pulmonary disease. Iran Red Crescent Med J. 2013;15(11):e7508. doi:10.5812/ircmj.7508
  • Rajendrasozhan S, Yao H, Rahman I. Current perspectives on role of chromatin modifications and deacetylases in lung inflammation in COPD. COPD. 2009;6(4):291-297..
  • Sahakian E, Chen J, Powers JJ, et al. Essential role for histone deacetylase 11 (HDAC11) in neutrophil biology. J Leukoc Biol. 2017;102(2):475–486. doi:10.1189/jlb.1A0415-176RRR
  • Zuo L, Wijegunawardana D. Redox role of ROS and inflammation in pulmonary diseases. Adv Exp Med Biol. 2021;1304:187–204.
  • Cui W, Zhang Z, Zhang P, et al. Nrf2 attenuates inflammatory response in COPD/emphysema: crosstalk with Wnt3a/β-catenin and AMPK pathways. J Cell Mol Med. 2018;22(7):3514–3525. doi:10.1111/jcmm.13628
  • Mizumura K, Maruoka S, Shimizu T, Gon Y. Role of Nrf2 in the pathogenesis of respiratory diseases. Respir Investig. 2020;58(1):28–35. doi:10.1016/j.resinv.2019.10.003
  • Fratta PA, Stranieri C, Ferrari M, et al. Oxidative stress and Nrf2 expression in peripheral blood mononuclear cells derived from COPD patients: an observational longitudinal study. Respir Res. 2020;21(1):37. doi:10.1186/s12931-020-1292-7
  • Genschmer KR, Russell DW, Lal C, et al. Activated PMN exosomes: pathogenic entities causing matrix destruction and disease in the lung. Cell. 2019;176(1–2):113–126. doi:10.1016/j.cell.2018.12.002
  • Mahor D, Kumari V, Vashisht K, et al. Elevated serum matrix metalloprotease (MMP-2) as a candidate biomarker for stable COPD. BMC Pulm Med. 2020;20(1):302. doi:10.1186/s12890-020-01323-3
  • Lu J, Xie L, Liu C, Zhang Q, Sun S. PTEN/PI3k/AKT regulates macrophage polarization in emphysematous mice. Scand J Immunol. 2017;85(6):395–405. doi:10.1111/sji.12545
  • Sun X, Chen L, He Z. PI3K/Akt-Nrf2 and anti-inflammation effect of macrolides in chronic obstructive pulmonary disease. Curr Drug Metab. 2019;20(4):301–304. doi:10.2174/1389200220666190227224748
  • Li N, Liu Y, Cai J. LncRNA MIR155HG regulates M1/M2 macrophage polarization in chronic obstructive pulmonary disease. Biomed Pharmacother. 2019;117:109015. doi:10.1016/j.biopha.2019.109015
  • Huang H, Feng H, Zhuge D. M1 macrophage activated by notch signal pathway contributed to ventilator-induced lung injury in chronic obstructive pulmonary disease model. J Surg Res. 2019;244:358–367. doi:10.1016/j.jss.2019.06.060
  • Takiguchi H, Yang CX, Yang C, et al. Macrophages with reduced expressions of classical M1 and M2 surface markers in human bronchoalveolar lavage fluid exhibit pro-inflammatory gene signatures. Sci Rep. 2021;11(1):8282. doi:10.1038/s41598-021-87720-y
  • El-Gazzar AG, Kamel MH, Elbahnasy O, El-Naggar ME. Prognostic value of platelet and neutrophil to lymphocyte ratio in COPD patients. Expert Rev Respir Med. 2020;14(1):111–116. doi:10.1080/17476348.2019.1675517
  • Shyam PSB, Chaya SK, Kumar VS, et al. Inflammatory biomarkers interleukin 1 beta (IL-1β) and tumour necrosis factor alpha (TNF-α) are differentially elevated in tobacco smoke associated COPD and biomass smoke associated COPD. Toxics. 2021;9(4):72. doi:10.3390/toxics9040072
  • Wang Z, Locantore N, Haldar K, et al. Inflammatory endotype-associated airway microbiome in chronic obstructive pulmonary disease clinical stability and exacerbations: a multicohort longitudinal analysis. Am J Respir Crit Care Med. 2021;203(12):1488–1502. doi:10.1164/rccm.202009-3448OC
  • Di Stefano A, Coccini T, Roda E, et al. Blood MCP-1 levels are increased in chronic obstructive pulmonary disease patients with prevalent emphysema. Int J Chron Obstruct Pulmon Dis. 2018;13:1691–1700. doi:10.2147/COPD.S159915
  • Hoffmann A, Levchenko A, Scott ML, Baltimore D. The IkappaB-NF-kappaB signaling module: temporal control and selective gene activation. Science. 2002;298(5596):1241–1245. doi:10.1126/science.1071914
  • Lee UJ, Choung SR, Prakash KV, et al. Dual knockdown of p65 and p50 subunits of NF-kappaB by siRNA inhibits the induction of inflammatory cytokines and significantly enhance apoptosis in human primary synoviocytes treated with tumor necrosis factor-alpha. Mol Biol Rep. 2008;35(3):291–298. doi:10.1007/s11033-007-9084-4
  • Zhuan B, Yu Y, Yang Z, Li P, Li P. Mechanisms of oxidative stress effects of the NADPH oxidase-ROS-NF-κB transduction pathway and VPO1 on patients with chronic obstructive pulmonary disease combined with pulmonary hypertension. Eur Rev Med Pharmacol Sci. 2017;21(15):3459–3464.
  • Mojiri-Forushani H, Hemmati AA, Khodadadi A, et al. Valsartan attenuates bleomycin-induced pulmonary fibrosis by inhibition of NF-κB expression and regulation of Th1/Th2 cytokines. Immunopharmacol Immunotoxicol. 2018;40(3):225–231. doi:10.1080/08923973.2018.1431924
  • Balamayooran G, Batra S, Cai S, et al. Role of CXCL5 in leukocyte recruitment to the lungs during secondhand smoke exposure. Am J Respir Cell Mol Biol. 2012;47(1):104–111. doi:10.1165/rcmb.2011-0260OC
  • Starrett W, Blake DJ. Sulforaphane inhibits de novo synthesis of IL-8 and MCP-1 in human epithelial cells generated by cigarette smoke extract. J Immunotoxicol. 2011;8(2):150–158. doi:10.3109/1547691X.2011.558529
  • Kaur G, Batra S. Regulation of DNA methylation signatures on NF-κB and STAT3 pathway genes and TET activity in cigarette smoke extract-challenged cells/COPD exacerbation model in vitro. Cell Biol Toxicol. 2020;36(5):459–480. doi:10.1007/s10565-020-09522-8
  • Drosten M, Barbacid M. Targeting the MAPK pathway in KRAS-driven tumors. Cancer Cell. 2020;37(4):543–550. doi:10.1016/j.ccell.2020.03.013
  • Yang G, Chang CC, Yang Y, et al. Resveratrol alleviates rheumatoid arthritis via reducing ROS and inflammation, inhibiting MAPK signaling pathways, and suppressing angiogenesis. J Agric Food Chem. 2018;66(49):12953–12960. doi:10.1021/acs.jafc.8b05047
  • Knobloch J, Jungck D, Kronsbein J, Stoelben E, Ito K, Koch A. LABAs and p38MAPK inhibitors reverse the corticosteroid-insensitivity of IL-8 in airway smooth muscle cells of COPD. J Clin Med. 2019;8(12):2058. doi:10.3390/jcm8122058
  • Banerjee A, Koziol-White C, Panettieri RJ. p38 MAPK inhibitors, IKK2 inhibitors, and TNFα inhibitors in COPD. Curr Opin Pharmacol. 2012;12(3):287–292. doi:10.1016/j.coph.2012.01.016
  • 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
  • Keränen T, Moilanen E, Korhonen R. Suppression of cytokine production by glucocorticoids is mediated by MKP-1 in human lung epithelial cells. Inflamm Res. 2017;66(5):441–449. doi:10.1007/s00011-017-1028-4
  • Pinart M, Hussain F, Shirali S, et al. Role of mitogen-activated protein kinase phosphatase-1 in corticosteroid insensitivity of chronic oxidant lung injury. Eur J Pharmacol. 2014;744:108–114. doi:10.1016/j.ejphar.2014.10.003
  • Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K pathway in human disease. Cell. 2017;170(4):605–635. doi:10.1016/j.cell.2017.07.029
  • Desale SE, Chidambaram H, Chinnathambi S. G-protein coupled receptor, PI3K and Rho signaling pathways regulate the cascades of Tau and amyloid-βin Alzheimer’s disease. Mol Biomed. 2021;2(1):17. doi:10.1186/s43556-021-00036-1
  • De la Cruz-Herrera CF, Baz-Martínez M, Lang V, et al. Conjugation of SUMO to p85 leads to a novel mechanism of PI3K regulation. Oncogene. 2016;35(22):2873–2880. doi:10.1038/onc.2015.356
  • Wang Q, Zhang P, Zhang W, et al. PI3K activation is enhanced by FOXM1D binding to p110 and p85 subunits. Signal Transduct Target Ther. 2020;5(1):105. doi:10.1038/s41392-020-00218-3
  • An X, Wei Z, Ran B, et al. Histone deacetylase inhibitor trichostatin A suppresses cell proliferation and induces apoptosis by regulating the PI3K/AKT signalling pathway in gastric cancer cells. Anticancer Agents Med Chem. 2020;20(17):2114–2124. doi:10.2174/1871520620666200627204857
  • Mercado N, Thimmulappa R, Thomas CM, et al. Decreased histone deacetylase 2 impairs Nrf2 activation by oxidative stress. Biochem Biophys Res Commun. 2011;406(2):292–298. doi:10.1016/j.bbrc.2011.02.035
  • Malhotra D, Thimmulappa RK, Mercado N, et al. Denitrosylation of HDAC2 by targeting Nrf2 restores glucocorticosteroid sensitivity in macrophages from COPD patients. J Clin Invest. 2011;121(11):4289–4302. doi:10.1172/JCI45144
  • Aydemir Y, Aydemir Ö, Şengül A, et al. Comparison of oxidant/antioxidant balance in COPD and non-COPD smokers. Heart Lung. 2019;48(6):566–569. doi:10.1016/j.hrtlng.2019.07.005
  • Tse HN, Raiteri L, Wong KY, et al. High-dose N-acetylcysteine in stable COPD: the 1-year, double-blind, randomized, placebo-controlled HIACE study. Chest. 2013;144(1):106–118. doi:10.1378/chest.12-2357
  • Johnson K, Mcevoy CE, Naqvi S, et al. High-dose oral N-acetylcysteine fails to improve respiratory health status in patients with chronic obstructive pulmonary disease and chronic bronchitis: a randomized, placebo-controlled trial. Int J Chron Obstruct Pulmon Dis. 2016;11:799–807. doi:10.2147/COPD.S102375
  • Li D, Shao R, Wang N, et al. Sulforaphane activates a lysosome-dependent transcriptional program to mitigate oxidative stress. Autophagy. 2021;17(4):872–887. doi:10.1080/15548627.2020.1739442
  • Wise RA, Holbrook JT, Criner G, et al. Lack of effect of oral sulforaphane administration on Nrf2 expression in COPD: a randomized, double-blind, placebo controlled trial. PLoS One. 2016;11(11):e163716. doi:10.1371/journal.pone.0163716
  • Zhang XY, Zhang C, Sun QY, et al. Infliximab protects against pulmonary emphysema in smoking rats. Chin Med J. 2011;124(16):2502–2506.
  • Rennard SI, Flavin SK, Agarwal PK, Lo KH, Barnathan ES. Long-term safety study of infliximab in moderate-to-severe chronic obstructive pulmonary disease. Respir Med. 2013;107(3):424–432. doi:10.1016/j.rmed.2012.11.008
  • 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
  • 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
  • Nagase H, Ueki S, Fujieda S. The roles of IL-5 and anti-IL-5 treatment in eosinophilic diseases: asthma, eosinophilic granulomatosis with polyangiitis, and eosinophilic chronic rhinosinusitis. Allergol Int. 2020;69(2):178–186. doi:10.1016/j.alit.2020.02.002
  • Pavord ID, Chanez P, Criner GJ, et al. Mepolizumab for eosinophilic chronic obstructive pulmonary disease. N Engl J Med. 2017;377(17):1613–1629. doi:10.1056/NEJMoa1708208
  • 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
  • Criner GJ, Celli BR, Brightling CE, et al. Benralizumab for the prevention of COPD exacerbations. N Engl J Med. 2019;381(11):1023–1034. doi:10.1056/NEJMoa1905248
  • 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
  • Kuna P, Jenkins M, O’Brien CD, Ahy 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
  • Nordenmark LH, Taylor R, Jorup C. Feasibility of computed tomography in a multicenter COPD trial: a study of the effect of AZD9668 on structural airway changes. Adv Ther. 2015;32(6):548–566. doi:10.1007/s12325-015-0215-3
  • Navratilova Z, Kolek V, Petrek M. Matrix metalloproteinases and their inhibitors in chronic obstructive pulmonary disease. Arch Immunol Ther Exp. 2016;64(3):177–193. doi:10.1007/s00005-015-0375-5
  • 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
  • Ntontsi P, Detta A, Bakakos P, Loukides S, Hillas G. Experimental and investigational phosphodiesterase inhibitors in development for asthma. Expert Opin Investig Drugs. 2019;28(3):261–266. doi:10.1080/13543784.2019.1571582
  • Martin C, Burgel PR, Roche N. Inhaled dual phosphodiesterase 3/4 inhibitors for the treatment of patients with COPD: a short review. Int J Chron Obstruct Pulmon Dis. 2021;16:2363–2373. doi:10.2147/COPD.S226688
  • Singh D, Abbott-Banner K, Bengtsson T, Newman K. The short-term bronchodilator effects of the dual phosphodiesterase 3 and 4 inhibitor RPL554 in COPD. Eur Respir J. 2018;52(5):1801074. doi:10.1183/13993003.01074-2018
  • Martinez FJ, Calverley PM, Goehring UM, Brose M, Fabbri LM, Rabe KF. Effect of roflumilast on exacerbations in patients with severe chronic obstructive pulmonary disease uncontrolled by combination therapy (REACT): a multicentre randomised controlled trial. Lancet. 2015;385(9971):857–866. doi:10.1016/S0140-6736(14)62410-7
  • Wedzicha JA, Calverley PM, Rabe KF. Roflumilast: a review of its use in the treatment of COPD. Int J Chron Obstruct Pulmon Dis. 2016;11:81–90. doi:10.2147/COPD.S89849
  • Lee SU, Sung MH, Ryu HW, et al. Verproside inhibits TNF-α-induced MUC5AC expression through suppression of the TNF-α/NF-κB pathway in human airway epithelial cells. Cytokine. 2016;77:168–175. doi:10.1016/j.cyto.2015.08.262
  • Li T, Wu YN, Wang H, Ma JY, Zhai SS, Duan J. Dapk1 improves inflammation, oxidative stress and autophagy in LPS-induced acute lung injury via p38MAPK/NF-κB signaling pathway. Mol Immunol. 2020;120:13–22. doi:10.1016/j.molimm.2020.01.014
  • Arora S, Dev K, Agarwal B, Das P, Syed MA. Macrophages: their role, activation and polarization in pulmonary diseases. Immunobiology. 2018;223(4–5):383–396. doi:10.1016/j.imbio.2017.11.001
  • Macnee W, Allan RJ, Jones I, Das P, Syed MA. Efficacy and safety of the oral p38 inhibitor PH-797804 in chronic obstructive pulmonary disease: a randomised clinical trial. Thorax. 2013;68(8):738–745. doi:10.1136/thoraxjnl-2012-202744
  • Singh D, Smyth L, Borrill Z, Sweeney L, Tal-Singer R. A randomized, placebo-controlled study of the effects of the p38 MAPK inhibitor SB-681323 on blood biomarkers of inflammation in COPD patients. J Clin Pharmacol. 2010;50(1):94–100. doi:10.1177/0091270009347873
  • Cahn A, Hamblin JN, Begg M, et al. Safety, pharmacokinetics and dose-response characteristics of GSK2269557, an inhaled PI3Kδ inhibitor under development for the treatment of COPD. Pulm Pharmacol Ther. 2017;46:69–77. doi:10.1016/j.pupt.2017.08.008
  • 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
  • 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
  • Rohrbacher L, Brauchle B, Ogrinc WA, von Bergwelt-Baildon M, Bücklein VL, Subklewe M. The PI3K∂-selective inhibitor idelalisib induces T- and NK-cell dysfunction independently of B-cell malignancy-associated immunosuppression. Front Immunol. 2021;12:608625. doi:10.3389/fimmu.2021.608625
  • Kim RY, Pinkerton JW, Essilfie AT, et al. Role for NLRP3 inflammasome-mediated, IL-1β-dependent responses in severe, steroid-resistant asthma. Am J Respir Crit Care Med. 2017;196(3):283–297. doi:10.1164/rccm.201609-1830OC
  • Nachmias N, Langier S, Brzezinski RY, et al. NLRP3 inflammasome activity is upregulated in an in-vitro model of COPD exacerbation. PLoS One. 2019;14(5):e214622. doi:10.1371/journal.pone.0214622
  • Yang W, Ni H, Wang H, Gu H. NLRP3 inflammasome is essential for the development of chronic obstructive pulmonary disease. Int J Clin Exp Pathol. 2015;8(10):13209–13216.
  • Wu X, Gong L, Xie L, et al. NLRP3 deficiency protects against intermittent hypoxia-induced neuroinflammation and mitochondrial ROS by promoting the PINK1-parkin pathway of mitophagy in a murine model of sleep apnea. Front Immunol. 2021;12:628168. doi:10.3389/fimmu.2021.628168
  • Yang H, Lv H, Li H, et al. Oridonin protects LPS-induced acute lung injury by modulating Nrf2-mediated oxidative stress and Nrf2-independent NLRP3 and NF-κB pathways. Cell Commun Signal. 2019;17(1):62. doi:10.1186/s12964-019-0366-y
  • Li C, Zhihong H, Wenlong L, et al. The nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 inflammasome regulates bronchial epithelial cell injury and proapoptosis after exposure to biomass fuel smoke. Am J Respir Cell Mol Biol. 2016;55(6):815–824. doi:10.1165/rcmb.2016-0051OC