Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Nov 14.
Published in final edited form as: Expert Opin Ther Pat. 2019 Jun 16;29(7):555–578. doi: 10.1080/13543776.2019.1630379

A patenting perspective on human neutrophil elastase (HNE) inhibitors (2014–2018) and their therapeutic applications

L Crocetti a, MT Quinn b, IA Schepetkin b, MP Giovannoni a
PMCID: PMC9642779  NIHMSID: NIHMS1847152  PMID: 31204543

Abstract

Introduction:

Human neutrophil elastase (HNE) is involved in a variety of serious chronic diseases, especially cardiopulmonary pathologies. For this reason, the regulation of HNE activity represents a promising therapeutic approach, which is evident by the development of a number of new and selective HNE inhibitors, both in the academic and pharmaceutical environments.

Areas covered:

The present review analyzes and summarizes the patent literature regarding human neutrophil elastase inhibitors for the treatment of cardiopulmonary diseases over 2014–2018.

Expert opinion:

HNE is an interesting and defined target to treat various inflammatory diseases, including a number of cardiopulmonary pathologies. The research in this field is quite active, and a number of HNE inhibitors are currently in various stages of clinical development. In addition, new opportunities for HNE inhibitor development stem from recent studies demonstrating the involvement of HNE in many other inflammatory pathologies, including rheumatoid arthritis, inflammatory bowel disease, skin diseases, and cancer. Furthermore, the development of dual HNE/proteinase 3 inhibitors is being pursued as an innovative approach for the treatment of neutrophilic inflammatory diseases. Thus, these new developments will likely stimulate new and increased interest in this important therapeutic target and for the development of novel and selective HNE inhibitors.

Keywords: Human neutrophil elastase, elastase inhibitors, cardiopulmonary diseases, inflammation, protease-antiproteases imbalance

1. Introduction

Proteases are enzymes that catalyze the hydrolysis of peptide bonds, thus converting proteins into smaller fragments. They are involved in many physiological processes and are considered important signaling molecules [1,2]. Serine proteases represent one-third of all known proteolytic enzymes, and they are grouped into 13 clans (superfamilies) and 40 families [3]. The family name ‘serine’ arises from the amino acid serine located in the active site, which is responsible for the nucleophilic attack on the carbonyl moiety of the substrate peptide bond to form an acyl-enzyme intermediate [4]. The four main clans of serine proteases include the superfamily A or PA clan (e.g. chymotrypsin), the subtilisin clan (SB), clan SC (e.g. carboxypeptidase C), and clan SF (e.g. bacterial leader peptidase I) [2,5]. Significant differences exist in the distribution of each clan across species. For example, proteases of superfamily A are highly represented in eukaryotes and less in prokaryotes and plant genomes, while clans SB and SC are most represented in other organisms. Among serine proteases belonging to the chymotrypsin family are the neutrophil serine proteases (NSP) [6], including neutrophil elastase (NE), cathepsin G (CG), proteinase 3 (PR3), and the recently discovered neutrophil serine protease 4 (NSP4) [7]. In addition, human neutrophils also express azurocidin (AZU), an inactive serine protease homolog with chemotactic and antimicrobial activities [8,9].

Neutrophils are the first line of defense against infection and are efficient phagocytes able to engulf and degrade microorganisms through the combined action of reactive oxygen species (ROS), proteases, and antimicrobial peptides [10,11]. Neutrophils are polymorphonuclear cells containing four types of granules: azurophil granules (also known as primary), specific granules (also known secondary), gelatinase granules (also known as tertiary), and secretory granules [12]. The granules are classified according to their protein content and their different ability to undergo exocytosis after neutrophil activation by inflammatory stimuli or phagocytosis of invading microorganisms. Azurophil granules are mainly involved in the intracellular degradation of microorganisms in the phagolysosome due to their high content of myeloperoxidase (MPO), bactericidal permeability-increasing protein, defensins, and serine proteases, such as NE, CG and PR3 [13]. The extracellular release of serine proteases from azurophil granules plays an important role in modulation of the inflammatory response by proteolytically modifying chemokines and cytokines, activating latent forms of cytokines and growth factors, and cleaving specific cell surface receptors. In addition, neutrophil serine proteases can induce caspase-independent cell apoptosis and initiate adaptive immune responses by activating lymphocytes [14]. Clearly, NSP play an important role in the inflammatory process and immune response and, thus, represent a potential target for therapeutic intervention. In particular, we will discuss human neutrophil elastase (HNE) inhibitors and their therapeutic applications in this review.

2. HNE

HNE (EC 3.4.21.37) is a globular glycoprotein of about 30 kDa belonging to the chymotrypsin family. It consists of a single polypeptide chain of 218 amino acids and two asparagine-linked carbohydrate side chains at Asn95 and Asn144. It is stabilized by four disulfide bridges and exhibits basic properties due to the presence of 19 arginine residues, resulting in an isoelectric point around 10–11. The active HNE structure is composed of an N-terminal activation domain and three flexible loops [15]. The primary structure has homology with PR3 (57%) and CG (37%), since they evolved from a common gene by duplication [16]. ELA2, the gene for HNE, is located in the terminal region of the short arm of chromosome 19 at p13.3 and consists of five exons and four introns. This region also contains the genes for the homologous proteins PR3 and azurocidin [17]. Mutations in ELA2 can be a risk factor for severe congenital and cyclic neutropenia, a rare disease characterized by oscillations in the production of neutrophils and other blood cells [18,19]. HNE is found not only in neutrophils but also in mast cells, monocytes, eosinophils, keratinocytes, and fibroblasts [20,21]. The concentration of HNE in neutrophils exceeds 5 mM, and the total amount in a single cell has been estimated to be up to 3 pg [22], which is sequestered in the cell by compartmentalization in azurophil granules. Active HNE is rapidly released from the granules into the extracellular space, although some released HNE remains associated with the outer plasma membrane [23,24]. HNE performs its proteolytic action through a catalytic triad consisting of Ser195-Asp102-His57, where the powerful nucleophile Ser195 OH group attacks the carbonyl carbon involved in the peptide bond. Close by the catalytic triad, an oxyanion binding site comprised of the backbone amide NH of Ser195 and Gly193 is present (Figure 1) [25]. Hydrolysis of the peptide bond occurs via two tetrahedral intermediates [26]. The OH group of Ser195, activated by His 57 as a general base, attacks the carbonyl group of the substrate leading to the first tetrahedral intermediate. This intermediate is then stabilized by the backbone N atoms of Gly193 and Ser195, which together generate a positively charged pocket within the active site, known as the oxyanion hole. Hydrogen bond interactions in the oxyanion hole contribute 1.5–3.0 kcal/mol to ground and transition state stabilization [27]. His57 transferring a proton to the amine of the tetrahedral intermediate causes this bond to break of this last one, resulting in the formation of the covalent acyl-enzyme complex. A molecule of water attacks the acyl-enzyme complex to generate a new tetrahedral intermediate, stabilized by the oxyanion hole as well. Finally, the collapse of this second intermediate results in the regeneration of active HNE [4,25].

Figure 1.

Figure 1.

HNE catalytic triad.

3. Physiological functions of HNE

The term elastase is not quite correct, since HNE does not only degrade elastin, a protein responsible for the elasticity of lung tissue. Rather, HNE degrades a variety of extracellular matrix and plasma proteins, including collagen (types I-IV), fibronectin, laminin, and proteoglycans [2831]. Other HNE substrates are coagulation factors (fibrinogen and factors V, VII, XII, and XIII), plasminogen, immunoglobulin G (IgG), IgA and IgM, thrombomodulin, platelet, complement factors C3 and C5, complement receptors [3234], and gp120, the coat protein of the human immunodeficiency virus [35]. HNE can also degrade other proteases present inside neutrophil granules [36], as well as some protease inhibitors, leading to their activation or inactivation [37]. HNE is currently considered a multifunctional enzyme involved in the killing of pathogens, regulation of inflammation, and tissue homeostasis [13] via its proteolytic activity against the proteins listed above. Moreover, HNE appears to play an important role in chemotaxis and migration of neutrophils by the degradation of adhesion molecules at cell junctions [38,39]. Although neutrophils may normally migrate to sites of infection in the absence of elastase, the presence of HNE is required for maximal intracellular killing of Gram-negative bacteria [40,41]. In addition, neutrophils can form neutrophil extracellular traps (NETs), which are web-like chromatin structures extruded from neutrophils and can bind/trap bacteria. These NETs sequester high concentrations of released serine proteases, including elastase, where they can degrade virulence factors and kill bacteria extracellularly [42].

Since a single azurophilic granule activated from neutrophils releases millimolar concentrations of HNE, the potent proteolytic activity of this serine protease must be adequately regulated by endogenous inhibitors. However, why HNE can still degrade extracellular matrix, despite the presence of functioning anti-protease molecules, is still not fully understood. According to the literature, neutrophils could release oxidant molecules that locally turn off anti-proteases, thereby permitting the action of HNE focused on local tissues [43]. Campbell and co-workers [22] measured the function of HNE in regards to quantum proteolysis and found that ~ 67,000 molecules of HNE are stored in each azurophilic granule, reaching a concentration of about 5.33 mM, which exceeds pericellular inhibitor concentrations in vivo by nearly three orders of magnitude. Consequently, other non-oxidative mechanisms must limit the proteolytic activity of HNE together with the endogenous inhibitors [43]. Azurophilic granules contain acidic medium that prevents proteolytic activity. Thus, storage in specialized compartments is important to control inadvertent protease activity inside the cell. Elastase is activated in mild alkaline medium, which results from the fusion of the granules with phagocytic vacuoles to form phagolysosomes where the engulfed bacteria are destroyed [44] or extracellular release.

Upon neutrophil activation, the intracellular granules fuse with the plasma membrane, which dramatically enlarges the neutrophil cell surface area and triggers a neutrophil oxidative burst. At this point, the most of the active enzyme is released in the extracellular space (~10% of the released HNE remains bound to the cell membrane), and it can be neutralized by the endogenous inhibitors in the blood. However, the concentration of elastase at the site of liberation is still higher than that of the extracellular anti-proteases. More distant from the site of degranulation, the elastase concentration decreases due to diffusion/dilution until free elastase is completely neutralized [45]. Consequently, only a small percentage of elastase liberated upon neutrophil activation is involved in a healthy situation [22,46].

The principal class of endogenous proteins which work as HNE inhibitors are serpins (serine protease inhibitors) [47,48], tissue inhibitors of metalloproteases (TIMPS) [49], and cystatins (cysteine protease inhibitors) [50]. The activity of HNE is primarily regulated by serpins, α1-antitrypsin (α1-AT, ATT or α1-proteinase inhibitor, α1-PI) [51], monocyte/NE inhibitor (MNEI, also called serpin B1) [52], and secretory leukocytes proteinase inhibitor (SLPI) [53] and elafin [54], which belongs to the chelonianin family. α1-AT, a blood plasma glycoprotein, regulates the activity of HNE in the low respiratory tract, while SLPI protects the upper airways from proteolysis [55]. Serpin B1, a potent inhibitor of HNE, is expressed at high concentrations mainly in macrophages and neutrophil. Elafin, a small soluble protein of 6 kDa, is a potent HNE inhibitor present in bronchial secretions [52,54]. In addition to regulation of the inflammatory process by direct enzyme inhibition, there is increasing evidence that the serine proteases inhibitors α1-AT, SLPI, and elafin also affect leukocyte chemotaxis and pro-inflammatory mediator release and may contribute to defense against invading pathogens [56].

4. Involvement of HNE in pathologic conditions

As discussed above, the potent proteolytic activity of HNE is strictly controlled by several endogenous inhibitors, which are particularly abundant in the respiratory tract and circulatory system and protect tissues from damage [57,58]. According to the protease/antiprotease imbalance hypothesis, many pathologies of the respiratory system result from damage to the connective tissue that is caused by the massive migration and infiltration of neutrophils and the subsequent release of proteolytic enzymes, including serine proteases (Figure 2). Hence, inadequate control of the protease’s activity due to low levels of their physiological inhibitors ultimately leads to the degradation of elastin, the elastic component of lung connective tissue, and other components of the extracellular matrix [56,59].

Figure 2.

Figure 2.

Protease-antiprotease imbalance in pulmonary diseases where HNE plays a key role.

Out-of-balance elastase activity is at the basis of severe diseases, especially those associated with the cardiopulmonary system [60], such as chronic obstructive pulmonary disease (COPD) [6164], bronchiectasis (BE) [65,66], cystic fibrosis (CF) [43,67], acute lung injury (ALI) [68], acute respiratory distress syndrome (ARDS) [69], pulmonary arterial hypertension (PAH) [70], and idiopathic pulmonary fibrosis (IPF) [71]. HNE significantly contributes to chronic inflammatory airway diseases by inducing mucin production in airway epithelial cells [72]. The exact mechanism at the basis of this effect is unclear, but it was demonstrated to involve protein kinase C and production of ROS. ROS, in turn, activate tumor necrosis factor (TNF)-converting enzyme, leading to TNF-converting enzyme-dependent release of soluble transforming growth factor-α (TGFα) and TGFα stimulation of epidermal growth factor receptor (EGFR), which induces the production of mucin [73].

HNE is present in psoriatic lesions and induces keratinocyte hyperproliferation via the epidermal growth factor receptor signaling pathway [74]. The low concentration of specific elastase inhibitors in psoriatic skin increases the excessive hydrolytic activity of HNE released from migrating cells [75]. Recently, Marto et al. [76] published a promising strategy for the treatment of psoriasis and inflammatory skin disease using a coumarin-based oxo-β-lactam compound to inhibit HNE. This HNE inhibitor, ER-143, was successfully formulated in starch-based nanocapsules with adequate pharmaceutical characteristics (such as particle size distribution, surface charge, and encapsulation efficiency), and it represents a promising approach for topical application to treat inflammatory skin diseases [77,78].

HNE is also involved in rheumatoid arthritis, since it can directly degrade the matrix, destroying cartilage components [79]. Di Cesare Mannelli et al. [80] described the effects of a potent and selective HNE inhibitor, EL-17 (IC50 = 20 nM) [81], in the rat model of rheumatoid arthritis. In this adjuvant-induced arthritis model, a single administration of EL-17 reduced pain evoked by noxious stimuli, and repeated treatment with EL-17 significantly prevented the development of spontaneous pain and hypersensitivity induced by mechanical noxious and non-noxious stimuli. The preventive efficacy seems to be related to a disease-modifying effect, since the articular damage clearly present in the histological examination of the tibiotarsal joint was greatly reduced, at least by the higher dose of EL-17.

HNE also seems to be involved in the development and progression of cancer, which is consistent with the observation that many types of cancer originate in areas of the body where chronic inflammatory states occur [82], and neutrophils and their proteases participate actively, contributing to the progression of the tumor itself [83]. A certain analogy can be found between tumor cells and neutrophils: cancer cells enter the bloodstream, adhere to endothelial cells to pass through them and invade tissues in different organs, and give rise to metastases. Likewise, neutrophils transported by the bloodstream throughout the body adhere to adhesive molecules present on activated endothelial cells to then penetrate through the vessel wall and migrate to the areas of inflammation. The biggest difference between these cells is the lifetime: neutrophils undergo apoptosis after ~72 h from their synthesis in the bone marrow, while the cancer cells do not die [84]. The tumor cells themselves are also able to release proteases, whose proteolytic activity is very important during tissue invasion and metastasis formation, as it allows them to overcome a series of physiological barriers consisting of elastin, collagen, and proteoglycan [85]. In vitro immunoassay studies demonstrate that these proteases, and in particular NE, are directly produced by tumor cells of the breast and lung [86]. In breast cancer, high levels of HNE represent a negative prognosis indicator, as demonstrated by the decrease in survival both in the absence and in the presence of metastasis [87]. In lung cancer, elevated serological levels of HNE are correlated not only with the stage of illness but also with its progression. In fact, the activity of the enzyme is three or five times greater in broncho-alveolar lavage fluid of patients with lung cancer compared to healthy individuals [88]. Furthermore, HNE actively participates in the degradation of elastin fibers, thus promoting the penetration and spread of cancer cells, but it can also generate elastin fragments, called elastochines, with properties similar to cytokines. These fragments act at various levels, promoting the progression of lung cancer [89]. Thus, the inhibition of HNE may be beneficial both in preventing invasion and in metastasis of various types of cancer [84]. In cancer, the production of myeloid cells in the bone marrow is generally accelerated, and the number of circulating neutrophils and myeloid-derived suppressor cells (MDSCs) appears to be correlated with the progression of the disease and patient survival. A large number of MDSCs often correlates with negative clinical outcomes [90,91]. Recently, it has been shown that infiltrating myeloid cells exert pro-tumorigenic action through HNE activity in prostate cancer [92,93]. A recent study demonstrated that the protease-antiprotease imbalance is also related to the pathogenesis of type 1 diabetes [94]. In these patients, α1-AT levels decrease, whereas circulating HNE concentrations and HNE enzymatic activity significantly increase. These changes correlate with diabetes-associated auto-antibody levels [15].

Vicuña et al. [95,96] showed that inhibitors of HNE can reverse ongoing neuropathic pain in mice, demonstrating a new role for this enzyme in the modulation of this type of pain. They also showed that NE contributing to neuropathic pain can be expressed not only by neutrophils but also by T-lymphocytes. In particular, the authors demonstrated that the administration of Sivelestat (see Figure 3) in mice after spinal nerve injury reduced mechanical pain behaviors during the first two weeks after injury as a result of its inhibitory action on serine proteases. Furthermore, treatment with Sivelestat left baseline nociceptive sensitivity unaffected. The evidence that the block of HNE activity by Sivelestat led to positive effects on mechanical allodynia in a model of neuropathic pain suggests a functional involvement of this protease in the complex mechanism of neuropathic pain [97,98]. Lastly, in a recent publication, HNE has been related to chronic kidney disease (CKD) [99], a pathology with an inflammatory basis. Chronic inflammation in patients with CKD under hemodialysis has been associated with an imbalance between oxidant and anti-oxidant mechanisms, including neutrophil activation with release of ROS and granule contents, such as HNE [100,101]. Finally, other important neutrophil-driven inflammatory diseases such as inflammatory bowel disease, Crohn’s disease [102], atherosclerosis [103], severe pneumonia [104], and graft-versus-host disease [105] are related to excessive HNE activity. Based on the discussion above, it is clear that inflammation is a physiological and pathological process that is found in many diseases and that serine proteases, including HNE, are key elements in these processes. Hence, the control of this proteolytic activity represents an important therapeutic aspect for the resolution of these diseases.

Figure 3.

Figure 3.

Sivelestat (marketed as Elaspol® 100), Alvelestat and BAY85-8501 (in Phase II clinical trials).

5. HNE inhibitors

In the last three decades, many pharmaceutical companies and academia have discovered a variety of innovative elastase inhibitors. At present, only two HNE inhibitors are on the market. The first one, Prolastin® (purified α1-AT), is a peptide drug used in the treatment of α1-AT deficiency [106]. The second is Sivelestat (ONO-5046, Elaspol® 100), which is a non-peptide inhibitor approved for intravenous use in Japan and South Korea for the treatment of ALI and ARDS associated with systemic inflammatory response syndrome [107]. Sivelestat is a highly specific and effective HNE inhibitor with an IC50 = 44 nM and Ki = 200 nM [108] (Figure 3).

Low-molecular-weight synthetic HNE inhibitors may offer several advantages over peptide inhibitors, including enhanced enzyme selectivity, oral bioavailability, lower susceptibility to proteolytic inactivation, and decreased risk of an immunogenic response. Moreover, a ‘small molecule’ is more easily modifiable in order to allow for the greater ability to modify the inhibitor structure and optimize physical and pharmacokinetic properties for the production of viable clinical candidates [109]. In literature, there are also many HNE inhibitors from natural products reported [110113], although they generally have low potency, selectivity, and metabolic stability, and none of these compounds have become drug candidates [63]. An important exception to this trend is represented by Lyngbyastatin 7 isolated from Cyanobacteria of marine environment by Salvador et. al in 2013 [114] and later obtained for total synthesis via 31 steps by Luo and co-workers [115]. It shows a 19-membered cyclic hexadepsi-peptide core and is a potent HNE inhibitor (IC50 = 23 nM) provided with a good selectivity for HNE against a panel of 68 proteases.

Patents on HNE inhibitors released in the period 2010–2014 were summarized in an excellent review by Tsai and Hwang (2015). In the current review, we report a perspective on low-molecular-weight HNE inhibitors and their possible therapeutic application by considering the patents registered from 2014 to 2018. Since in this range of time some company have published several licenses on HNE inhibitors, we decided to present the patents and the HNE inhibitors reported therein according to the parent pharmaceutical company.

Before providing a systematic description of the most interesting ‘small molecules’ discovered by the pharmaceutical industry in the selected period, we first report a short presentation of drugs in clinical trials. Table 1 shows the HNE inhibitors currently in clinical trials, and we included the target pathology, the clinical trial phase, the company/institute involved in the clinical trial, and the drug generation. For the latter, we followed the recent classification by Von Nussbaum et al. [45], which categorizes HNE inhibitors into five generations. The peptide inhibitors α1-AT [116123] and Elafin [124,125] are in clinical trials for different pathologies, including COPD, α1-AT deficiency (ADAT) or PAH. All the others are synthetic non-peptide inhibitors, with the exception of POL6014 [126,127], a new macrocyclic peptide mimetic developed by the University of Zürich and Polyphor Ltd using Protein Epitope Mimetics (PEM) drug discovery [128130]. The structures of Alvelestat [45,131133] and BAY-858501 [45,134,135] are reported in Figure 3, while the formula of CHF6333 from Chiesi Farmaceutici is undisclosed [66,136,137]. These latter drugs will be discussed in detail in the corresponding sections.

Table 1.

HNE inhibitors in clinical trials.

HNE Inhibitor Condition/Disease Phase Clinical Trials Generation Company/Institute Reference
Alpha 1-Antitrypsin (ATT) (Glassia, Aralast) COPD Alpha1-antitrypsin Deficiency (ADAT) III 1 Shire (Baxalta now part of Shire) [114,115]
Alpha 1-Antitrypsin (ATT) (Prolastin, Aralast) Acute Myocardial Infarction II 1 Virginia Commonwealth University [116,117]
Alpha 1 Anti-Trypsin (AAT) (Zemaria) Steroid Refractory Acute Graft vs Host Disease II 1 University of Michigan Cancer Center [118,119]
Alpha 1 Anti-Trypsin (AAT) (Glassia) Graft-Versus-Host Disease (GVHD) Acute on Chronic II 1 Fred Hutchinson Cancer Research Center [120,121]
ELAFIN (Tiprelestat) Pulmonary Arterial Hypertension (PAH) I 1 Roham T. Zamanian, Stanford University [122,123]
POL6014 Cystic Fibrosis II 1/2 Santhera Pharmaceuticals [124128]
MPH 966 (Alvelestat oral tablet) ADAT Enphysema COPD II 3 Mereo BioPharma [45,129,130]
AZD9668 (Alvelestat) Bronchiolitis Obliterans Syndrome (BOS) Chronic Graft vs Host Disease II 3 National Cancer Institute (NCI) [45,131]
CHF6333 Cystic Fibrosis Bronchiectasis I 5 Chiesi Farmaceutici S.p.A. [66,134,135]
BAY 85-8501 Bronchiectasis II 5 Bayer [45,132,133]

5.1. Chiesi Farmaceutici S.p.A

Chiesi Farmaceutici S.p.A has been working for years in the development of bronchodilator drugs for the treatment of pulmonary diseases, such as COPD, and is also interested in the identification of new HNE inhibitors to use both alone and in association with other drugs. Before considering the patents, it is important to highlight that the structure of the best compound developed by this company (CHF6333) is not known. CHF6333, currently in phase I clinical trials, is the first inhaled HNE inhibitor under development for the treatment of HNE-driven lung diseases using a dry powder inhaler. In vitro, CHF6333 is a highly potent HNE inhibitor (IC50 = 0.2 nM) with good selectivity against other proteases. In addition, CHF6333 significantly reduces lung neutrophil recruitment induced by cigarette smoke exposure in mice and reduces both lung tissue infection and inflammation when administered intratracheally to P. aeruginosa-infected rats for 7 days [138].

In the literature, there are many 2,3,5,8-tetrahydro-[1,2,4] triazolo[4,3-a]pyrimidine derivatives reported as HNE inhibitors patented by Chiesi [139143] and active by an inhalatory route. The reported IC50 values indicate compounds endowed with a notable potency, but no clear structure–activity relationships were deduced from the compound library. In Table 2, a general structure of tetrahydrotriazolopirimidine and some significant compounds (1–8) are presented. Compounds 7 and 8 were also tested in at least two separate experiments against activated rat and human neutrophils, with IC50 of <10 nM (rat protease inhibition) and <1 nM (human protease inhibition), respectively [142]. Among the bicyclic compounds patented by Chiesi, the [1,2,4]triazolo[1,5-a]pyridine derivatives 9–11 shown in Figure 4 are active in the nanomolar range [144]. Moreover, there are three patents claiming monocyclic heteroaromatic compounds with imidazolone and pyrazolone scaffolds [145147]. These compounds exhibit HNE inhibitory activity in the subnanomolar range and are suitable for administration in microparticles using a dry powder inhaler (DPI). Table 3 shows some representative compounds with imidazolone (12–17) and pyrazolone (18,19) scaffolds, all containing a m-trifluoromethylphenyl ring at N-1 of the heterocyclic system and a p-cyanophenyl group [145,146]. Noteworthy, the two pairs of isomers with imidazolone and pyrazolone scaffolds (16 versus 18 and 17 versus 19) retain the same level of inhibitory activity.

Table 2.

Examples of 2,3,5,8-tetrahydro[1,2,4]triazolo[4,3-a]pyrimidine compounds (5th generation) developed by Chiesi Farmaceutici S.p.A.

graphic file with name nihms-1847152-t0001.jpg
Compound R R1 IC50 (nM)a Reference
1 CH3 graphic file with name nihms-1847152-t0002.jpg < 20 [137]
2 CH3 graphic file with name nihms-1847152-t0003.jpg < 20 [138]
3 CH3 graphic file with name nihms-1847152-t0004.jpg < 20 [138]
4 CH3 graphic file with name nihms-1847152-t0005.jpg < 20 [139]
5 CH3 graphic file with name nihms-1847152-t0006.jpg < 20 [139]
6 CH3 graphic file with name nihms-1847152-t0007.jpg < 20 [139]
7 CH2CH2OH H < 1 [140]
8 graphic file with name nihms-1847152-t0008.jpg H < 1 [140]
a

The results are means of two independent experiments, each performed in duplicate (HNE enzyme assay).

Figure 4.

Figure 4.

Examples of [1,2,4]triazolo[1,5-a]pyridine derivatives developed by Chiesi Farmaceutici S.p.A.

Table 3.

Examples of representative imidazolone (12–17) and pyrazolone derivatives (18, 19) developed by Chiesi Farmaceutici S.p.A.

graphic file with name nihms-1847152-t0009.jpg
Compound R1 R2 IC50 (nM)a Reference
12 graphic file with name nihms-1847152-t0010.jpg H < 1 [143]
13 graphic file with name nihms-1847152-t0011.jpg H < 1 [143]
14 graphic file with name nihms-1847152-t0012.jpg H < 1 [143]
15 graphic file with name nihms-1847152-t0013.jpg H < 1 [143]
16 graphic file with name nihms-1847152-t0014.jpg graphic file with name nihms-1847152-t0015.jpg < 1 [144]
17 graphic file with name nihms-1847152-t0016.jpg graphic file with name nihms-1847152-t0017.jpg < 1 [144]
18 graphic file with name nihms-1847152-t0018.jpg graphic file with name nihms-1847152-t0019.jpg < 1 [144]
19 graphic file with name nihms-1847152-t0020.jpg graphic file with name nihms-1847152-t0021.jpg < 1 [144]
a

The results are means of two independent experiments, each performed in duplicate (HNE enzyme assay).

5.2. Boehringer Ingelheim GmbH

A number of 4-oxo-1,4-dihydropyridine were patented by Boehringer Ingelheim International GmbH as HNE inhibitors for the treatment and/or prevention of pulmonary, gastrointestinal (GI) and genitourinary diseases, inflammatory diseases of the skin and eye, other autoimmune and allergic disorders, allograft rejection, and oncological diseases [148150]. In Table 4 the main representative compounds 20–29 are shown. Their HNE inhibitory activity is in the sub/low nanomolar range. The presence of the sulfur atom as chiral center in compounds 20–22 did not influence HNE inhibitory activity, and the reported IC50 values are related to experiments performed on the racemate. In contrast, the chiral center in the substituent at nitrogen in position 1 [the best is the 1-(4-cyanophenyl)ethyl group] had an important effect on activity. For these compounds, it is possible to identify the (R)-enantiomer as the eutomer whose IC50 values are reported in Table 4. The comparison of IC50 values of (R)- and (S)-enantiomers clearly shows that the eutomer (R) is always at least four fold more potent than the (S)-enantiomer [i.e. IC50 < 1 nM for 24(R) and IC50 = 225 nM for 24(S); IC50 = 2.3 nM for 27(R) and IC50 = 1299 nM for 27(S)] [149]. Some compounds reported in this patent exhibited favorable pharmacokinetic properties, metabolic stability, and permeability suitable for oral administration.

Table 4.

Examples of representative 4-oxo-1,4-dihydropyridine derivatives developed by Boehringer Ingelheim GmbH.

graphic file with name nihms-1847152-t0022.jpg
Compound R1 R3 R5 X IC50 (nM)a Reference
20 CH2CH3 graphic file with name nihms-1847152-t0023.jpg CF3 C < 1b [146]
21 CH(CH3)2 graphic file with name nihms-1847152-t0024.jpg CHF2 C < 1b [146]
22 graphic file with name nihms-1847152-t0025.jpg graphic file with name nihms-1847152-t0026.jpg CHF2 C < 1b [146]
23 graphic file with name nihms-1847152-t0027.jpg graphic file with name nihms-1847152-t0028.jpg CF3 C < 1c [147]
24 graphic file with name nihms-1847152-t0029.jpg CH3 CF3 C < 1c [147]
25 graphic file with name nihms-1847152-t0030.jpg CH3 CF3 N 3.7c [147]
26 graphic file with name nihms-1847152-t0031.jpg graphic file with name nihms-1847152-t0032.jpg CF3 N 2.8c [147]
27 graphic file with name nihms-1847152-t0033.jpg graphic file with name nihms-1847152-t0034.jpg CF3 N 2.3c [147]
28 graphic file with name nihms-1847152-t0035.jpg graphic file with name nihms-1847152-t0036.jpg CHF2 C < 1c [147]
29 CH(CH3)2 graphic file with name nihms-1847152-t0037.jpg CHF2 C 3 [148]
a

The results are means of two independent experiments, each performed in duplicate (HNE enzyme assay).

b

IC50 values are related to the racemate.

c

IC50 values are related to eutomer [(R)-enantiomer].

Boehringer Ingelheim also investigated other monocyclic scaffolds, such as 2-pyridone [151], 2-pyrazinone [151], and dihydropyrimidone [152] as HNE inhibitors reaching IC50 values in the nanomolar/picomolar range. In Figure 5 are presented two potent compounds belonging to the 2-oxo-1,2-dihydropyridine (30) and 3-oxo-3,4-dihydropyrazine (31) series, respectively, and a general formula (32) representative of a library of about 215 compounds with 2-oxo-1,2,3,4-tetrahydropyrimidine scaffold, which are very potent HNE inhibitors exhibiting picomolar IC50 values and in some cases, an additional inhibitory effect on neutrophil PR3, which is favorable for pharmacological efficacy. Moreover, the dihydropyrimidone derivatives of structure 32 exhibited favorable metabolic stability, appropriate permeability, and aqueous solubility. Among this library of compounds, the derivative that has R = CH3 and R1 = 1-methylpyridin-2(1H)-one exhibited the best properties. In particular, its half-life (t1/2 in vitro) in human liver microsomes was >130 min, its aqueous stability was 0.073 mg/mL, and its metabolic stability with human hepatocytes calculated as hepatic intrinsic clearance value (Qh [%]) was 5%. Another important parameter for oral absorption is the measurement of the ability of a compound to pass across the cell membrane. The in vitro model used for determination of drug transport across the membrane is human cancer colon carcinoma cells (Caco-2) grown on permeable filter supports. The selected compound exhibited a permeability coefficient [PE] and drug adsorption from the intestine into the blood [AB] (PEAB) value of 12 × 10−6 cm/s and a PE and drug secretion from the blood back into intestine [BA] (PEBA) value of 120 × 10−6. These values make it an optimal candidate for oral administration [152].

Figure 5.

Figure 5.

Examples of 2-oxo-1,2-dihydropyridine (30), 3-oxo-3,4-dihydropyrazine (31), and general structure of 2-oxo-1,2,3,4-tetrahydropyrimidine (32) derivatives developed by Boehringer Ingelheim GmbH.

The encouraging results obtained with dihydropyrimidone derivatives (32) encouraged Boehringer researchers to further explore this scaffold and developed new series of tetrahydrocycloalkylpyrimidone analogs [153155], which are summarized in Tables 5 and 6. These inhibitors were developed for pharmaceutical composition and methods for the treatment of pulmonary disease and other inflammatory pathologies. The representative tetrahydrociclopentane pyrimidone derivatives (33–47) shown in Table 5 are very potent HNE inhibitors, with IC50 values in the low nanomolar/picomolar range. Compounds 33–42 and 47 which contain various groups R4 belong to the 5th generation [45]. The presence of a group at position 2 on the p-cyanophenyl ‘freezes’ the structure in an ideal bioactive conformation, thus pre-organizing the inhibitor for the forthcoming binding event. This additional substituent increases the potency of inhibitor, although it does not directly interact with the target. Compounds 43–46, although missing the ‘freezing’ group, are equally powerful HNE inhibitors, with IC50 < 1 nM but belong to the 4th generation and, as a result, are less potent and stable than the compounds belonging to the 5th generation.

Table 5.

Examples of 3,4,6,7-tetrahydro-1H-cyclopentane[d]pyrimidine-2,5-dione derivatives developed by Boehringer Ingelheim GmbH.

graphic file with name nihms-1847152-t0038.jpg
Compound R4 R3 X IC50 (nM)a Reference
33 SO2CH3 CH3 C < 1 [151]
34 SO2CH2CH3 graphic file with name nihms-1847152-t0039.jpg N < 1 [151]
35 SO2CH3 CH2CH2OH C <1 [151]
36 graphic file with name nihms-1847152-t0040.jpg CH3 C 2.2 [152]
37 graphic file with name nihms-1847152-t0041.jpg CH3 C 1.8 [152]
38 graphic file with name nihms-1847152-t0042.jpg CH3 C 1.3 [152]
39 graphic file with name nihms-1847152-t0043.jpg CH3 C 7.5 [152]
40 graphic file with name nihms-1847152-t0044.jpg CH3 C 2.1 [152]
41 graphic file with name nihms-1847152-t0045.jpg CH3 C 1.2 [152]
42 graphic file with name nihms-1847152-t0046.jpg CH3 C < 1 [152]
43 H H C 11.5 [153]
44 H CH3 C 2.4 [153]
45 H graphic file with name nihms-1847152-t0047.jpg C <1 [153]
46 H graphic file with name nihms-1847152-t0048.jpg C <1 [153]
47 SO2CH3 graphic file with name nihms-1847152-t0049.jpg C <1 [153]
a

The results are means of two independent experiments, each performed in duplicate (HNE enzyme assay).

Table 6.

Examples of condensed dihydropyrimidone derivatives developed by Boehringer Ingelheim GmbH.

graphic file with name nihms-1847152-t0050.jpg
Compound R4 R3 X IC50 (nM)a Reference
48 b SO2CH3 H CH2 4.8 [154]
49 b H graphic file with name nihms-1847152-t0051.jpg CH2 < 1 [154]
50 b H graphic file with name nihms-1847152-t0052.jpg CH2 < 1 [154]
51 b SO2CH3 graphic file with name nihms-1847152-t0053.jpg CH2 < 1 [154]
52 b SO2CH3 graphic file with name nihms-1847152-t0054.jpg CH2 < 1 [154]
53 b SO2CH3 graphic file with name nihms-1847152-t0055.jpg CH2 1 [155]
54 b SO2CH3 CH2CH2OH CH2 < 1 [155]
55 b CONH2 CH3 CH2 1.6 [152]
56 c H H NH 3.1 [156]
57 c H graphic file with name nihms-1847152-t0056.jpg NH 1.3 [156]
58 c H graphic file with name nihms-1847152-t0057.jpg NH < 1 [156]
59 c graphic file with name nihms-1847152-t0058.jpg CH3 NH < 1 [156]
60 c CONH2 CH3 N-CH3 < 1 [156]
61 c SO2CH3 CH3 NH < 1 [156]
a

The results are means of two independent experiments, each performed in duplicate (HNE enzyme assay).

b

The absolute configuration (S) has been unambiguously assigned by X-ray structure analysis.

c

The absolute configuration (R) has been unambiguously assigned by X-ray structure analysis.

For the condensed dihydropyrimidones (48–61) presented in Table 6, the absolute configuration responsible for HNE inhibitory activity was unambiguously assigned, leading to IC50 < 1nM [156158]. They exhibit favorable in vivo potency in models of HNE-induced lung injury in mice, rats, or hamsters; good metabolic stability (t1/2 > 130 min); good permeability across the cellular membrane; and reduced in vivo intrinsic clearance (CL) [156158]. Compound 51, with a predicted human hepatic in vivo blood clearance of 0 ml/min/Kg, a PEAB of 9.86 × 10−6, a PEBA of 53.5 × 10−6, and aqueous stability of 0.086 mg/mL, represents one of the best pharmacokinetic profiles. However, numerous examples of these compounds containing a carbamoyl substituent at position 3 of the dihydropyrimidine scaffold exhibited improved AB permeability and/or reduced efflux ratio.

5.3. Bayer Healthcare AG

Currently, Bayer Healthcare AG has one HNE inhibitor in phase II clinical trials (BAY 85-8501) (already reported in Figure 3 and Table 1), which belongs to the 5th generation [45]. Analogous to the other pyrimidone derivatives developed by Bayer, BAY 85-8501 contains a methylsulfonyl group at R4 of the p-cyanophenyl ring, which induces a frozen bioactive conformation leading to a significant increase in potency, selectivity, and stability (Table 7). The high binding efficiency of the pyrimidone class is based on an induced-fit binding mode: upon binding of these molecules, the S2 pocket of HNE widens significantly to accommodate the northern para-cyanoaryl residue. The presence of a substituent R4 is important to increase potency into picomolar range. Other potent and interesting dihydropyrimidones are shown in Table 7 (compounds 62–67) [159], all exhibiting good in vivo stability, also under oxidative conditions. Due to the high impact of this new class of HNE inhibitors, Bayer started to explore bi-heterocycles containing the pyrimidine nucleus with different substituents (Figure 6) [159,160]. Compounds 68–71 exhibit excellent activities against HNE with IC50 < 0.3 nM. However, pyrimidopyridazine derivative 71 (BAY-8040) [161] is the best in vivo candidate from this series. This compound showed good potency and in vivo pharmacokinetic properties, and it was tested in vivo in a monocrotaline rat model of PAH. Treatment with BAY-8040 (71) (50 mg Kg−1, p.o) from days 14 to 28 significantly decreased right ventricular systolic pressure and right ventricular hypertrophy relative to placebo, with a significant beneficial effect on right ventricular dysfunction and preservation of systemic arterial pressure. Furthermore, compound 71 improved the cardiac output and arterial oxygenation in rats with MCT-induced PAH. Thus, BAY-8040 was able not only to suppress the inflammation at the basis of the pathology but also reverse tissue remodeling in the PAH setting [161].

Table 7.

Examples of 4-(4-cyano-2-thionyl)dihydropyrimidones developed by Bayer HealthCare AG.

graphic file with name nihms-1847152-t0059.jpg
Compoundb R4 R3 IC50 (nM)a Reference
62 SO2CH3 H 0.5 [157]
63 SO2CH3 graphic file with name nihms-1847152-t0060.jpg < 0.3 [157]
64 SO2CH3 SO2CH3 < 0.3 [157]
BAY 858501 SO2CH3 CH3 < 0.3 [129,130,157]
65 SO2CH3 graphic file with name nihms-1847152-t0061.jpg < 0.3 [157]
66 SO2CH3 CH2CN < 0.3 [157]
67 SO2Ph H < 0.3 [157]
a

The results are means of two independent experiments, each performed in duplicate (HNE enzyme assay).

b

The absolute configuration (S) has been unambiguously assigned by X-ray structure analysis.

Figure 6.

Figure 6.

Heterocyclic-fused pyrimidine derivatives developed by Bayer Healthcare AG.

5.4. Other companies

AstraZeneca has extensively investigated HNE inhibitors, producing a large number of compounds with a 2-pyridone scaffold. Among these products is Alvelestat (AZD9668) (see Figure 3), which is currently in clinical trials, as previously mentioned in Table 1. AstraZeneca also developed 2-pyrazinone derivatives, and compound 72 (AZD9819), shown in Figure 7, is an interesting and representative example [162]. KRP-109 (73) is claimed to be a selective, water-soluble competitive HNE inhibitor developed by Kyorin Pharmaceutical Co., Ltd. (Tokyo, Japan) [163]. A distinct feature of KRP-109 is its ability to accumulate in high concentrations in lung tissue, thereby inhibiting the activity of HNE released by infiltrating neutrophils under inflammatory conditions. Moreover, KRP-109 reduced lung inflammation in a murine model of severe pneumococcal pneumonia and was also able to decrease the degradation of mucin in CF sputum in vitro [164].

Figure 7.

Figure 7.

2-pyrazinone derivative (72) developed by Astrazeneca and KRP-109 (73) developed by Kyorin Pharmaceutical Co.

5.5. Miscellaneous

The California Institute for Biomedical Research has recently developed a series of dimeric compounds with the general formula Y1-A1-X-A2-Y2 (Figure 8), where A1 and A2 are linear or branched alkyl chains, X is a group such as -C(O)-, -C(O) C(O)-, -NHCO-, and Y1 and Y2 are fragments of drugs, for example, Sivelestat and Alvelestat. These compounds are disclosed in a patent [165] as potentially useful for the treatment of lung diseases, such as COPD and cystic fibrosis, but also for intestinal disease, including inflammatory bowel disease, and a representative product (compound 74) exhibiting IC50 < 150 nM is shown in Figure 8.

Figure 8.

Figure 8.

Example of a dimeric compound developed by the California Institute for Biomedical Research.

Figure 9 shows compounds 75–77 with isoxazol-5(2H)-one scaffolds, which were developed by Giovannoni and co-workers [166,167] as potent and stable HNE inhibitors exhibiting IC50 values in the low nanomolar range. These compounds, which were designed as molecular simplifications of other compounds synthesized in the same research laboratory [81,168172], act as competitive, pseudo-irreversible HNE inhibitors, and docking studies seem to highlight that the attack of Ser195 is directed to the endocyclic CO group at position 5 to form the acyl-enzyme complex.

Figure 9.

Figure 9.

Examples of HNE inhibitors reported in the recent literature.

Finally, worthy of mention is a new class of HNE inhibitors with a diazaborine nucleus [173]. Figure 9 shows two examples of these compounds (78, 79). Although endowed with moderate activity in the micromolar range, these compounds exhibit good selectivity toward elastase, and no inhibition was observed against a panel of five closely related serine proteases. Moreover, they are very stable in phosphate buffer at pH 7.4 and in human plasma. Computational studies performed on this system suggest that the mechanism of action involves the formation of a reversible covalent bond between the diazoborine boron center and the catalytic serine oxygen.

6. Conclusion

The selected patents reported for the period 2014–2018 represent a large number of HNE inhibitors obtained through the use of rational drug design and chemical modifications of potent lead compounds and by the study of the structural properties of neutrophil elastase. Mono- and bi-heterocycle nitrogen-containing scaffolds are the most investigated, leading to products with similar chemical characteristics in many cases. Most of the new compounds belong to the 4th and 5th generations, inserting inside the active site in a perpendicular manner. This particular interaction produces a new S2 pocket or allosteric site without interacting with the catalytic triad. Both in the monocyclic and bicycles compounds, a p-cyanophenyl fragment is present at the north position, and a m-trifluorophenyl at nitrogen at the south position, both being important for binding to HNE via an induced fit with a frozen bioactive conformation. These new potent, nonreactive molecules exhibit significant boosts in potency, selectivity, and metabolic stability, especially under pathophysiological conditions.

7. Expert opinion

Human neutrophil elastase (HNE) is a therapeutic target of considerable interest because of its involvement in many inflammatory diseases, as is demonstrated by the number of HNE inhibitors patents developed in the last decade by pharmaceutical companies. Currently, the two HNE inhibitors on the market are used for the treatment of α1-antitrypsin deficiency (Prolastin®) and ALI and ARDS associated with systemic inflammatory response syndrome (SIRS) (Sivelestat®). In addition to their use for cardiopulmonary diseases, HNE inhibitors were proposed to be useful for other inflammatory pathologies, such as psoriasis, rheumatoid arthritis, bowel intestinal inflammation, and also cancer, since HNE activity has been directly related to the progression and metastasis of cancer cells.

Several key advances in the last couple of years will likely stimulate further development of newer HNE inhibitors. The first originates from the observation that in addition to HNE, other proteases contribute to tissue damage; thus, dual HNE/PR3 inhibitors have been proposed as an innovative approach for the treatment of neutrophilic inflammatory diseases by Hwang and coworkers [174,175], opening a new window in the discovery of new drugs. Second, the activity of neutrophil serine proteases, including elastase and Pr3, has been implicated in some autoimmune diseases, such as type 1 diabetes (T1D) [94]. Although the role of neutrophils in the development of T1D remains unknown, a recent patent [176] reports that the serum concentration and the enzymatic activities of neutrophil elastase and Pr3 are diagnostic biomarkers for the risk of developing autoimmune diabetes. Another interesting approach being considered to regulate levels of protease activity and restore the protease-antiprotease balance is to indirectly modulate HNE function by inhibiting those peptidases that active this enzyme during development in the bone marrow. For example, DPP1 (dipeptidyl peptidase 1 or Cathepsin C) is a lysosomal cysteine protease that plays a key role in the activation of the proinflammatory neutrophil serine proteases, HNE, PR3, and Cathepsin G by cleaving the N-terminal dipeptide during neutrophil maturation. The inhibition of DPP1 could generate neutrophils lacking these active proteases, thereby reducing the local release and the subsequent damage [177].

Finally, the study published by Marto and coworkers [76] opens a new perspective on the administration routes for HNE inhibitors, till now used intravenously. The recent demonstration of the involvement of HNE in psoriasis and atopic dermatitis stimulated the research in this field, in particular, the use of starch-based nanocapsules as nanobiomaterial for topical drug delivery. The low anti-inflammatory activity of the potent ER143 inhibitor (IC50 = 0.67 nM), when used as a solution, is considerably increased if the same compound is associated with a starch-based nanoparticulate system (StNC). The resulted formulation exhibited interesting characteristics and in vivo studies showed that the topical application of StNC-ER143 strongly inhibited the acute inflammatory response as evident by the decrease of edema (about 92% inhibition), erythema and neutrophil infiltration.

In conclusion, taking into account the interesting results achieved in the field of HNE inhibitors and their medical applications, this topic appears worthy of further in-depth analysis. At the present, the main effort by pharmaceutical companies is pointed to improve quality of life and reduce the risk of death in pulmonary pathologies, overall COPD which in 2020 is expected to be the 4th cause of death in the world. Moreover, the new recent discoveries regarding as the involvement of HNE in other pathologies as the possibility to indirectly modulate HNE function, open new intriguing possibilities.

Article highlights.

  • This review focuses on neutrophil elastase (NE) inhibitor patents reported in the literature over 2014–2018.

  • The main application of HNE inhibitors concerns the treatment of (cardio)pulmonary inflammatory diseases; other possible uses include skin pathologies, rheumatoid arthritis, and cancer.

  • This review follows the recent classification proposed for HNE inhibitors, which considers five generations, starting from biologicals (1st generation) to ‘pre-adaptive pharmacophore derived from 4th generation inhibitors’ (5th generation). Many of these compounds show picomolar activity, high selectivity, and drug-like pharmacokinetic properties.

  • In this review, a systematic investigation of pharmaceutical companies working on HNE inhibitors was done. Accordingly, for each company, the most interesting results are reported. A miscellaneous from literature follows.

  • Non-peptidic NE inhibitors are low-molecular-mass synthetic compounds containing nitrogen heterocycles such as pyrimidinone, pyrimidinedione, 2-pyrazinone, pyrimidine, pyridine, 4-pyridone, 2-pyridone, and pyridazine.

This box summarizes key points contained in the article.

Funding

This paper was funded in part by the National Institutes of Health IDeA Program COBRE Grant GM110732, the USDA National Institute of Food and Agriculture Hatch project 1009546, and the Montana State University Agricultural Experiment Station.

Footnotes

Declaration of interest

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

Reviewer disclosures

A reviewer on this manuscript has disclosed that they are trying to develop HNE inhibitors and filed patent applications. All other peer reviewers on this manuscript have no relevant financial relationships or otherwise to disclose.

References

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

  • 1.Soualmia F, El Amri C. Serine protease inhibitors to treat inflammation: a patent review (2011–2016). Expert Opin Ther Pat. 2018;28 (2):93–110. [DOI] [PubMed] [Google Scholar]; • Useful review on the application of recent serine protease inhibitors in inflammatory pathologies.
  • 2.Page MJ, Di Cera E. Serine peptidases: classification, structure and function. Cell Mol Life Sci. 2008;65(7–8):1220–1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Di Cera E. Serine proteases. IUBMB Life. 2009;61(5):510–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hedstrom L. Serine protease mechanism and specificity. Chem Rev. 2002;102:4501–4524. [DOI] [PubMed] [Google Scholar]
  • 5.Rawlings ND, Barrett AJ, Bateman A. Using the MEROPS database for proteolytic enzymes and their inhibitors and substrates. Curr Protoc Bioinformatics. 2014;12(48):1–33. [DOI] [PubMed] [Google Scholar]
  • 6.Heutinck KM, Ten Berge IJ, Hack CE, et al. Serine proteases of the human immune system in health and disease. Mol Immunol. 2010;47(11–12):1943–1955. [DOI] [PubMed] [Google Scholar]
  • 7.Perera NC, Schilling O, Kittel H, et al. NSP4, an elastase-related protease in human neutrophils with arginine specificity. Proc Natl Acad Sci USA. 2012;109(16):6229–6234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Almeida RP, Vanet A, Witko-Sarsat V, et al. Azurocidin, a natural antibiotic from human neutrophils: expression, antimicrobial activity, and secretion. Protein Expr Purif. 1996;7(4):355–366. [DOI] [PubMed] [Google Scholar]
  • 9.Chertov O, Ueda H, Xu LL, et al. Identification of human neutrophil-derived cathepsin G and azurocidin/CAP37 as chemoattractants for mononuclear cells and neutrophils. J Exp Med. 1997;186(5):739–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Reeves EP, Lu H, Jacobs HL, et al. Killing activity of neutrophil is mediated through activation of proteases by K+ flux. Nature. 2002;416(6878):291–297. [DOI] [PubMed] [Google Scholar]
  • 11.Stuart LM, Ezekowitz RA. Phagocytosis: elegant complexity. Immunity. 2005;22:539–550. [DOI] [PubMed] [Google Scholar]
  • 12.Faurschou M, Borregaard N. Neutrophil granules and secretory vesicles in inflammation. Microbes Infect. 2003;5(14):1317–1327. [DOI] [PubMed] [Google Scholar]
  • 13.Pham CTN. Neutrophil serine proteases: specific regulators of inflammation. Nat Rev Immunol. 2006;6:541–550. [DOI] [PubMed] [Google Scholar]; •• Important review on the role serine protease in inflammation.
  • 14.Pham CTN. Neutrophil serine proteases fine-tune the inflammatory response. Int J Biochem Cell Biol. 2008;40:1317–1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tsai YF, Hwang TL. Neutrophil elastase inhibitors: a patent review and potential applications for inflammatory lung disease (2010–2014). Expert Opin Ther Pat. 2015;25(10):1145–1158. [DOI] [PubMed] [Google Scholar]
  • 16.Korkmaz B, Moreau T, Gauthier F. Neutrophil elastase, proteinase 3 and cathepsin G: physicochemical properties, activity and physio-pathological functions. Biochimie. 2008;90:227–242. [DOI] [PubMed] [Google Scholar]; • An interesting overview of elastase, proteinase 3 and cathepsin G and their characteristic.
  • 17.Zimmer M, Medcalf RL, Fink TM, et al. Three human elastase-like genes coordinately expressed in the myelomonocyte lineage are organized as a single genetic locus on 19pter. Proc Natl Acad Sci USA. 1992;89(17):8215–8219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Horwitz M, Benson KF, Person RE, et al. Mutations in ELA2, encoding neutrophil elastase, define a 21-day biological clock in cyclic hematopoiesis. Nat Genet. 1999;23:433–436. [DOI] [PubMed] [Google Scholar]
  • 19.Horwitz MS, Duan Z, Korkmaz B, et al. Neutrophil elastase in cyclic and severe congenital neutropenia. Blood. 2007;109 (5):1817–1824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lungarella G, Menegazzi R, Gardi C, et al. Identification of elastase in human eosinophils: immunolocalization, isolation, and partial characterization. Arch Biochem Biophys. 1992;292(1):128–135. [DOI] [PubMed] [Google Scholar]
  • 21.Tsuji N, Moriwaki S, Suzuki Y, et al. The role of elastases secreted by fibroblasts in wrinkle formation: implication through selective inhibition of elastase activity. Photochem Photobiol. 2001;74(2):283–290. [DOI] [PubMed] [Google Scholar]
  • 22.Liou TG, Campbell EJ. Nonisotropic enzyme-inhibitor interactions: a novel non-oxidative mechanism for quantum proteolysis by human neutrophils. Biochemistry. 1995;34(49):16171–16177. [DOI] [PubMed] [Google Scholar]
  • 23.Owen CA, Campbell EJ. Neutrophil proteinases and matrix degradation. The cell biology of pericellular proteolysis. Semin Cell Biol. 1995;6:367–376. [DOI] [PubMed] [Google Scholar]
  • 24.Allen DH, Tracy PB. Human coagulation factor V is activated to the functional cofactor by elastase and cathepsin G expressed at the monocyte surface. J Biol Chem. 1995;270:1408–1415. [DOI] [PubMed] [Google Scholar]
  • 25.Walker B, Lynas JF. Strategies for the inhibition of serine proteases. Cell Mol Life Sci. 2001;58:596–624. [DOI] [PMC free article] [PubMed] [Google Scholar]; • An interesting treatise of the mechanism of action of serine proteases.
  • 26.Hartley BS, Kilby BA. The reaction of p-nitrophenyl esters with chymotrypsin and insulin. Biochem J. 1954;56:288–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bobofchak KM, Pineda AO, Mathews FS, et al. Energetic and structural consequences of perturbing Gly-193 in the oxyanion hole of serine proteases. J Biol Chem. 2005;280:25644–25650. [DOI] [PubMed] [Google Scholar]
  • 28.Chua F, Laurent GJ. Neutrophil elastase: mediator of extracellular matrix destruction and accumulation. Proc Am Thorac Soc. 2006;3:424–427. [DOI] [PubMed] [Google Scholar]
  • 29.Bieth JG. Elastases: catalytic and biological properties. In: Mecham R, editor. Regulationof matrix accumulation. New York: Academic Press; 1986. p. 217–230. [Google Scholar]
  • 30.Gadek JE, Fells GA, Wright DG, et al. Human neutrophil elastase functions as a type III collagen “collagenase.”. Biochem Biophys Res Commun. 1980;95:1815–1822. [DOI] [PubMed] [Google Scholar]
  • 31.Janoff A, Scherer J. Mediators of inflammation in leukocyte lysosomes: elastinolytic activity in granules of human polymorphonuclear leukocytes. J Exp Med. 1968;128:1137–1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Barret AJ, Salvesen G. An introduction to the proteinases. In: Barrett AJ, Salvesen G (eds). Protease inhibitors. Vol. 1. New York: Elsevier; 1986. p. 1–22. [Google Scholar]
  • 33.Havemann K, Janoff A. Neutral proteases of human polymorphonuclear leukocytes. Baltimore, (MD): Urban & Schwarzenberg; 1978. [Google Scholar]
  • 34.Abe H, Okajima K, Okabe H. Granulocyte protease and hydrogen peroxide synergistically inactivate thrombomodulin of endothelial cells in vitro. J Lab Clin Med. 1994;123:874–881. [PubMed] [Google Scholar]
  • 35.States DJ, Fells GA, Crystal RG. Susceptibility of the HIV envelope protein to attack by neutrophil elastase. Clin Res. 1989;37:482A. [Google Scholar]
  • 36.Senior RM, Campbell EJ. Neutral proteinases from human inflammatory cells. A critical review of their role in extracellular matrix degradation. Clin Lab Med. 1983;3:645–666. [PubMed] [Google Scholar]
  • 37.Okada Y, Watanabe S, Nakanishi I, et al. Inactivation of tissue inhibitor of metalloproteinases by neutrophil elastase and other serine proteinases. FEBS Lett. 1988;229:157–160. [DOI] [PubMed] [Google Scholar]
  • 38.Cepinskas G, Sanding M, Pr K. PAF-induced elastase-dependent neutrophil trans endothelial migration is associated with the mobilization of the elastase to the neutrophil surface and localization to the migration front. J Cell Sci. 1999;112:1937–1945. [DOI] [PubMed] [Google Scholar]
  • 39.Hermant B, Bibert S, Concord E, et al. Identification of proteases involved in the proteolysis of vascular endothelium cadherin during neutrophil transmigration. J Biol Chem. 2003;278:14002–14012. [DOI] [PubMed] [Google Scholar]
  • 40.Belaaouaj A, McCarthy R, Baumann M, et al. Mice lacking neutrophil elastase reveal impaired host defense against gram negative bacterial sepsis. Nat Med. 1998;4(5):615–618. [DOI] [PubMed] [Google Scholar]
  • 41.Bardoel BW, Kenny EF, Sollberger G, et al. The balancing act of neutrophils. Cell Host Microbe. 2014;15(5):526–536. [DOI] [PubMed] [Google Scholar]
  • 42.Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535. [DOI] [PubMed] [Google Scholar]
  • 43.Kelly E, Greene CM, McElvaney NG. Targeting neutrophil elastase in cystic fibrosis. Expert Opin Ther Targets. 2008;12(2):145–157. [DOI] [PubMed] [Google Scholar]
  • 44.Cowburn AS, Condliffe AM, Farahi N, et al. Advances in neutrophil biology: clinical implications. Chest. 2008;134(3):606–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Von Nussbaum F, Li VMJ. Neutrophil elastase inhibitors for the treatment of (cardio)pulmonary diseases: into clinical testing with pre-adaptive pharmacophores. Bioorg Med Chem. 2015;25:4370–4381. [DOI] [PubMed] [Google Scholar]; •• Excellent publication concerning the new classification of HNE inhibitors in generations.
  • 46.Stockley RA. Neutrophils and the pathogenesis of COPD. Chest. 2002;121(5):151S–155S. [DOI] [PubMed] [Google Scholar]
  • 47.Ekeowa U, Gooptu B, Belorgey D, et al. Alpha-1-antitrypsin deficiency, chronic obstructive pulmonary disease and the serpinopathies. Clin Sci (London). 2009;116:837–850. [DOI] [PubMed] [Google Scholar]
  • 48.Silverman GA, Bird PI, Carrell W, et al. The serpins are an expanding superfamily of structurally similar but functionally diverse proteins. Evolution, mechanism of inhibition, novel functions, and a revised nomenclature. J Biol Chem. 2001;276:33293–33296. [DOI] [PubMed] [Google Scholar]
  • 49.Brew K, Nagase H. The tissue inhibitors of metalloproteinases (TIMPs): an ancient family with structural and function diversity. Biochim Biophys Acta. 2010;1803:55–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Turk V, Stoka V, Turk D. Cystatins: biochemical and structural properties and medical relevance. Front Biosci. 2008;13:5406–5420. [DOI] [PubMed] [Google Scholar]
  • 51.Miravitlles M. Alpha-1-antitrypsin and other proteinase inhibitors. Curr Opin Pharmacol. 2012;12:309–314. [DOI] [PubMed] [Google Scholar]
  • 52.Cooley J, Takayama TK, Shapiro SD, et al. The serpin MNEI inhibits elastase-like and chymotrypsin-like serine protease through efficient reactions at two active sites. Biochemistry. 2001;40:15762–15770. [DOI] [PubMed] [Google Scholar]
  • 53.Sallenave JM. The role of secretory leukocyte proteinase inhibitor and elafin (elastase-specific inhibitor/skin-derived antileukopro-tease) as alarm antiproteinases in inflammatory lung disease. Respir Res. 2000;1:87–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Alam SR, Newby DE, Henriksen PA. Role of the endogenous inhibitor, elafin, in cardiovascular injury from epithelium to endothelium. Biochem Pharmacol. 2012;83:695–704. [DOI] [PubMed] [Google Scholar]
  • 55.Sun Z, Yang P. Role of imbalance between neutrophil elastase and alpha 1-antitrypsin in cancer development and progression. Lancet Oncol. 2004;5(3):182–190. [DOI] [PubMed] [Google Scholar]
  • 56.Rubio F, Accurso FJ, Remold-O’Donnell E. Linkage of neutrophil serine proteases and decreased surfactant protein-A (SP-A) levels in inflammatory lung disease. Thorax. 2004;59:318–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Boudier C, Bieth J. Mucous proteinase inhibitor: a fast acting inhibitor of leukocyte elastase. Biochem Biophys Acta. 1989;995:36–41. [DOI] [PubMed] [Google Scholar]
  • 58.Gadek JE, Pacht ER. The protease-antiprotease balance within the human lung: implications for the pathogenesis of emphysema. Lung. 1990;168(Suppl):552–564. [DOI] [PubMed] [Google Scholar]
  • 59.Groutas WC, Dou D, Alliston KR. Neutrophil elastase inhibitors. Expert Opin Ther Pat. 2011;21(3):339–354. [DOI] [PMC free article] [PubMed] [Google Scholar]; • Very useful review on neutrophil elastase inhibitors over 2010–2014.
  • 60.Polverino E, Rosales-Mayor E, Dale GE, et al. The role of neutrophil elastase inhibitors in lung diseases. Chest. 2017;152(2):249–262. [DOI] [PubMed] [Google Scholar]
  • 61.Lucas SD, Costa E, Guedes RC, et al. Targeting COPD: advances on low molecular-weight inhibitors of human neutrophil elastase. Med Res Rev. 2011;33:73–101. [DOI] [PubMed] [Google Scholar]; • Very useful review on human neutrophil elastase inhibitors for the treatment of COPD.
  • 62.Barnes PJ, Stockley RA. COPD: current therapeutic interventions and future approaches. Eur Respir J. 2005;25:1084–1106. [DOI] [PubMed] [Google Scholar]
  • 63.Pandey KC, De S, Mishra PK. Role of proteases in chronic obstructive pulmonary disease. Front Pharmacol. 2017;8:512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ohbayashi H. Neutrophil elastase inhibitors as treatment for COPD. Expert Opin Investig Drugs. 2002;11:965–980. [DOI] [PubMed] [Google Scholar]
  • 65.Chan SCH, Shum DKY, Ip MSM. Sputum sol neutrophil elastase activity in bronchiectasis: differential modulation by syndecan-1. Am J Respir Crit Care Med. 2003;168(2):192–198. [DOI] [PubMed] [Google Scholar]
  • 66.Gramegna A, Amati F, Terranova L, et al. Neutrophil elastase in bronchiectasis. Respir Res. 2017;18:211–224. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Excellent review on the role of neutrophil elastase in bronchiectasis.
  • 67.Voynow JA, Fischer BM, Zheng S. Proteases and cystic fibrosis. Int J Biochem Cell B. 2008;40:1238–1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kawabata K, Hagio T, Matsuoka S. The role of neutrophil elastase in acute lung injury. Eur J Pharmacol. 2002;451:1–10. [DOI] [PubMed] [Google Scholar]
  • 69.Wang Z, Chen F, Zhai R, et al. Plasma neutrophil elastase and elafin imbalance is associated with acute respiratory distress syndrome (ARDS) development. PLoS ONE. 2009;4:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rabinovitch M, Guignabert C, Humbert M, et al. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res. 2014;115(1):165–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Schaaf B, Wieghorst A, Aries SP, et al. Neutrophil inflammation and activation in bronchiectasis: comparison with pneumonia and idiopathic pulmonary fibrosis. Respiration. 2000;67(1):52–59. [DOI] [PubMed] [Google Scholar]
  • 72.Fischer BM, Voynow JA. Neutrophil elastase induces MUC5AC gene expression in airway epithelium via a pathway involving reactive oxygen species. Am J Respir Cell Mol Biol. 2002;26:447–452. [DOI] [PubMed] [Google Scholar]
  • 73.Shao MX, Ueki IF, Nadel JA. Tumor necrosis factor α-converting enzyme mediates MUC5AC mucin expression in cultured human airway epithelial cells. Proc Natl Acad Sci USA. 2003;100:11618–11623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Meyer-Hoffert U, Wingertszahn J, Wiedow O. Human leukocyte elastase induces keratinocyte proliferation by epidermal growth factor receptor activation. J Investig Dermatol. 2004;123:338–345. [DOI] [PubMed] [Google Scholar]
  • 75.Glinski W, Jarzabek-Chorzelska M, Kuligowski M, et al. Basement membrane zone as a target for human neutrophil elastase in psoriasis. Arch Dermatol Res. 1990;282:506–511. [DOI] [PubMed] [Google Scholar]
  • 76.Marto J, Ruivo E, Lucas SD, et al. Starch nanocapsules containing a novel neutrophil elastase inhibitor with improved pharmaceutical performance. Eur J Pharm Biopharm. 2018;127:1–11. [DOI] [PubMed] [Google Scholar]
  • 77.Marto J, Gouveia LF, Gonçalves LM, et al. A quality by design (QbD) approach on starch-based nanocapsules: a promising platform for topical drug delivery. Colloids Surf B Biointerfaces. 2016;143:177–185. [DOI] [PubMed] [Google Scholar]
  • 78.Areias L, Ruivo E, Gonçalves L, et al. A unified approach toward the rational design of selective low nanomolar human neutrophil elastase inhibitors. RSC Adv. 2015;5(64):51717–51721. [Google Scholar]
  • 79.Cawston TE, Young DA. Proteinases involved in matrix turnover during cartilage and bone breakdown. Cell Tissue Res. 2010;339:221–235. [DOI] [PubMed] [Google Scholar]
  • 80.Di Cesare Mannelli L, Micheli L, Cinci L, et al. Effects of the neutrophil elastase inhibitor EL-17 in rat adjuvant-induced arthritis. Rheumatology. 2016;55:1285–1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Crocetti L, Schepetkin IA, Cilibrizzi A, et al. Optimization of N-benzoylindazole derivatives as inhibitors of human neutrophil elastase. J Med Chem. 2013;56:6259–6272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Demaria S, Pikarsky E, Karin M, et al. Cancer and inflammation: promise for biologic therapy. J Immunother. 2010;33:335–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Sato T, Takahashi S, Mizumoto T, et al. Neutrophil elastase and cancer. J Surg Oncol. 2006;15:217–222. [DOI] [PubMed] [Google Scholar]
  • 85.Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol. 2012;196:395–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Yamashita J, Tashiro K, Yoneda S, et al. Local increase in polymorphonuclear leukocyte elastase is associated with tumor invasiveness in non-small cell lung cancer. Chest. 1996;109:1328–1334. [DOI] [PubMed] [Google Scholar]
  • 87.Akizuki M, Fukutomi T, Takasugi M, et al. Prognostic significance of immunoreactive neutrophil elastase in human breast cancer: long-term follow-up results in 313 patients. Neoplasia. 2007;9 (3):260–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Vaguliene N, Zemaitis M, Lavinskiene S, et al. Local and systemic neutrophilic inflammation in patients with lung cancer and chronic obstructive pulmonary disease. BMC Immunol. 2013;14:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Antonicelli F, Bellon G, Debelle L, et al. Elastin-elastases and inflamm-aging. Curr Top Dev Biol. 2007;79:99–155. [DOI] [PubMed] [Google Scholar]
  • 90.Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16(7):431–446. [DOI] [PubMed] [Google Scholar]
  • 91.Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor micro-environment: expect the unexpected. J Clin Invest. 2016;125(9):3356–3364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Lerman I, Hammes SR. Neutrophil elastase in the tumor microenvironment. Steroids. 2017;133:96–101. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Excellent publication on the role of neutrophil elastase in the tumor microenvironment.
  • 93.Lerman I, Garcia-Hernandez ML, Rangel-Moreno J, et al. Infiltrating myeloid cells exert pro-tumorigenic actions via neutrophil elastase. Mol Cancer Res. 2017;15(9):1138–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Wang Y, Xiao Y, Zhong L, et al. Increased neutrophil elastase and proteinase 3 and augmented NETosis are closely associated with beta-cell autoimmunity in patients with type I diabetes. Diabetes. 2014;63(12):4239–4248. [DOI] [PubMed] [Google Scholar]
  • 95.Vicuña L, Simonetti M, Bali KK, et al. Selective inhibitors of neutrophil elastase for treating neuropathic pain and chronic pain states harbouring a neuropathic component. WO 2016050835 A2. 2016.
  • 96.Vicuña L, Strochlic DE, Latremoliere A, et al. The serine protease inhibitor SerpinA3N attenuates neuropathic pain by inhibiting T cell-derived leukocyte elastase. Nat Med. 2015;21 (5):518–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Weyer AD, Stucky CL. Repurposing a leukocyte elastase inhibitor for neuropathic pain. Nat Med. 2015;21(5):429–430. [DOI] [PubMed] [Google Scholar]
  • 98.Balia KK, Kuner R. Therapeutic potential for leukocyte elastase in chronic pain states harboring a neuropathic component. Pain. 2017;158(11):2243–2258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Bronze-da-Rocha E, Santos-Silva A. Neutrophil elastase inhibitors and chronic kidney disease. Int J Biol Sci. 2018;14(10):1343–1360. [DOI] [PMC free article] [PubMed] [Google Scholar]; • An interesting work about a new and recent application of HNE inhibitors.
  • 100.Pereira R, Rocha S, Borges A, et al. Elastase release during the hemodialysis procedure seems to induce changes in red blood cell membrane proteins. Hemodial Int Int Symp Home Hemodial. 2011;15:429–431. [DOI] [PubMed] [Google Scholar]
  • 101.Polanska B, Augustyniak D, Makulska I, et al. Elastase, alpha1-proteinase inhibitor, and interleukin-8 in children and young adults with end-stage kidney disease undergoing continuous ambulatory peritoneal dialysis. Arch Immunol Ther Ex. 2014;62:239–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Langhorst J, Elsenbruch S, Koelzer J, et al. Noninvasive markers in the assessment of intestinal inflammation in inflammatory bowel diseases: performance of fecal lactoferrin, calprotectin, and PMN-elastase, CRP, and clinical indices. Am J Gastroenterol. 2008;103:162–169. [DOI] [PubMed] [Google Scholar]
  • 103.Wen G, An W, Chen J, et al. Genetic and pharmacologic inhibition of the neutrophil elastase inhibits experimental atherosclerosis. J Am Heart Assoc. 2018;7(4):e008187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Matsuse H, Yanagihara K, Mukae H, et al. Association of plasma neutrophil elastase levels with other inflammatory mediators and clinical features in adult patients with moderate and severe pneumonia. Respir Med. 2007;101(7):1521–1528. [DOI] [PubMed] [Google Scholar]
  • 105.Arafat SN, Robert MC, Abud T, et al. Elevated neutrophil elastase in tears of ocular graft-versus-host disease patients. Am J Ophthalmol. 2017;176:46–52. [DOI] [PubMed] [Google Scholar]
  • 106.Bayer Corp: Prolastin. 2002. Company World Wide Web Site. Available from: http://edoc.com/prolastin
  • 107.Iwata K, Doi A, Ohji G, et al. Effect of neutrophil elastase inhibitor (sivelestat sodium) in the treatment of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS): a systematic review and meta-analysis. Intern Med. 2010;49:2423–2432. [DOI] [PubMed] [Google Scholar]
  • 108.Aikawa N, Kawasaki Y. Clinical utility of the neutrophil elastase inhibitor sivelestat for the treatment of acute respiratory distress syndrome. Ther Clin Risk Manage. 2014;10:621–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bernstein PR, Edwards PD, Williams JC. 2 Inhibitors of human leukocyte elastase. In: Ellis GP, Luscombe DK, editors. Progress in medicinal chemistry. Vol. 31. Amsterdam: Elsevier; 1994. p. 59–120. [DOI] [PubMed] [Google Scholar]
  • 110.Siedle B, Hrenn A, Merfort I. Natural compounds as inhibitors of human neutrophil elastase. Planta Med. 2007;73(5):401–420. [DOI] [PubMed] [Google Scholar]; •• Excellent broad review on natural compounds with HNE inhibition activity.
  • 111.Martins FT, Assis DM, Dos Santos MH, et al. Natural polyprenylated benzophenones inhibiting cysteine and serine protease. Eur J Med Chem. 2009;44:1230–1239. [DOI] [PubMed] [Google Scholar]
  • 112.Kim JY, Wang Y, Uddin Z, et al. Competitive neutrophil elastase inhibitory isoflavones from the roots of flemingia Philippinensis. Bioorg Chem. 2018;78:249–257. [DOI] [PubMed] [Google Scholar]
  • 113.Uddin Z, Zuopeng L, Song YH, et al. Visconata: A rare flavonol having long chain fatty acid from Dodonaea viscosa which inhibits Human neutrophil elastase (HNE). Tetrahedron Lett. 2017;58 (21):2507–2511. [Google Scholar]
  • 114.Salvador LA, Taori K, Biggs JS, et al. Potent elastase inhibitors from cyanobacteria: structural basis and mechanisms mediating cytoprotective and anti-inflammatory effects in bronchial epithelial cells. J Med Chem. 2013;56:1276–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Luo D, Chen Q-Y, Luesch H. Total synthesis of the potent marine-derived elastase inhibitor lyngbyastatin 7 and in vitro biological evaluation in model systems for pulmonary diseases. J Org Chem. 2016;81:532–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Sorroche PB, Fernández Acquier M, López Jove O, et al. Alpha-1 antitrypsin deficiency in COPD patients: a cross-sectional study. Arch Bronconeumol. 2015;51(11):539–543. [DOI] [PubMed] [Google Scholar]
  • 117.Clinicaltrials.gov NCT study code NCT02722304. 2018.
  • 118.Abouzaki NA, Christopher S, Trankle C, et al. Inhibiting the inflammatory injury after myocardial ischemia reperfusion with plasma-derived alpha-1 antitrypsin: a post Hoc analysis of the VCU-α1RT Study. J Cardiovasc Pharmacol. 2018;71(6):375–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Clinicaltrials.gov. NCT study code NCT01936896. 2016.
  • 120.Magenau JM, Goldstein SC, Peltier D, et al. α1-antitrypsin infusion for treatment of steroid-resistant acute graft-versus-host disease. Blood. 2018;131(12):1372–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Clinicaltrials.gov. NCT study code NCT01700036. 2018.
  • 122.Marcondes AM, Karoopongse E, Lesnikova M, et al. α-1-antitrypsin (AAT)–modified donor cells suppress GVHD but enhance the GVL effect: a role for mitochondrial bioenergetics. Blood. 2014;124 (18):2881–2891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Clinicaltrials.gov. NCT study code NCT01523821. 2018.
  • 124.Chun HJ, Yu PB. Elafin in pulmonary arterial hypertension. Beyond targeting elastases. Am J Respir Crit Care Med. 2015;191 (11):1217–1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Clinicaltrials.gov. NCT study code NCT03522935. 2019.
  • 126.Luther A, Moehle K, Chevalier E, et al. Protein epitope mimetic macrocycles as biopharmaceuticals. Curr Opin Chem Biol. 2017;38:45–51. [DOI] [PubMed] [Google Scholar]
  • 127.Clinicaltrials.gov. NCT study core NCT03748199. 2018.
  • 128.Obrecht D, Chevalier E, Moehle K, et al. β-hairpin protein mimetic technology in drug discovery. Drug Discov Today. 2012;9:e63–e69. [Google Scholar]; • An interesting paper on the use of β-Hairpin protein mimetic as HNE inhibitors.
  • 129.Gombert FO, Obrecht D, Sellier-Kessler O, et al. Beta-hairpin peptidomimetics as selective elastase inhibitors. WO 2015/096873 A1. 2015.
  • 130.Ludin C, Keller M Beta-hairpin peptidomimetic with elastase inhibitory acivity and aereosol dosage forms thereof. WO 2017/207117 A1. 2017.
  • 131.Clinicaltrials.gov. NCT study code NCT03636347. 2019.
  • 132.Clinicaltrials.gov. NCT study code NCT03679598. 2019.
  • 133.Clinicaltrials.gov. NCT study code NCT02669251. 2018.
  • 134.Von Nussbaum F, Li VM-J, Allerheiligen S, et al. Freezing the bioactive conformation to boost potency: the identification of BAY 85-8501, a selective and potent inhibitor of human neutrophil elastase for pulmonary diseases. ChemMedChem. 2015;10:1163–1173. [DOI] [PMC free article] [PubMed] [Google Scholar]; • Detailed preclinical studies of a HNE inhibitor in bronchiectasis.
  • 135.Clinicaltrials.gov. NCT study core NCT01818544. 2015.
  • 136.Carnini C, Brogin G, Patacchini R, et al. CHF6333: pharmacological and pharmacokinetic characterization of a novel potent inhaled inhibitor of neutrophil elastase. Am J Respir Crit Care Med. 2017;195:A4420. [Google Scholar]
  • 137.Clinicaltrials.gov. NCT study core NCT03056326. 2017.
  • 138.Miglietta D, Carnini C, Puviani V, et al. Pharmacological characterization of CHF6333, a novel potent inhaled inhibitor of neutrophil elastase. ERS International Congress, London. 2016. [Google Scholar]
  • 139.Alcaraz L, Heald RA, Sutton JM, et al. Pyrimidone compounds which are HNE inhibitors. US 9868740 B2 2018.
  • 140.Alcaraz L, Heald RA, Sutton JM, et al. 3-Oxo-2,3,5,8-tetrahydro-[1,2,4]triazolo[4,3-A]pyrimidine derivatives for the treatment of respiratory diseases. EP 2935274 B1. 2018.
  • 141.Armani E, Capaldi C, Sutton JM, et al. Compounds. US 9487528 B2. 2016.
  • 142.Armani E, Capaldi C Triazolinone compounds as HNE inhibitors. US 9890169 B2. 2018.
  • 143.Edwards C, Kulagowski J, Finch H Pyrimidine derivatives and their use in the treatment of respiratory diseases such as COPD. EP 2545057 B1. 2017.
  • 144.Capaldi C, Heald RA, Ray NC, et al. Inhibition of enzymes. US 9199984 B2. 2015
  • 145.Armani E, Capaldi C, Sutton JM, et al. Compounds. US 9409870 B2. 2016.
  • 146.Capaldi C, Armani E, Jennings ASR Compounds. US 9862706 B2. 2018.
  • 147.Sutton JM, Heald RA, Jennings ASR, et al. Novel compounds. US 2017/0340611. 2017.
  • 148.Oost T, Fiegen D, Substituted GC 4-pyridones and their use as inhibitors of neutrophil elastase activity. EP 2888242 B1. 2016.
  • 149.Oost T, Fiegen D, Gnamm C, et al. Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity. EP 2888248 B1. 2016.
  • 150.Oost T, Fiegen D, Gnamm C, et al. Substituted 4-pyridones and their use as inhibitors of neutrophil elastase activity. EP 2888249 B1. 2016.
  • 151.Gnamm C, Oost T Substituted pyridones and pyrazinones and their use as inhibitors of neutrophil elastase activity. EP 3107911 B1. 2018.
  • 152.Peters S, Anderskewitz R, Morschhaeuser G, et al. Substituted dihydropyrimidones and their use as inhibitors of neutrophil elastase activity. EP 3174860 B1. 2018.
  • 153.Oost T, Anderskewitz R, Gnamm C, et al. Substituted bicyclic dihydropyrimidones and their use as inhibitors of neutrophil elastase activity. US 9475779 B2. 2016.
  • 154.Peters S, Anderskewitz R, Gnamm C, et al. Substituted bicyclic dihydropyrimidones and their use as inhibitors of neutrophil elastase activity. US 9458113 B2. 2016.
  • 155.Gnamm C, Oost T, Peters S, et al. Substituted bicyclic dihydropyrimidones and their use as inhibitors of neutrophil elastase activity. US 9670166 B2. 2017.
  • 156.Gnamm C, Oost T, Peters S, et al. Substituted bicyclic dihydropyrimidones and their use as inhibitors of neutrophil elastase activity. US 9115093 B2. 2015.
  • 157.Oost T, Anderskewitz R, Gnamm C, et al. Substituted bicyclic dihydropyrimidones and their use as inhibitors of neutrophil elastase activity. US 9440930 B2. 2016.
  • 158.Gnamm C, Anderskewitz R, Hoesch H, et al. Substituted bicyclic dihydropyrimidones and their use as inhibitors of neutrophil elastase activity. US 9657015 B2. 2017.
  • 159.Von Nussbaum F, Karthaus D, Anlauf S, et al. 4-(4-cyano-2-thioaryl)dihydro-pyrimidones and use thereof. US 2015/0045344 A1. 2015.
  • 160.Von Nussbaum F, Karthaus D, Anlauf S, et al. Triazolo and tetrazolo pyrimidine derivatives as HNE inhibitors for treating COPD. US 9359362 B2. 2016.
  • 161.Von Nussbaum F, Li VM, Meibom D, et al. Potent and selective human neutrophil elastase inhibitors with novel equatorial ring topology: in vivo efficacy of the polar pyrimidopyridazine BAY-8040 in a pulmonary arterial hypertension rat model. Chem Med Chem. 2016;11:199–206. [DOI] [PubMed] [Google Scholar]
  • 162.Hansen P, Ivarsoon M, Lawitz K, et al. 2-pyrazinone derivatives for the treatment of disease or condition in which inhibition of neutrophil elastase activity is beneficial. CA 2650553 C. 2014.
  • 163.Yamada K, Yanagihara K, Araki N, et al. In vivo efficacy of KRP-109, a novel elastase inhibitor, in a murine model of severe pneumococcal pneumonia. Pulm Pharmacol Ther. 2011;24:660–665. [DOI] [PubMed] [Google Scholar]
  • 164.Chillappagari S, Müller C, Mahavadi P, et al. A small molecule neutrophil elastase inhibitor, KRP-109, inhibits cystic fibrosis mucin degradation. J Cyst Fibros. 2016;15:325–331. [DOI] [PubMed] [Google Scholar]
  • 165.Chatterjee AK, Kumar M, Schultz PG Elastase inhibitors. US 2017/0144998 A1. 2017.
  • 166.Vergelli C, Schepetkin IA, Crocetti L, et al. Isoxazol5(2H)-one: a new scaffold for potent human neutrophil elastase (HNE) inhibitors. J Enz Inhib Med Chem. 2017;32(1):821–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Giovannoni MP, Schepetkin IA, Quinn MT, et al. Synthesis, biological evaluation, and molecular modelling studies of potent human neutrophil elastase (HNE) inhibitors. J Enz Inhib Med Chem. 2018;33(1):1108–1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Crocetti L, Giovannoni MP, Schepetkin IA, et al. Design, synthesis and evaluation of N-benzoylindazole derivatives and analogues as inhibitors of human neutrophil elastase. Bioorg Med Chem. 2011;19:4460–4472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Crocetti L, Schepetkin IA, Ciciani G, et al. Synthesis and pharmacological evaluation of indole derivatives as deaza analogues of potent human neutrophil elastase inhibitors. Drug Dev Res. 2016;77:285–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Giovannoni MP, Schepetkin IA, Crocetti L, et al. Cinnoline derivatives as human neutrophil elastase inhibitors. J Enz Inhib Med Chem. 2016;31(4):628–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Crocetti L, Bartolucci G, Cilibrizzi A, et al. Synthesis and analytical characterization of new thiazol-2- (3H)- ones as human neutrophil elastase (HNE) inhibitors. Chem Cent J. 2017;11:127–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Crocetti L, Giovannoni MP, Schepetkin IA, et al. 1H-pyrrolo[2,3-b] pyridine: a new scaffold for human neutrophil elastase (HNE) inhibitors. Bioorg Med Chem. 2018;26:5583–5595. [DOI] [PubMed] [Google Scholar]
  • 173.António JPM, GonçAlves LM, Guedes RC, et al. Diazaborines as new inhibitors of human neutrophil elastase. ACS Omega. 2018;3:7418–7423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Heisch P-W, Hwang T-L, Wang W-H, et al. Oxime-based compound, pharmaceutical composition containing the same and method for preparing the same. US 9073833 B1. 2015.
  • 175.Hwang T-L, Wang W-H, Wang T-Y, et al. Synthesis and pharmacological characterization of 2-aminobenzaldeyde oxime analogs as dual inhibitors of neutrophil elastase and proteinase 3. Bioorg Med Chem. 2015;23:1123–1134. [DOI] [PubMed] [Google Scholar]
  • 176.Xu A, Wang Y, Zhong L, et al. Methods and compositions for use of neutrophil elastase and proteinase 3 as diagnostic biomarkers. US 9625460 B2. 2017.
  • 177.Doyle K, Lönn H, Käck H, et al. Discovery of second generation reversible covalent DPP1 inhibitors leading to an oxazepane amidoacetonitrile based clinical candidate (AZD7986). J Med Chem. 2016;59:9457–9472. [DOI] [PubMed] [Google Scholar]

RESOURCES