Next Article in Journal
Peroxydisulfate Activation by Biochar from Banana Peel Promoted with Copper Phosphide for Bisphenol S Degradation in Aqueous Media
Previous Article in Journal
Modification of High-Surface-Area Carbons Using Self-Limited Atomic Layer Deposition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancements in Serine Protease Inhibitors: From Mechanistic Insights to Clinical Applications

1
College of Chemistry, Fuzhou University, Fuzhou 350116, China
2
The National & Local Joint Engineering Research Center on Biopharmaceutical and Photodynamic Therapy Technologies, Fuzhou University, Fuzhou 350116, China
*
Authors to whom correspondence should be addressed.
Catalysts 2024, 14(11), 787; https://doi.org/10.3390/catal14110787
Submission received: 14 September 2024 / Revised: 24 October 2024 / Accepted: 4 November 2024 / Published: 5 November 2024
(This article belongs to the Section Biocatalysis)

Abstract

:
Serine proteases, a significant class of enzymes comprising approximately one-third of known human proteases, are ubiquitously present across various organisms. These enzymes typically exhibit highly conserved catalytic domain structures, and their activity is stringently regulated within the body, playing a pivotal role in numerous physiological processes. Dysregulation of serine protease activity can result in severe consequences, including excessive inflammation, heightened risk of thrombosis and cancer, and even mortality. Serine protease inhibitors have emerged as critical regulators, offering a broad range of physiological functions such as maintaining the coagulation–fibrinolysis balance, modulating inflammatory responses, accelerating wound healing, promoting apoptosis, and providing antitumor and antiviral effects. As a result, the development of serine protease inhibitors has become increasingly vital. In recent years, significant progress in the study of serine proteases has led to the pivotal role of various serine protease inhibitors in clinical diagnosis and treatment. This review explores the fundamental mechanisms of serine protease inhibitors, summarizes those that have been successfully integrated into clinical practice, and discusses the challenges encountered in their development along with partial solutions. These advancements lay the groundwork for further refinement and innovation in serine protease inhibitor therapeutics.

Graphical Abstract

1. Introduction

Serine proteases are enzymes that specifically cleave peptide bonds within proteins and are widely distributed across various organisms. They are named for the nucleophilic serine residue located at their active site. Among the known proteases in humans, nearly one-third can be classified as serine proteases. The most representative families include chymotrypsin, subtilisin, carboxypeptidase Y, and Clp protease [1,2].
The hydrolytic activity of serine proteases is driven by a “catalytic triad” composed of the amino acids aspartate (Asp), histidine (His), and serine (Ser), all of which are located at the enzyme’s active site [2,3]. The hydroxyl group of the serine residue plays a key role in the catalytic process by acting as a nucleophile, often donating electrons. The catalytic triad functions through a coordinated mechanism where the imidazole ring of histidine activates the serine hydroxyl group, enabling acid–base catalysis. Meanwhile, the aspartate residue stabilizes this interaction by forming a hydrogen bond with the nitrogen atom of the histidine imidazole ring (Figure 1) [4].
There are numerous types of serine proteases widely distributed in nature, and their catalytic activity is strictly regulated within biological systems, playing important roles in many key physiological pathways. Trypsin is a very common serine protease. As a well-known pancreatic digestive enzyme, trypsin aids in the digestion of proteins in the human body and plays a significant role in the digestive process. However, when the function of trypsin becomes disrupted, it can trigger a series of gastrointestinal diseases, posing a threat to human health [5,6,7]. Plasmin plays a crucial role in the fibrinolytic system by dissolving fibrin [8]. Coagulation factors are vital in the human coagulation cascade [9]. Dysregulation of the activity or expression of these proteases can have serious consequences (Figure 2). Multiple serine proteases play essential roles in the immune complement system [10,11,12]. However, excessive activation of serine proteases in the complement system can lead to over-inflammation and various diseases [13]. Transmembrane serine protease 2 (TMPRSS2), regulated by androgens, is a key enzyme involved in numerous pathological and physiological processes [14]. Elevated levels of TMPRSS2 are closely associated with various cancers [15,16]. Serine proteases released from activated leukocytes and mast cells, or generated through the coagulation cascade, are key components in the inflammatory response [17,18]. They are involved in producing pro-inflammatory cytokines and activating immune cells, and any dysregulation can lead to serious consequences [19,20,21]. Some serine proteases have been found to serve as diagnostic and therapeutic targets in clinical applications [22]. Serine proteases also play critical roles in the viral replication cycle: cellular serine proteases can mediate the entry of the viruses into the cytoplasmic membrane or facilitate their entry into cells [23,24].
Serine proteases in the human body can trigger diseases and even threaten life if their activity becomes uncontrolled. Therefore, maintaining the delicate balance of serine protease activity within the body is crucial [18,25]. As indispensable regulatory factors in the fibrinolytic system, endogenous serine protease inhibitors—Plasminogen activator inhibitors (PAIs)—play a vital role. On the one hand, they effectively slow down the conversion of plasminogen to plasmin by inhibiting the activity of tPA, significantly reducing the risk of hemorrhagic complications and ensuring the stability and safety of the circulatory system. On the other hand, inhibition of uPA activity cleverly diminishes the invasiveness and metastatic ability of cancer cells, establishing a strong defense against malignant tumor invasion in the body [26]. The above research indicates that developing inhibitors capable of modulating serine protease activity has become an effective strategy for treating clinical conditions triggered by these enzymes.
Given the critical role that serine proteases play in clinical diagnosis and treatment, numerous clinical trials in recent years have revealed the effectiveness of serine protease inhibitors in treating various clinical conditions caused by these enzymes. These inhibitors open new avenues for the treatment of related diseases by precisely inhibiting the activity or expression of serine proteases. This review aims to briefly outline the mechanisms of action of serine protease inhibitors, describe the current state of research and development, and categorize the serine protease inhibitors that have entered clinical trials or treatment. It also summarizes the persistent challenges faced during the development of serine protease inhibitors and proposes strategies and solutions to address these issues.

2. Mechanisms and Classification of Serine Protease Inhibitors

The goal of developing serine protease inhibitors is to prevent and treat certain diseases by modulating their activity or expression levels. Classical serine protease inhibitors interact with serine proteases through exposed loops that possess specific scaffold conformations. The binding conformation of these inhibitors to serine proteases is referred to as the “typical” conformation, and the mechanism based on this conformation is known as the standard or Laskowski mechanism (Figure 3A) [27,28]. In this mechanism, the inhibitor binds to the serine protease like a substrate and is slowly hydrolyzed by the protease. However, the hydrolysis products do not release from the active site, and the inhibitor’s hydrolysis products remain bound to the protease’s active site; the peptide bond is reformed, causing a decrease in or even complete loss of the protease’s catalytic activity [29,30]. It is important to note that the classical mechanism is not the only mode of inhibition: non-canonical inhibitors are a class of reversible serine protease inhibitors that block the active site spatially. These inhibitors prevent other substrates from entering the active site upon binding but do not lead to any peptide bond hydrolysis (Figure 3B) [31,32,33]. Additionally, we reported a mechanism through which the catalytic activity of serine proteases is regulated by targeting the autolytic loop within the protease’s catalytic domain. The autolytic loop is a component of the activation domain and typically undergoes significant conformational changes during serine protease activation. Inhibitors that target the autolytic loop can disrupt not only its conformation but also the conformations of other components crucial for the catalytic activity of serine proteases, thereby interfering with their activity. This indicates that targeting the non-catalytic domains of serine proteases represents a potential mechanism for protease inhibition (Figure 4) [34]. Although some small-molecule inhibitors mimic peptide inhibitors to suppress the activity of serine proteases through the Laskowski mechanism or non-standard pathways, there exists another class of small-molecule inhibitors that exhibit a more direct mode of action. They form tight binding with the key catalytic active site Ser 195 of serine proteases or the binding sites involved in protein–protein interactions, which can be either covalent or non-covalent. By directly competing for the occupancy of the active site with the substrate, inducing changes in protein structure, or obstructing the necessary transitions in protein conformation and configuration, these inhibitors effectively inhibit the catalytic activity of proteases [35,36]. Currently, in addition to the traditional approach of directly inhibiting protease activity, there exists a more refined inhibitory mechanism: nucleotide inhibitors demonstrate their efficacy through a highly specific and precise mode of action. They can selectively block or inhibit the expression process of specific proteases while cleverly avoiding unnecessary inhibition of other enzyme activities. Carefully designed nucleotide inhibitors can accurately target and bind to the mRNA of specific proteases, affecting the normal translation of these proteases [37]. More importantly, nucleotide inhibitors regulate the expression levels of the target proteases in a precise and targeted manner, while avoiding interference with a broad range of enzyme activities.

3. Different Types of Serine Protease Inhibitors

Serine protease inhibitors hold significant applications and research value in the fields of biology and medicine. Over decades of development, serine protease inhibitors can be broadly categorized into the following types.

3.1. Small-Molecule Inhibitors

Small-molecule inhibitors of serine protease are advantageous compared to other types of inhibitors due to their low production costs, lack of immunogenicity, long shelf life, and high formulation flexibility. When small molecules enter the active site pocket of serine proteases, they form covalent or non-covalent bonds with the catalytic active site, leading to reduced activity or inactivation of the protease.
Pradaxa® is clinically used for the prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation (NVAF) and one or more risk factors, with Dabigatran as its main component [38]. Dabigatran is a direct thrombin inhibitor that reduces intracranial bleeding in patients with atrial fibrillation and decreases major and clinically relevant non-major bleeding in acute venous thromboembolism (VTE) treatment. However, its use in patients with dynamic renal impairment due to dehydration may lead to bleeding complications, and the substructure of Dabigatran may cause glomerular hyperfiltration, reducing its efficacy [39]. Melagatran is also a direct thrombin inhibitor, while Ximelagatran is its oral prodrug, which is converted to Melagatran through enzymatic reactions. Clinical studies indicate that Melagatran has a rapid onset of action, stable absorption, and does not require routine monitoring, making it suitable for VTE prevention in patients undergoing hip or knee replacement surgery [40,41]. However, due to its potential severe hepatic toxicity, Melagatran has been withdrawn from the market [42]. Argatroban binds to thrombin reversibly and is the only small-molecule oral thrombin inhibitor approved by the FDA for treating Heparin-Induced Thrombocytopenia (HIT) in the U.S. [43,44]. AZD0837, developed as a follow-up compound to Ximelagatran, has shown effects similar to Warfarin in clinical Phase II trials for preventing ventricular-fibrillation-related strokes, without the hepatic toxicity seen with Ximelagatran [45]. The developed extended-release formulation allows for once-daily dosing. Currently, AZD0837 is in Phase III clinical trials for treating chronic ventricular fibrillation [46]. Bivalirudin, as a direct thrombin inhibitor, demonstrates better efficacy in the early intervention of acute coronary syndromes but is subject to temporary, reversible thrombin inhibition due to cleavage by thrombin after binding [47,48].
Apixaban, Betrixaban, Edoxaban, and Rivaroxaban are a class of potent reversible oral inhibitors that act directly on FXa [49]. Although these medications can prevent and treat thromboembolic diseases by inhibiting FXa activity, their clinical indications for treatment differ. Apixaban inhibits free FXa and prothrombin activity while also possessing the ability to indirectly inhibit thrombin-induced platelet aggregation [50,51,52,53]. It is currently used to reduce the risk of stroke in patients with non-valvular atrial fibrillation, for thromboprophylaxis after hip or knee replacement surgery, and to treat deep vein thrombosis or pulmonary embolism, as well as to prevent recurrent deep vein thrombosis and pulmonary embolism [54]. Betrixaban is primarily used for the prevention of VTE and stroke in patients with atrial fibrillation [55,56,57]. Edoxaban reduces thrombin production, inhibiting clot formation and is currently indicated for the prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation as well as for treating deep vein thrombosis and pulmonary embolism [58]. Rivaroxaban functions by inhibiting free FXa and shows a concentration-dependent relationship with FXa bound to prothrombin and thrombus-associated FXa [59,60]. However, it has no direct effect on platelet aggregation induced by adenosine diphosphate, collagen, or thrombin [59]. Other direct oral anticoagulants targeting FXa are also entering clinical trials, including Darexaban, Eribaxaban, Otamixaban, TAK-442, and LY517717 [61].
Milvexian (BMS-986177/JNJ0033093) is a reversible, highly efficient small-molecule inhibitor that acts on the active site of FXIa, exhibiting excellent antithrombotic effects mainly for thromboembolic prevention [62,63]. LNP023 is a reversible, specific, direct small-molecule inhibitor of Complement Factor B, preventing complement activation in the serum of C3 glomerulopathy patients and hemolysis of human PNH red blood cells by inhibiting CFB [64].
Nafamostat is a broad-spectrum small-molecule inhibitor covalently binding to the active site of serine proteases, effective against multiple conditions including acute pancreatitis and disseminated intravascular coagulation. Recent clinical results indicate that Nafamostat can prevent viral entry into human cells by inhibiting the transmembrane protease serine 2 (TMPRSS2), demonstrating high antiviral efficacy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [65]. Camostat is an oral serine protease inhibitor available for treating chronic pancreatitis and post-operative reflux esophagitis in Japan and South Korea [66,67]. Current research shows that Camostat can inhibit TMPRSS2 and prevent targeted cell infection with SARS-CoV-2, suggesting potential use in managing COVID-19 [68]. Upamostat is an oral prodrug with bioavailability that targets serine protease inhibitor WX-UK1, showing the highest activity against trypsin and closely related enzymes. Although currently under Phase II clinical trials for pancreatic and breast cancers with significant inhibitory effects on tumor metastasis [69], upamostat lacks specificity for uPA and shows similar inhibition levels against various homologous proteases [70]. Recent clinical studies suggest broad-spectrum inhibitory effects of upamostat could be applicable in treating COVID-19, gastrointestinal diseases, and malignant tumors [71].
In summary, traditional small-molecule inhibitors typically contain P1 and P1′ groups. The structures of these inhibitors include basic groups such as guanidine or ami-dine, which can be precisely and specifically recognized by the active site within the S1 pocket of serine proteases. The hydrolysis of the peptide bond occurs precisely at the junction between the P1 and P1′ groups. Crucially, the basic group in the P1 group of small-molecule inhibitors can form strong electrostatic interactions with the highly conserved Asp189 residue deeply embedded in the S1 pocket [72,73]. Additionally, the P1 group often contains aromatic groups that can form stable interactions with the S1 pocket of serine proteases. This interaction stabilizes the P1 group inserted into the S1 pocket of the serine protease, ensuring a tight binding and effective action between the inhibitor and the enzyme. Non-traditional small-molecule inhibitors, such as flavonoids like quercetin and myricetin [74], do not necessarily contain basic or weakly basic groups like amidine or guanidine in their structures. Instead, the P1 groups of non-traditional small-molecule inhibitors feature phenyl ring groups or halogen groups that replace aromatic groups to form stable interactions with the S1 pocket [75]. Despite the numerous significant advantages of small-molecule inhibitors, their inherent drawbacks, such as off-target effects and solubility issues, cannot be overlooked. These drawbacks will be discussed and analyzed in detail in the following sections (Table 1).

3.2. Nucleotide Drugs

Nucleotide-based drugs are a new class of inhibitors with a broad range of applications in clinical settings. RNA-based therapeutic agents include small interfering RNAs (siRNAs), which are short double-stranded RNA molecules that mediate mRNA degradation by binding to complementary mRNA target sequences. Antisense oligonucleotides (ASOs), unlike siRNAs, are single-stranded RNA or DNA molecules, but they also bind to complementary target mRNA sequences to block protein translation.

3.2.1. Antisense Oligonucleotides (ASOs)

Through the Watson–Crick complementary pairing principles, antisense oligonucleotides (ASOs) bind to the mRNA of targeted serine protease genes, leading to the selective degradation by endogenous nucleases or inhibiting mRNA processing and/or function solely through occupancy mechanisms [37]. This effectively restricts the expression of serine proteases. Antisense technology has a high specificity for inhibiting unique targets and can be employed to suppress the synthesis of key proteases involved in diseases [76]. Successful ASOs offer advantages such as low drug design costs, high target selectivity, minimal drug–food interactions, high patient compliance, infrequent dosing, and a low incidence of clinical adverse events [77], making them a promising medical technology; selecting an appropriate ASO allows for highly specific target suppression.
AZD8233, primarily used for treating hyperlipidemia, targets PCSK9 mRNA sequences by inhibiting the synthesis of PCSK9 protein within cells [78]. Fesomersen and IONIS-FXIRX are FXI-targeting ASOs that are complementary to human FXI mRNA sequences, significantly lowering FXI mRNA levels in the liver without increasing bleeding risk, thereby demonstrating clear antithrombotic effects [79]. However, IONIS-FXIRX requires subcutaneous injections twice a week and takes 3 to 4 weeks to bring FXI levels into the therapeutic range [80]. Fesomersen, a second-generation FXI-targeting ASO, binds partially with N-acetylgalactosamine (GalNAc3) to promote its uptake by liver cells and requires administration only once a month, reducing FXI levels into the therapeutic range within a week. Compared to the first generation, IONIS-FXIRX, Fesomersen shows better efficacy and a faster onset of action [80].

3.2.2. siRNA

siRNA is a class of non-coding small RNAs that can silence target genes. siRNA induces the cleavage of specific mRNA sequences through the principle of base complementary pairing, interfering with the gene expression of the intended protein and thereby suppressing the synthesis of the corresponding protease. siRNA can inhibit gene function without causing mutations in the organism, making it an up-and-coming gene therapy technology. Inclisiran (Leqvio®) is a siRNA that requires subcutaneous administration only once every six months, which prevents the synthesis of PCSK9 in the liver, thereby lowering circulating low-density lipoprotein cholesterol (LDL-C). Clinically, it can prevent and treat atherosclerotic cardiovascular disease (ASCVD) [81]. However, the PCSK9 siRNA causes a significant reduction in PCSK9 expression, leading to the overexpression of CD36, and long-term use may trigger cancers such as acute myeloid leukemia, rodent tumors, breast cancer, and colorectal cancer [82].

4. Biomacromolecular Inhibitors

4.1. Peptides and Peptide Mimetics

Peptides as serine protease inhibitors are widely found in both animals and plants [83,84,85,86]. These inhibitors reduce the catalytic activity of serine proteases, sometimes rendering them completely inactive by forming a tight substrate-like binding with them [27]. Peptide inhibitors include families such as Kunitz, Bowman–Birk, and Kazal [87].
Kunitz inhibitors are widely occurring serine protease inhibitors with significant therapeutic potential. These inhibitors belong to two families: one derived from animals and the other from plants. The Kunitz-type inhibitor isolated from soybeans is known as the soybean trypsin inhibitor (STI), while Kunitz-type inhibitors sourced from plants other than soybeans are referred to as Kunitz trypsin inhibitors (KTIs). Research indicates that there are considerable differences in the structure and binding modes of Kunitz inhibitors from different sources [88,89]: STIs bind to proteases like substrates and are slowly hydrolyzed by the proteases. However, the products do not release from the active site, and, while the inhibitor is being hydrolyzed, peptide bonds are reformed while still bound to the active site of the protease [29,30]. Most KTIs consist of a single polypeptide chain containing 181 amino acid residues and four cysteine residues, forming two internal disulfide bonds and a unique reactive site. The amino acid residue sequence involved in protease inhibition within Kunitz-type inhibitors is known as the protease binding loop. Almost all KTIs adopt a canonical conformation in the protease binding loop, forming a complex with the active site of serine proteases. Serine proteases hydrolyze the peptide bonds between the P1–P1′ group of KTI, and the resulting products retain a structure similar to that of the intact inhibitor, trapping the enzyme in a hydrolysis/resynthesis cycle, thereby inhibiting enzyme activity [90]. Animal-derived Kunitz-type inhibitors contain one or more Kunitz domains [91], typically consisting of 50–70 amino acids. Disulfide bonds within the structure maintain the integrity of the inhibitor [92]. The presence of disulfide bonds ensures that Kunitz inhibitors possess such binding loops on their surface [92], allowing them to tightly bind to the active sites of serine proteases in a substrate-like manner, thereby inhibiting enzyme activity. This section discusses Kunitz inhibitors sourced from animals.
Aprotinin inhibits the activity of kallikrein and also of trypsin, which is why it is often named bovine pancreatic trypsin inhibitor (BPTI) and trypsin-kallikrein inhibitor (TKI); it acts as a natural serine protease inhibitor. Although Aprotinin is not an antifibrinolytic agent, it can improve hemostatic deficiencies related to elevated plasmin activity [93], thereby being useful in reducing the risk of substantial blood loss during strokes and repeat surgeries [94]. Due to its ability to inhibit the proteolytic activation of certain viruses, including the novel coronavirus, by TMPRSS2 and its binding to the ACE2 receptors on the surface of epithelial cells, it is considered a potential drug for the prevention and treatment of acute respiratory viral infections [95,96,97].
Desmolaris, another Kunitz-type inhibitor extracted from the vampire bat Desmodus rotundus, inhibits FXIa in a slow, tight, non-competitive manner, preventing FXIa from activating FXa, which leads to a dose-dependent extension of activated partial thromboplastin time (aPTT) without significant bleeding. Additionally, Desmolaris can inhibit various serine proteases such as trypsin, α-chymotrypsin, and neutrophil-derived elastase, offering great promise as a novel anticoagulant in clinical thrombotic treatment [98].
Ir-CPI is a Kunitz-type inhibitor originating from the tick Ixodes ricinus that selectively binds to three factors during their activation, thereby inhibiting the mutual activation of FXII, prekallikrein, and FXI in human plasma while preserving thrombotic balance. It is the first known selective inhibitor of the coagulation contact pathway capable of preventing both venous and arterial thrombosis [99].
Bowman–Birk Inhibitors (BBIs) are a different class of serine protease inhibitors derived from soybeans, distinct from SKTIs, primarily acting on trypsin, chymotrypsin, and elastase. BBIs have a molecular weight of only 6–9 kDa and consist of 60–80 amino acids. They possess a symmetrical structure with two cyclic domains located within a non-peptide region connected by disulfide bonds formed between cysteine residues. These two inhibitory domains are highly exposed, making them easily accessible to proteolytic enzymes [100,101]. As a result, BBIs can independently interact with two types of serine proteases simultaneously. The number and distribution of cysteine residues in the disulfide bonds play a crucial role in maintaining the structural stability and functionality of the domains under extreme temperature and pH conditions [100,101], indicating that BBIs have promising potential for the prevention and treatment of gastric cancer in the future. The P1 group of BBI inserts into the S1 pocket of serine proteases, forming a stable enzyme-inhibitor complex. Although the therapeutic targets and mechanisms of action of BBIs remain unknown [101], current research suggests that BBIs can exert therapeutic and preventive effects against inflammation and cancer by inhibiting serine proteases [101,102,103,104].
Kazal-type serine protease inhibitors (KTSPIs) are widely found in mammals, birds, and various invertebrates, consisting of one or multiple Kazal domains. They inhibit the catalytic activity of serine proteases from the S1 family, including trypsin, chymotrypsin, thrombin, elastase, plasmin, proteinase K, and Bacillus subtilis protease A [29]. Kazal-type inhibitors are particularly suited to act as thrombin inhibitors, as there is no spatial hindrance between Trp60 (the chymotrypsin symbol for thrombin) and the inhibitor [105]. KTSPI has been shown to modulate inflammatory responses [106], innate immunity, and antimicrobial defense [91,107,108]. AcKTSPI, derived from the venom gland of Apis cerana, contains a Kazal domain and exhibits inhibitory effects on Bacillus subtilis protease A and proteinase K but shows no activity against α-chymotrypsin, trypsin, factor Xa, thrombin, tissue plasminogen activator, or elastase. Additionally, AcKTSPI demonstrates antibacterial activity against Gram-positive bacteria, suggesting its potential as an effective antimicrobial agent in the future [109]. The first known Kazal-type inhibitor with anticoagulant activity, rhodiin, originated from the insect Rhodnius prolixus and specifically inhibits thrombin [105]. Another specific thrombin inhibitor, dipetalogastin, comes from the insect Dipetalogaster maximus [110]. Furthermore, infestin 1–2, found in the intestines of the hematophagous insect Triatoma infestins, is also a specific thrombin inhibitor [111]. Similarly, infestin-4 from the kissing bug Triatoma infestins inhibits not only factor XIIa but also plasmin and factor Xa [19,111,112]. AaTl, extracted from Aedes aegypti, is another KTSPI that can non-competitively inhibit thrombin, prolonging prothrombin time, activating partial thromboplastin time, and thrombin time [113]. In addition, the endogenous protease inhibitor serine protease inhibitor Kazal type 1 (SPINK1) in KTSPI specifically inhibits the secretion of pancreatic proteases and tumor-associated proteases, preventing the premature activation of proteases and protecting against self-digestion of the pancreas or ductal system [114,115,116]. In the tumor microenvironment, it plays multiple roles that facilitate cancer progression [116,117], being functionally related to processes like cell resistance, differentiation, metastasis, inflammation, and cancer spread [118]. SPINK1 is extensively researched as a cancer biomarker [119,120]. Other endogenous inhibitors will be elaborated on in the following text.
Antipain, bacitracin A, chymostatin, and leupeptin are broad-spectrum serine protease inhibitors that inactivate serine proteases by covalently or non-covalently binding to their active sites. Recent studies have found that leupeptin can form strong interactions with key amino acids in TMPRSS2, suggesting its potential as a drug for preventing and treating COVID-19 [121,122]. Hirudin, extracted from leeches, is the most effective known natural thrombin inhibitor; however, studies indicate that the bleeding incidence associated with hirudin is significantly higher than that with heparin, thus, it is not recommended for treating acute coronary syndrome and currently has no other clinical indications [123]. Desirudin, developed from modified hirudin, is used for the prevention of venous thromboembolism and is currently undergoing multiple Phase III clinical trials [44]. Additionally, PEG-Hirudin, a mixture of natural hirudin, has shown a significant extension of plasma half-life [124]. This new combination has demonstrated the ability to inhibit arterial thrombus formation in in vitro human models [125]. Although Phase II clinical trials have been completed, this medication remains experimental without any indications or recommendations.
MK-0616 is an oral macrocyclic peptide inhibitor that binds to PCSK9, inhibiting its interaction with LDLR. MK-0616 shows good inhibitory effects and selectivity against PCSK9 [126]. Sylvestin is a peptide inhibitor that can simultaneously inhibit PKal and FXIIa, demonstrating excellent efficacy against ischemic stroke by reducing thrombosis and inflammation in vivo [127]. VX-950 (LY-570310) is a highly selective, covalent reversible, potent peptidomimetic inhibitor of hepatitis C virus (HCV) NS3/4A protease, which reduces HCV RNA levels in replicating cells in a time- and dose-dependent manner, making it a potential treatment for hepatitis C infection without significant cytotoxicity at high doses [128].
Two cyclic peptides, upain-1 (CSWRGLENHRMC) and mupain-1 (CPAYSRYLDC), isolated from a phage display peptide library, exhibit potent inhibitory effects and selectivity against human-uPA and mouse-uPA, leading to the development of uPA inhibitors with comparable efficacy and specificity to monoclonal antibodies [129].
The Serine protease inhibitor (Serpins) superfamily within peptide inhibitors comprises a range of proteins that display low sequence homology but have highly conserved core structures [87,130]. The majority demonstrate significant inhibitory effects on serine proteases. Given that Serpins possess the unique ability to be autonomously secreted by human tissues, this characteristic encourages their classification as endogenous protease inhibitors for further exploration and summarization.
Neuroserpin (NS), widely expressed in the brain, and PAI-1, produced by various cells, are classic examples of SERPINE 1. These similar extracellular serine proteases, once secreted by cells, inhibit the activity of tissue-type plasminogen activator (tPA) and block the cleavage of several substrates, including plasminogen [131]. Serpin peptidase inhibitor, clade A, member 1 (SERPINA1) is primarily expressed in the liver and lungs, with expression observed in monocytes and macrophages [132]. It is a major protein in human serum, inhibiting various serine proteases such as neutrophil elastase, trypsin, chymotrypsin, and thrombin [133,134]. SERPINA1 regulates the immune system and inflammation [134], and defects in SERPINA1 can lead to severe lung and liver diseases [135].
SERPINA3, also known as α-1-antichymotrypsin (AACT, ACT), is mainly synthesized in the liver and secreted into the bloodstream [136]. It inhibits serine proteases, such as chymotrypsins, cathepsin G, and mast-cell chymase, by forming stable complexes, which diminishes their proteolytic activity and subsequently alters the extracellular matrix (ECM) [137,138,139]. The SERPINA3 protein plays a crucial role in tumorigenesis, with elevated levels associated with poorer prognosis in certain cancers, leading to its use as a diagnostic factor for colon, breast, lung, and gastric cancers. The serine protease inhibitor encoded by the SERPINC1 gene, antithrombin III (ATIII), possesses an exposed reactive center loop that interacts with the active site of proteases to inhibit their activity [140]. ATIII is a key inhibitor of coagulation factors, with even minute changes significantly affecting thromboembolism formation [141]. Furthermore, ATIII can inhibit inflammation through both coagulation-dependent and -independent mechanisms [142,143].

4.2. Antibody

Monoclonal antibodies consist of two heavy polypeptide chains and two light polypeptide chains, capable of targeting and binding to specific proteins anywhere in the body. The antigens bound by mAbs can elicit antibody-dependent cellular cytotoxicity or complement-mediated cytotoxicity, leading to the blockade of cell membrane receptors and inhibition of intracellular signaling [144]. Sibrotuzumab is an anti-FAP antibody that accumulates at tumor sites, aiding in the clearance of tumor cells and reducing tumor size. It is used to treat metastatic FAP-positive cancers, including breast cancer, colorectal cancer, and non-small cell lung cancer [145].
Evolocumab (AMG 145) and alirocumab (REGN727/SAR236553) are monoclonal antibodies that bind to the serine protease PCSK9 and can significantly lower plasma levels of low-density lipoprotein cholesterol (LDL-C), potentially reducing cardiovascular risk [146,147]. Narsoplimab (OMS721) is a human IgG4 monoclonal antibody that binds and inhibits mannan-binding lectin-associated serine protease-2 (MASP-2), showing significant therapeutic effects in hematopoietic stem cell transplant-associated thrombotic microangiopathy (HSCT-TMA). XIsomab 3G3 (AB023) is a humanized 14E11 antibody derived from recombinant mouse FXI, which binds to a highly conserved region of the apple 2 domain of FXI. Although 14E11 can inhibit FXIIa-mediated activation of FXI, it shows no significant inhibitory effect on FXI activated by thrombin or FXIa pathways, suggesting that XIsomab 3G3 may be specifically suitable for FXIa, functioning as a monoclonal antibody that inhibits thrombosis in a dose-dependent manner without increasing bleeding time [148]. Osocimab (BAY1213790) is a durable fully human monoclonal G1 antibody targeting FXIa, binding to a specific region near the enzyme’s active site, causing significant structural changes in FXIa that reduce its binding to substrates, particularly FXI. This minimizes thrombus formation with minimal disruption to hemostasis [149], generally applicable during surgical procedures.
Based on investigations of uPA activity and inactive conformation transitions, we and collaborators developed inhibitory antibodies against huPA, mAb-112, and Fab-112. mAb-112 stabilizes the zymogen conformation of the uPA precursor and/or delays activation of the single-chain uPA precursor into its active double-chain form through steric hindrance. Fab-112 binds to the autolytic loop, causing its relocation, thereby inducing the complete conversion of activated uPA to its inactive zymogen form [150]. Additionally, recognizing features of the protease domain of muPA, we developed a set of nanobodies (Nb22) and an allosteric nanobody (Nb7) against muPA. Nb22 binds to the S1 specificity pocket of muPA in a substrate-like manner to inhibit its activity, while Nb7 can recognize both substrate-bound and unbound muPA with nearly identical affinity, indicating that Nb7 can act as a mixed inhibitor of muPA [151].

5. Challenges of Serine Protease Inhibitors in Clinical Applications

So far, serine protease inhibitors have been used clinically not only for treating diseases caused by serine proteases but also as probes in disease diagnosis. The small-molecule inhibitor FAPI-04, which inhibits FAP, shows a high affinity for FAP and exhibits excellent stability in human serum. PET/CT scan results from two patients with metastatic breast cancer revealed that after a small amount of FAPI-04 treatment, the uptake of the tracer in metastatic lesions was elevated, and pain symptoms were alleviated. Therefore, FAPI-04 holds promise as a clinical tracer for diagnostic imaging and targeted therapy of malignancies such as cancers with high levels of activated fibroblasts [152]. In terms of treatment, the endogenous inhibitor that has been studied most extensively is PAI-1, which is the primary endogenous inhibitor of tissue-type plasminogen activator (tPA) and also of urokinase (uPA). PAI-1 regulates fibrin dissolution and tumor cell invasion by inhibiting uPA and tPA. However, recent studies suggest that PAI-1 may promote tumor cell deterioration, positioning it as a critical target for novel cancer therapies [153] with the ongoing development of PAI-1 inhibitors. Furthermore, the serine protease inhibitor Kazal type 1 (SPINK1), synthesized by pancreatic acinar cells, is aberrantly overexpressed at both mRNA and protein levels in samples of oral tongue squamous cell carcinoma (OTSCC). SPINK1 promotes the proliferation, invasion, and motility of OTSCC cells while reducing apoptosis, indicating that SPINK1 could serve as a potential prognostic biomarker for OTSCC [154].
Serine protease inhibitors have shown considerable promise in disease treatment. After decades of in-depth research, numerous compounds and biological agents exhibiting effective inhibition of serine proteases have been discovered, presenting opportunities for development into clinical therapeutic drugs. Currently, various serine protease inhibitors have entered clinical trial stages (Table 2). However, very few serine protease inhibitors have transitioned into clinical therapeutic use. The numerous challenges faced during the development of serine protease inhibitors have limited their clinical application.
The catalytic active sites of serine proteases are highly conserved, with only certain differences in the amino acid residues within and around the active pocket. Some small-molecule inhibitors of serine proteases, while exhibiting high inhibitory efficacy, primarily act through covalent or non-covalent interactions with the catalytic active sites in the active pocket. This lack of precise target identification can lead to off-target effects, potentially resulting in adverse clinical events. Consequently, the high similarity of active sites among different serine proteases poses a challenge for developing highly selective inhibitors targeting these sites. Moreover, the negative charge of the protease active sites makes the development of small-molecule competitive inhibitors challenging [195,196]. Furthermore, targeting specific serine proteases does not always guarantee successful pathological outcomes. If serine proteases possess non-catalytic domains, these domains often participate in regulating catalytic activity [197,198,199].
While antibody-based serine protease inhibitors exhibit strong specificity, their mechanism of action primarily relies on protein–protein interactions, which restricts the spectrum of diseases they can effectively treat. Compared to small-molecule inhibitors, antibody inhibitors are associated with higher production costs, greater susceptibility to inactivation, a limited mode of administration, and stringent storage requirements. Furthermore, antibodies have high immunogenicity, increasing the likelihood of triggering immune responses that can lead to allergic reactions, and, in severe cases, pose life-threatening risks.
Some serine protease inhibitors also face issues of instability and short half-lives in vivo. ASOs typically utilize a phosphodiester backbone during design and synthesis, which is susceptible to nuclease activity, thereby affecting the stability and efficacy of ASOs in inhibiting serine proteases [200].
Additionally, overcoming the poor solubility of small-molecule drugs is one of the greatest challenges in clinical drug development. Small-molecule inhibitors with low solubility tend to precipitate in aqueous solutions, resulting in extremely low concentrations in water that fail to reach the inhibitory levels required against serine proteases in the human body, thus hindering optimal therapeutic effects. Since solubility is a critical factor for the oral absorption of drug formulations and a necessary condition for the drugs to penetrate cell membranes and be transported within the body, serine protease inhibitors with poor solubility typically exhibit low oral bioavailability. Consequently, these inhibitors are often developed as injectables. To improve the solubility or stability of small-molecule inhibitors, those that are generally insoluble in water can often dissolve in organic solvents such as anhydrous ethanol, acetone, or tetrahydrofuran, particularly when surfactants are added to enhance solubility. However, excessive concentrations of these organic solvents may cause irreversible alterations to the structure of serine proteases, leading to complete inactivation. Moreover, some organic solvents are metabolized very slowly within the human body, increasing the risk of bioaccumulation and resulting in solvent poisoning, which can trigger adverse reactions [201,202].
Another issue is also drawing attention. For multifunctional serine proteases, serine protease inhibitors exhibit a complex dual action: on one hand, Factor Xa and Thrombin play a crucial role in the intrinsic coagulation pathway, making the development of antithrombotic drugs targeting these two key points a hot topic in the research field [203,204]. By precisely inhibiting the activity of FXa or thrombin, unnecessary thrombus formation can be effectively curtailed, opening new avenues for the prevention and treatment of cardiovascular diseases. Although numerous preclinical trials have confirmed that some FXa inhibitors and thrombin inhibitors have achieved significant results in reducing the risk of cardiovascular events, the use of these inhibitors is also associated with an increased risk of bleeding [205,206]. This finding reveals that serine protease inhibitors, when applied to multifunctional serine proteases, demonstrate both therapeutic potential and potential safety challenges.

6. General Strategies for Overcoming Challenges in the Development of Serine Protease Inhibitors

How to address the various challenges in the development of serine protease inhibitors to ensure their widespread clinical application has long been a significant topic for consideration. The following two directions may provide ideas for the development and widespread use of serine protease inhibitors.

6.1. Structure-Based Drug Design Remains the Primary Approach for Inhibitor Development

With in-depth studies in structural biology, certain key sites on serine proteases have become references for the development of inhibitor structures. For example, ecotoin is known as a broad-spectrum serine protease inhibitor that interacts with both the active site Ser195 and the exosite of serine proteases to exert its inhibitory effects. By modifying ecotoin to enhance its interaction with serine proteases, it has been transformed into a high-affinity uPA inhibitor [207]. However, some inhibitors exhibit significant variability in their inhibitory effects on different proteins, as seen with rivaroxaban, which shows high selectivity against human FXa (Ki = 0.4 nmol/L), surpassing the selectivity for other biologically relevant serine proteases by 10,000 times [60]. The relatively conserved catalytic active site structures of serine proteases indicate that other factors may interfere with the binding of the same inhibitor to different serine proteases. We hypothesize that the S1 protease active pocket and surrounding amino acid residues may influence this effect. Therefore, attention should be paid to the S1 pocket and surrounding residues in inhibitor development, utilizing slight geometric variations in the S1 pocket and its sub-sites to achieve the development of specific serine protease inhibitors [208,209,210].
Small cyclized peptides have demonstrated significant potential in developing specific serine protease inhibitors. Compared to linear peptides, cyclized peptides possess greater conformational constraints, allowing them to engage with areas of serine proteases beyond the S1 pocket. These areas exhibit much larger variations among homologous enzymes, resulting in higher specificity for substrate and drug recognition, thus rendering cyclized peptides more selective toward their targets [211]. We have also reported that by altering certain key amino acid residues within cyclized peptides, it is possible to transform a specificity cyclized peptide inhibitor targeting one serine protease into a specificity inhibitor for another serine protease. Moreover, increasing the flexibility of cyclized peptides can enhance their inhibitory effects on targets [129].
Additionally, the relatively large and variable autolysis loops present across different serine proteases may serve as possible targets for highly specific serine protease activity modulation. Therefore, we propose that targeting autolysis loops may become a general strategy for pharmacological interventions in serine protease activity in the future [34].
Structure is also an important consideration in the development of serine protease inhibitors. Research on Kunitz-STI inhibitors has shown that there are significant sequence variations among the Kunitz-STI inhibitor class, yet these sequences maintain highly similar structural conformations, which are crucial for inhibiting the catalytic activity of serine proteases [89,212]. Therefore, merely classifying based on simple amino acid residue sequences and using that as a basis for developing serine protease inhibitors is clearly inadequate; structural comparisons may provide a better starting point for this development.
ASOs have great potential for future clinical disease treatment. The stability of ASOs can be improved in several ways: modifying the ASO backbone can enhance resistance to nuclease degradation. Thiophosphoramidate and isostere backbone modifications can replace the phosphodiester backbone, increasing ASO stability [213,214]. Phosphorothioate (PS) backbone modifications not only enhance ASOs’ resistance to nuclease degradation [215], ensuring that PS-ASOs can reach the target RNA in cells and tissues while recruiting RNase H enzymes to promote target RNA cleavage, but they also strengthen the attachment of ASOs to plasma proteins, facilitating ASO binding to cells and specific tissue uptake [216]. Modifications at the 2′-position of the sugar moiety can enhance the therapeutic effect of ASOs by promoting target binding. Further, 2′-O-methyl and 2′-O-methoxyethyl sugar modifications not only improve resistance to nucleases but also reduce nonspecific protein binding and lower the toxicity of ASOs [217]. Locked nucleic acids (LNAs) are another type of sugar modification; LNA-ASOs increase the binding capability of ASOs to target RNA and nuclease resistance [218], although they also enhance ASO toxicity [219]. Recent studies have shown that several LNA analogs have been synthesized, demonstrating improved activity and toxicity profiles in animal models [220,221]. However, modified ASOs have exhibited dose-dependent transient and mild to moderate toxicity, with the most common toxicities being coagulation cascade activation and increased liver enzymes observed when the plasma concentrations of PS-ASOs are high [222,223]. Current research indicates that second- and third-generation chemically modified ASOs do not have significant side effects in human patients irrespective of dose, making clinical application of ASOs in humans feasible [224]. Therefore, reducing dose-dependent toxicity or developing safer ASOs has become a pressing challenge to overcome in clinical treatments.

6.2. Drug Delivery Systems Enhance the Clinical Utilization of Poorly Soluble Inhibitors and Broad-Spectrum Inhibitors

The plasma concentrations of low-water-soluble small-molecule inhibitors of serine protease can be increased using various strategies. The primary approach typically involves chemical methods, particularly structure optimization, to enhance hydrophilic groups within the small-molecule structure, thereby improving solubility. Beyond structural optimization, solubility can also be increased through methods such as the complexation of active molecules, the use of emulsions, micelles, microemulsions, cosolvents, polymer micelle preparation, particle size reduction techniques, pharmaceutical salts, prodrugs, solid-state substitution techniques, soft gel technology, drug nanocrystals, solid dispersion methods, crystal engineering, and nanomorphology techniques [225].
Using biological methods to enhance the therapeutic effects of poorly soluble drugs is currently a research hotspot. Insoluble drugs can be covalently or non-covalently incorporated into carriers, which can then target the specific protein of interest through targeting agents [226]. The most common biological method for improving the treatment efficacy of insoluble drugs is encapsulation. In this process, the poorly soluble drug is embedded in a carrier, which then targets the relevant serine protease, releasing the drug near the target site to achieve therapeutic effects. The carrier is subsequently metabolized and eliminated by the liver. Common carriers used in clinical studies include microcapsules, microspheres, nanoparticles, liposomes, proteins (such as human serum albumin, ferritin, low-density lipoprotein, chaperone proteins, and fibrillar proteins), as well as extracellular vehicles (EVs) [227]. Protease-activated prodrugs (PAPs) are carriers that precisely target tumor-associated proteases. The peptide precursor binds to the inhibitor, rendering the drug temporarily ineffective. When entering the human circulatory system, the prodrug is either undetected or in an inactive state. It is only when the prodrug is cleaved at specific sites by tumor-associated endopeptidases and the residual peptide segments are removed by nonspecific exopeptidases that the active agent is fully enabled to exert its inhibitory effects [228].
Common serine protease targets include uPA [229,230], plasmin [231], and FXa [232]. Pharmaceutical carriers offer several advantages in clinical disease treatment; they can deliver not only poorly soluble small-molecule drugs but also siRNA, miRNA, ASOs, proteins, small-molecule drugs, nanoparticles, and CRISPR/Cas9 gene-editing systems [227,233,234,235]. However, common drug carriers have limited encapsulation capacity; as the size of the drug increases, the amount that can be encapsulated decreases. Therefore, developing carriers with larger encapsulation capacities or focusing on smaller molecular weight drugs presents ongoing challenges for serine protease inhibitor delivery systems in clinical applications.
Dual-targeted combination therapy is a common treatment approach in clinical practice. While it is known that certain serine proteases are key targets for treating specific clinical indications, current research on individual proteins does not always guarantee a complete cure. Furthermore, specific inhibitors that can be used in clinical treatments targeting these key points are limited. Therefore, dual-targeted combination therapy aims to achieve precise treatment of diseases by inhibiting important serine proteases involved in disease progression. This approach not only enhances the efficacy of treatment but also reduces the toxicity risks associated with each drug, thereby minimizing the occurrence of adverse clinical events. For instance, in vitro cell experiments have shown that combinations of the three drugs Omipalisib, Remdesivir, and Tipifarnib display strong synergistic effects in inhibiting Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This drug combination has improved antiviral efficacy against SARS-CoV-2 infections while simultaneously lowering the toxicity risks of each medication [236].
Improving the specificity of serine protease inhibitors can also be achieved through the emerging and rapidly developing approach of Antibody–Drug Conjugates (ADCs). An ADC consists of three components: a monoclonal antibody, a linker, and a cytotoxic drug. By combining the precise targeting capability of monoclonal antibodies with the cancer-killing potential of cytotoxic agents, ADCs can selectively kill cancer cells [237]. The cytotoxic agent is covalently attached to the monoclonal antibody via an appropriate linker, resulting in targeted delivery of the cytotoxic agent to the target cells, which reduces toxicity compared to using the cytotoxic agent alone [238]. Compared to other treatment options, ADCs exhibit better efficacy and longer serum half-lives, reduced immunogenicity, and improved specificity toward cancer cells. Currently, there are 11 FDA-approved ADCs for tumor indications, with hundreds undergoing advanced clinical evaluations and many more in preclinical development [239]. One of the biggest challenges in ADC development is selecting the appropriate linker that effectively conjugates the cytotoxic payload to the monoclonal antibody. The chemical properties of the linker influence various characteristics of the ADC, including toxicity, specificity, stability, and efficacy [238]. uPAR has been shown to be a molecular target for ADC therapy in some cancers [240], and it is believed that ADCs may become widely used in the treatment of diseases caused by serine proteases in the near future.
We developed a drug carrier based on human serum albumin (HSA) that is fused with a targeting agent known as the Amino-Terminal Fragment (ATF) of uPA. This fusion protein, named ATF-HSA, can bind to the tumor surface marker uPAR. By loading chemotherapy drugs and photosensitizers into this carrier, we obtained a relatively stable macromolecular complex. Compared to free drugs, this macromolecular complex showed more precise targeting in vivo, significantly improved therapeutic effects, and markedly reduced side effects [241,242,243,244]. This indicates that such HSA packaging strategies targeting tumors could also be applied to treat other disease targets. The fusion protein ATF-HSA not only serves as a therapeutic drug delivery carrier but also has been demonstrated to function as an imaging agent delivery carrier, showing specificity for tumors expressing uPAR [241]. However, preclinical studies have found that the human uPAR targeting agent typically does not bind to mouse uPAR, indicating a species-specific limitation [244]. Recently, we developed a novel fusion protein (uPARTC) that can bind to both mouse and human uPAR. The macromolecular complex formed by embedding drugs into uPARTC exhibited effective proliferation inhibition in both mouse and human uPAR-overexpressing cells, as well as potent antitumor effects in animal studies. This suggests that uPARTC is a promising tumor-targeting drug carrier capable of overcoming the challenge of species-specificity in uPAR-targeted drugs and can be used to load other cytotoxic compounds [226]. In the future, modifications of fusion proteins based on uPARTC show immense potential for enhancing the specificity of inhibitors and precise targeting for clinical treatment indications, paving the way for advancements in precision medicine.

7. Conclusions

Serine proteases play an indispensable role in human life activities, widely participating in and profoundly influencing numerous metabolic pathways. As a result, they have become important targets for clinical diagnosis and treatment of various diseases. Given their significant status, the development of serine protease inhibitors has demonstrated extraordinary value and potential in the medical field, aiming to precisely intervene and treat clinical conditions caused by abnormal activities of these enzymes. In recent years, the field of serine protease inhibitors has experienced rapid development, with increasingly in-depth theoretical research and several inhibitors successfully entering clinical application stages, marking significant progress in the translation of research outcomes into clinical practice. This review thoroughly analyzes the mechanisms of action of serine protease inhibitors and systematically summarizes the types of inhibitors that have entered clinical practice. At the same time, we acknowledge and summarize the various challenges faced in the development of serine protease inhibitors, including but not limited to issues of inhibitor specificity, efficiency enhancement, and long-term safety. In response to these challenges, we further explore feasible solutions and strategies, aiming to provide directional guidance for future research.

Author Contributions

L.J. conceived the manuscript; Y.W. wrote the initial version of the manuscript and generated the figures. L.J., Y.W. and M.H. contributed to manuscript revision and finalization. All authors have read and agreed to the published version of the manuscript.

Funding

This study is financially supported by grants from the National Natural Science Foundation of China (32370990, 82070142, and 22077016), the National Key R&D Program of China (2023YFE0118400), the Natural Science Foundation of Fujian Province (2023Y4016), the Key Project of Science and Technology Innovation of Fujian Province (2021G02004), and the National Key Clinical Specialty Discipline Construction Program and Clinical Research Center for hematological malignancies of Fujian province (2020Y2006).

Conflicts of Interest

The authors declare no competing financial interests.

Abbreviations

ACS: acute coronary syndrome; AACT/ACT, α-1-antichymotrypsin; AF, atrial fibrillation; ADCs, Antibody–Drug Conjugates; AKI, acute kidney injury; aPTT, activated partial thromboplastin time; ASCVD, atherosclerotic cardiovascular disease; Asp, amino acids aspartate; ASO, antisense oligonucleotide; ATIII, antithrombin III; ATF, Amino-Terminal Fragment; BBI, Bowman–Birk inhibitor; BPTI: bovine pancreatic trypsin inhibitor; CAM, drug–drug co-amorphous system; CD36, cluster of differentiation 36; CFB, Complement Factor B; DTV, deep vein thrombosis; ECM, extracellular matrix; EVs, extracellular vehicles; FAP, fibroblast activation protein; FAPI, Fibroblast activation protein inhibitor; FIIa, Activated Coagulation Factor II; FVIIa, Activated Coagulation Factor VII; FIXa, Activated Coagulation Factor IX; FXa, Activated Coagulation Factor X; FXIa, Activated Coagulation Factor XI; FAP, Fibroblast activation protein-alpha; GalNAc3, N-acetylgalactosamine; HCV, hepatitis C virus; His, amino acids histidine; HIT, heparin-induced thrombocytopenia; HSA, human serum albumin; HSCT-TMA, hematopoietic stem cell transplant-associated thrombotic microangiopathy; IgAN, immunoglobulin A nephropathy; KTI, Kunitz trypsin inhibitor; KTSPI, Kazal-type serine protease inhibitor; LDL-C, low-density lipoprotein cholesterol; LDLR, low-density lipoprotein receptor; LNA, locked nucleic acids; mRNA, message RNA; mAb, monoclonal antibody; NS, neuroserpin; NVAF, non-valvular atrial fibrillation; OTSCC, oral tongue squamous cell carcinoma; PAI-1, plasminogen activator inhibitor 1; PAPs, protease-activated prodrugs; PCI, percutaneous coronary intervention; PCSK9, proprotein convertase subtilisin/kexin type 9; PE, pulmonary embolism; PET/CT, positron emission tomography/computerized tomography; PNH, paroxysmal nocturnal hemoglobinuria; PS, phosphorothioate; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; Serpins, serine protease inhibitors; SERPINA1, serpin peptidase inhibitor, clade A, member 1; SERPINA3, serine protease inhibitor A3; SERPINC1, serpin peptidase inhibitor, clade C, member 1; siRNA, small interfering RNA; Ser, amino acids serine; STI, soybean trypsin inhibitor; SPINK1, serine protease inhibitor Kazal type 1; TKI, trypsin-kallikrein inhibitor; TMPRSS2, transmembrane serine protease 2; tPA, tissue-type plasminogen activator; uPA, urokinase-type plasminogen activator; uPAR, urokinase-type plasminogen activator receptor; VTE, venous thromboembolism.

References

  1. Rawlings, N.D.; Barrett, A.J. MEROPS: The peptidase database. Nucleic Acids Res. 2000, 28, 323–325. [Google Scholar] [CrossRef] [PubMed]
  2. Hedstrom, L. Serine protease mechanism and specificity. Chem. Rev. 2002, 102, 4501–4524. [Google Scholar] [CrossRef] [PubMed]
  3. Shen, H.B.; Chou, K.C. Identification of proteases and their types. Anal. Biochem. 2009, 385, 153–160. [Google Scholar] [CrossRef] [PubMed]
  4. Burchacka, E.; Pięta, P.; Łupicka-Słowik, A. Recent advances in fungal serine protease inhibitors. Biomed. Pharmacother. 2022, 146, 112523. [Google Scholar] [CrossRef]
  5. Soreide, K.; Janssen, E.A.; Körner, H.; Baak, J.P. Trypsin in colorectal cancer: Molecular biological mechanisms of proliferation, invasion, and metastasis. J. Pathol. 2006, 209, 147–156. [Google Scholar] [CrossRef]
  6. Andriulli, A.; Masoero, G.; Felder, M.; Vantini, I.; Petrillo, M.; Cavallini, G.; Bianchi Porro, G.; Dobrilla, G.; Verme, G. Circulating trypsin-like immunoreactivity in chronic pancreatitis. Dig. Dis. Sci. 1981, 26, 532–537. [Google Scholar] [CrossRef]
  7. Yoshida, N.; Yoshikawa, T. Basic and translational research on proteinase-activated receptors: Implication of proteinase/proteinase-activated receptor in gastrointestinal inflammation. J. Pharmacol. Sci. 2008, 108, 415–421. [Google Scholar] [CrossRef]
  8. Keragala, C.B.; Medcalf, R.L. Plasminogen: An enigmatic zymogen. Blood 2021, 137, 2881–2889. [Google Scholar] [CrossRef]
  9. Ali, A.E.; Becker, R.C. The foundation for investigating factor XI as a target for inhibition in human cardiovascular disease. J. Thromb. Thrombolysis 2024, 1–14, online ahead of print. [Google Scholar] [CrossRef]
  10. Dunkelberger, J.R.; Song, W.C. Complement and its role in innate and adaptive immune responses. Cell Res. 2010, 20, 34–50. [Google Scholar] [CrossRef]
  11. Degn, S.E.; Thiel, S. Humoral pattern recognition and the complement system. Scand. J. Immunol. 2013, 78, 181–193. [Google Scholar] [CrossRef]
  12. Carroll, M.C. Complement and humoral immunity. Vaccine 2008, 26 (Suppl. 8), I28–I33. [Google Scholar] [CrossRef] [PubMed]
  13. Noris, M.; Remuzzi, G. Overview of complement activation and regulation. Semin. Nephrol. 2013, 33, 479–492. [Google Scholar] [CrossRef] [PubMed]
  14. Lucas, J.M.; Heinlein, C.; Kim, T.; Hernandez, S.A.; Malik, M.S.; True, L.D.; Morrissey, C.; Corey, E.; Montgomery, B.; Mostaghel, E.; et al. The androgen-regulated protease TMPRSS2 activates a proteolytic cascade involving components of the tumor microenvironment and promotes prostate cancer metastasis. Cancer Discov. 2014, 4, 1310–1325. [Google Scholar] [CrossRef] [PubMed]
  15. Ko, C.J.; Huang, C.C.; Lin, H.Y.; Juan, C.P.; Lan, S.W.; Shyu, H.Y.; Wu, S.R.; Hsiao, P.W.; Huang, H.P.; Shun, C.T.; et al. Androgen-Induced TMPRSS2 Activates Matriptase and Promotes Extracellular Matrix Degradation, Prostate Cancer Cell Invasion, Tumor Growth, and Metastasis. Cancer Res. 2015, 75, 2949–2960. [Google Scholar] [CrossRef]
  16. Cheng, J.; Zhou, J.; Fu, S.; Fu, J.; Zhou, B.; Chen, H.; Fu, J.; Wei, C. Prostate adenocarcinoma and COVID-19: The possible impacts of TMPRSS2 expressions in susceptibility to SARS-CoV-2. J. Cell Mol. Med. 2021, 25, 4157–4165. [Google Scholar] [CrossRef]
  17. Sharony, R.; Yu, P.J.; Park, J.; Galloway, A.C.; Mignatti, P.; Pintucci, G. Protein targets of inflammatory serine proteases and cardiovascular disease. J. Inflamm. 2010, 7, 45. [Google Scholar] [CrossRef]
  18. Heutinck, K.M.; ten Berge, I.J.; Hack, C.E.; Hamann, J.; Rowshani, A.T. Serine proteases of the human immune system in health and disease. Mol. Immunol. 2010, 47, 1943–1955. [Google Scholar] [CrossRef]
  19. Meyer-Hoffert, U. Reddish, scaly, and itchy: How proteases and their inhibitors contribute to inflammatory skin diseases. Arch. Immunol. Ther. Exp. 2009, 57, 345–354. [Google Scholar] [CrossRef]
  20. Moldoveanu, B.; Otmishi, P.; Jani, P.; Walker, J.; Sarmiento, X.; Guardiola, J.; Saad, M.; Yu, J. Inflammatory mechanisms in the lung. J. Inflamm. Res. 2009, 2, 1–11. [Google Scholar]
  21. Soualmia, F.; El Amri, C. Serine protease inhibitors to treat inflammation: A patent review (2011–2016). Expert. Opin. Ther. Pat. 2018, 28, 93–110. [Google Scholar] [CrossRef]
  22. Micalet, A.; Tappouni, L.J.; Peszko, K.; Karagianni, D.; Lam, A.; Counsell, J.R.; Quezada, S.A.; Moeendarbary, E.; Cheema, U. Urokinase-type plasminogen activator (uPA) regulates invasion and matrix remodelling in colorectal cancer. Matrix Biol. Plus 2023, 19–20, 100137. [Google Scholar] [CrossRef] [PubMed]
  23. Zhou, Y.; Vedantham, P.; Lu, K.; Agudelo, J.; Carrion, R., Jr.; Nunneley, J.W.; Barnard, D.; Pöhlmann, S.; McKerrow, J.H.; Renslo, A.R.; et al. Protease inhibitors targeting coronavirus and filovirus entry. Antiviral Res. 2015, 116, 76–84. [Google Scholar] [CrossRef]
  24. Mykytyn, A.Z.; Breugem, T.I.; Riesebosch, S.; Schipper, D.; van den Doel, P.B.; Rottier, R.J.; Lamers, M.M.; Haagmans, B.L. SARS-CoV-2 entry into human airway organoids is serine protease-mediated and facilitated by the multibasic cleavage site. Elife 2021, 10, e64508. [Google Scholar] [CrossRef]
  25. Wang, Y.; Luo, W.; Reiser, G. Trypsin and trypsin-like proteases in the brain: Proteolysis and cellular functions. Cell Mol. Life Sci. 2008, 65, 237–252. [Google Scholar] [CrossRef]
  26. Kruithof, E.K. Plasminogen activator inhibitors—A review. Enzyme 1988, 40, 113–121. [Google Scholar] [CrossRef]
  27. Bode, W.; Huber, R. Natural protein proteinase inhibitors and their interaction with proteinases. Eur. J. Biochem. 1992, 204, 433–451. [Google Scholar] [CrossRef]
  28. Krowarsch, D.; Cierpicki, T.; Jelen, F.; Otlewski, J. Canonical protein inhibitors of serine proteases. Cell Mol. Life Sci. 2003, 60, 2427–2444. [Google Scholar] [CrossRef]
  29. Laskowski, M., Jr.; Kato, I. Protein inhibitors of proteinases. Annu. Rev. Biochem. 1980, 49, 593–626. [Google Scholar] [CrossRef]
  30. Zakharova, E.; Horvath, M.P.; Goldenberg, D.P. Structure of a serine protease poised to resynthesize a peptide bond. Proc. Natl. Acad. Sci. USA 2009, 106, 11034–11039. [Google Scholar] [CrossRef]
  31. Micheelsen, P.O.; Vévodová, J.; De Maria, L.; Ostergaard, P.R.; Friis, E.P.; Wilson, K.; Skjøt, M. Structural and mutational analyses of the interaction between the barley alpha-amylase/subtilisin inhibitor and the subtilisin savinase reveal a novel mode of inhibition. J. Mol. Biol. 2008, 380, 681–690. [Google Scholar] [CrossRef] [PubMed]
  32. Richardson, J.L.; Fuentes-Prior, P.; Sadler, J.E.; Huber, R.; Bode, W. Characterization of the residues involved in the human alpha-thrombin-haemadin complex: An exosite II-binding inhibitor. Biochemistry 2002, 41, 2535–2542. [Google Scholar] [CrossRef] [PubMed]
  33. Rydel, T.J.; Tulinsky, A.; Bode, W.; Huber, R. Refined structure of the hirudin-thrombin complex. J. Mol. Biol. 1991, 221, 583–601. [Google Scholar] [CrossRef]
  34. Jiang, L.; Yuan, C.; Huang, M. A general strategy to inhibit serine protease by targeting its autolysis loop. FASEB J. 2021, 35, e21259. [Google Scholar] [CrossRef]
  35. Redondo, S.; Martínez, M.P.; Ramajo, M.; Navarro-Dorado, J.; Barez, A.; Tejerina, T. Pharmacological basis and clinical evidence of dabigatran therapy. J. Hematol. Oncol. 2011, 4, 53. [Google Scholar] [CrossRef]
  36. Deftereos, S.; Anatoliotakis, N.; Giannopoulos, G.; Kaoukis, A.; Mavri, M.; Pyrgakis, V.; Stefanadis, C. Novel direct factor IIa and Xa inhibitors: Mechanisms of action and preclinical studies. Curr. Clin. Pharmacol. 2012, 7, 149–165. [Google Scholar] [CrossRef]
  37. Bennett, C.F.; Swayze, E.E. RNA targeting therapeutics: Molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 259–293. [Google Scholar] [CrossRef]
  38. Blair, H.A.; Keating, G.M. Dabigatran Etexilate: A Review in Nonvalvular Atrial Fibrillation. Drugs 2017, 77, 331–344. [Google Scholar] [CrossRef]
  39. Antonijevic, N.M.; Zivkovic, I.D.; Jovanovic, L.M.; Matic, D.M.; Kocica, M.J.; Mrdovic, I.B.; Kanjuh, V.I.; Culafic, M.D. Dabigatran—Metabolism, Pharmacologic Properties and Drug Interactions. Curr. Drug Metab. 2017, 18, 622–635. [Google Scholar] [CrossRef]
  40. Evans, H.C.; Perry, C.M.; Faulds, D. Ximelagatran/Melagatran: A review of its use in the prevention of venous thromboembolism in orthopaedic surgery. Drugs 2004, 64, 649–678. [Google Scholar] [CrossRef]
  41. Brighton, T.A. The direct thrombin inhibitor melagatran/ximelagatran. Med. J. Aust. 2004, 181, 432–437. [Google Scholar] [CrossRef] [PubMed]
  42. Mohapatra, R.; Tran, M.; Gore, J.M.; Spencer, F.A. A review of the oral direct thrombin inhibitor ximelagatran: Not yet the end of the warfarin era. Am. Heart J. 2005, 150, 19–26. [Google Scholar] [CrossRef]
  43. Berry, C.N.; Girardot, C.; Lecoffre, C.; Lunven, C. Effects of the synthetic thrombin inhibitor argatroban on fibrin- or clot-incorporated thrombin: Comparison with heparin and recombinant Hirudin. Thromb. Haemost. 1994, 72, 381–386. [Google Scholar] [CrossRef]
  44. O’Brien, P.J.; Mureebe, L. Direct thrombin inhibitors. J. Cardiovasc. Pharmacol. Ther. 2012, 17, 5–11. [Google Scholar] [CrossRef] [PubMed]
  45. Lip, G.Y.; Rasmussen, L.H.; Olsson, S.B.; Jensen, E.C.; Persson, A.L.; Eriksson, U.; Wåhlander, K.F. Oral direct thrombin inhibitor AZD0837 for the prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation: A randomized dose-guiding, safety, and tolerability study of four doses of AZD0837 vs. vitamin K antagonists. Eur. Heart J. 2009, 30, 2897–2907. [Google Scholar] [CrossRef]
  46. Squizzato, A.; Dentali, F.; Steidl, L.; Ageno, W. New direct thrombin inhibitors. Intern. Emerg. Med. 2009, 4, 479–484. [Google Scholar] [CrossRef] [PubMed]
  47. Stone, G.W.; Witzenbichler, B.; Guagliumi, G.; Peruga, J.Z.; Brodie, B.R.; Dudek, D.; Kornowski, R.; Hartmann, F.; Gersh, B.J.; Pocock, S.J.; et al. Bivalirudin during primary PCI in acute myocardial infarction. N. Engl. J. Med. 2008, 358, 2218–2230. [Google Scholar] [CrossRef]
  48. Parry, M.A.; Maraganore, J.M.; Stone, S.R. Kinetic mechanism for the interaction of Hirulog with thrombin. Biochemistry 1994, 33, 14807–14814. [Google Scholar] [CrossRef]
  49. Siddiqui, F.; Hoppensteadt, D.; Jeske, W.; Iqbal, O.; Tafur, A.; Fareed, J. Factor Xa Inhibitory Profile of Apixaban, Betrixaban, Edoxaban, and Rivaroxaban Does Not Fully Reflect Their Biologic Spectrum. Clin. Appl. Thromb. Hemost. 2019, 25, 1076029619847524. [Google Scholar] [CrossRef]
  50. Mandernach, M.W.; Beyth, R.J.; Rajasekhar, A. Apixaban for the prophylaxis and treatment of deep vein thrombosis and pulmonary embolism: An evidence-based review. Ther. Clin. Risk Manag. 2015, 11, 1273–1282. [Google Scholar] [CrossRef]
  51. Mekaj, Y.H.; Mekaj, A.Y.; Duci, S.B.; Miftari, E.I. New oral anticoagulants: Their advantages and disadvantages compared with vitamin K antagonists in the prevention and treatment of patients with thromboembolic events. Ther. Clin. Risk Manag. 2015, 11, 967–977. [Google Scholar] [CrossRef] [PubMed]
  52. Bauer, K.A. Recent progress in anticoagulant therapy: Oral direct inhibitors of thrombin and factor Xa. J. Thromb. Haemost. 2011, 9 (Suppl. 1), 12–19. [Google Scholar] [CrossRef] [PubMed]
  53. Bonar, R.; Favaloro, E.J.; Mohammed, S.; Ahuja, M.; Pasalic, L.; Sioufi, J.; Marsden, K. The effect of the direct factor Xa inhibitors apixaban and rivaroxaban on haemostasis tests: A comprehensive assessment using in vitro and ex vivo samples. Pathology 2016, 48, 60–71. [Google Scholar] [CrossRef]
  54. Byon, W.; Garonzik, S.; Boyd, R.A.; Frost, C.E. Apixaban: A Clinical Pharmacokinetic and Pharmacodynamic Review. Clin. Pharmacokinet. 2019, 58, 1265–1279. [Google Scholar] [CrossRef]
  55. Chan, N.C.; Bhagirath, V.; Eikelboom, J.W. Profile of betrixaban and its potential in the prevention and treatment of venous thromboembolism. Vasc. Health Risk Manag. 2015, 11, 343–351. [Google Scholar] [CrossRef]
  56. Connolly, S.J.; Eikelboom, J.; Dorian, P.; Hohnloser, S.H.; Gretler, D.D.; Sinha, U.; Ezekowitz, M.D. Betrixaban compared with warfarin in patients with atrial fibrillation: Results of a phase 2, randomized, dose-ranging study (Explore-Xa). Eur. Heart J. 2013, 34, 1498–1505. [Google Scholar] [CrossRef]
  57. Feng, W.; Wu, K.; Liu, Z.; Kong, G.; Deng, Z.; Chen, S.; Wu, Y.; Chen, M.; Liu, S.; Wang, H. Oral direct factor Xa inhibitor versus enoxaparin for thromboprophylaxis after hip or knee arthroplasty: Systemic review, traditional meta-analysis, dose-response meta-analysis and network meta-analysis. Thromb. Res. 2015, 136, 1133–1144. [Google Scholar] [CrossRef]
  58. Srinivasan, S.; Ajmal, M.; Pecci, C.; Lassar, T. Edoxaban in cardiovascular disease management: Review. Br. J. Clin. Pharmacol. 2022, 88, 535–540. [Google Scholar] [CrossRef]
  59. Perzborn, E.; Roehrig, S.; Straub, A.; Kubitza, D.; Mueck, W.; Laux, V. Rivaroxaban: A new oral factor Xa inhibitor. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 376–381. [Google Scholar] [CrossRef]
  60. Perzborn, E.; Strassburger, J.; Wilmen, A.; Pohlmann, J.; Roehrig, S.; Schlemmer, K.H.; Straub, A. In vitro and in vivo studies of the novel antithrombotic agent BAY 59-7939—An oral, direct Factor Xa inhibitor. J. Thromb. Haemost. 2005, 3, 514–521. [Google Scholar] [CrossRef]
  61. Bondarenko, M.; Curti, C.; Montana, M.; Rathelot, P.; Vanelle, P. Efficacy and toxicity of factor Xa inhibitors. J. Pharm. Pharm. Sci. 2013, 16, 74–88. [Google Scholar] [CrossRef] [PubMed]
  62. Birchansky, J.; Frishman, W.H. Milvexian: A Focus on a New Oral Anticoagulant that Targets Factor XIa for Thromboembolism Prophylaxis. Cardiol. Rev. 2024, online ahead of print. [CrossRef]
  63. Perera, V.; Wang, Z.; Luettgen, J.; Li, D.; DeSouza, M.; Cerra, M.; Seiffert, D. First-in-human study of milvexian, an oral, direct, small molecule factor XIa inhibitor. Clin. Transl. Sci. 2022, 15, 330–342. [Google Scholar] [CrossRef]
  64. Schubart, A.; Anderson, K.; Mainolfi, N.; Sellner, H.; Ehara, T.; Adams, C.M.; Mac Sweeney, A.; Liao, S.M.; Crowley, M.; Littlewood-Evans, A.; et al. Small-molecule factor B inhibitor for the treatment of complement-mediated diseases. Proc. Natl. Acad. Sci. USA 2019, 116, 7926–7931. [Google Scholar] [CrossRef]
  65. Hernández-Mitre, M.P.; Tong, S.Y.C.; Denholm, J.T.; Dore, G.J.; Bowen, A.C.; Lewin, S.R.; Venkatesh, B.; Hills, T.E.; McQuilten, Z.; Paterson, D.L.; et al. Nafamostat Mesylate for Treatment of COVID-19 in Hospitalised Patients: A Structured, Narrative Review. Clin. Pharmacokinet. 2022, 61, 1331–1343. [Google Scholar] [CrossRef]
  66. Ohshio, G.; Saluja, A.K.; Leli, U.; Sengupta, A.; Steer, M.L. Esterase inhibitors prevent lysosomal enzyme redistribution in two noninvasive models of experimental pancreatitis. Gastroenterology 1989, 96, 853–859. [Google Scholar] [CrossRef] [PubMed]
  67. Talukdar, R.; Saikia, N.; Singal, D.K.; Tandon, R. Chronic pancreatitis: Evolving paradigms. Pancreatology 2006, 6, 440–449. [Google Scholar] [CrossRef]
  68. Hoffmann, M.; Hofmann-Winkler, H.; Smith, J.C.; Krüger, N.; Sørensen, L.K.; Søgaard, O.S.; Hasselstrøm, J.B.; Winkler, M.; Hempel, T.; Raich, L.; et al. Camostat mesylate inhibits SARS-CoV-2 activation by TMPRSS2-related proteases and its metabolite GBPA exerts antiviral activity. bioRxiv 2020, 65, 103255. [Google Scholar] [CrossRef]
  69. Mack, G.S.; Marshall, A. Lost in migration. Nat. Biotechnol. 2010, 28, 214–229. [Google Scholar] [CrossRef]
  70. Setyono-Han, B.; Stürzebecher, J.; Schmalix, W.A.; Muehlenweg, B.; Sieuwerts, A.M.; Timmermans, M.; Magdolen, V.; Schmitt, M.; Klijn, J.G.; Foekens, J.A. Suppression of rat breast cancer metastasis and reduction of primary tumour growth by the small synthetic urokinase inhibitor WX-UK1. Thromb. Haemost. 2005, 93, 779–786. [Google Scholar] [CrossRef]
  71. Plasse, T.F.; Fathi, R.; Fehrmann, C.; McComsey, G.A. Upamostat: A serine protease inhibitor for antiviral, gastrointestinal, and anticancer indications. Expert Opin. Investig. Drugs 2023, 32, 1095–1103. [Google Scholar] [CrossRef] [PubMed]
  72. Ekici, O.D.; Paetzel, M.; Dalbey, R.E. Unconventional serine proteases: Variations on the catalytic Ser/His/Asp triad configuration. Protein Sci. 2008, 17, 2023–2037. [Google Scholar] [CrossRef] [PubMed]
  73. Franco, F.M.; Jones, D.E.; Harris, P.K.; Han, Z.; Wildman, S.A.; Jarvis, C.M.; Janetka, J.W. Structure-based discovery of small molecule hepsin and HGFA protease inhibitors: Evaluation of potency and selectivity derived from distinct binding pockets. Bioorg. Med. Chem. 2015, 23, 2328–2343. [Google Scholar] [CrossRef] [PubMed]
  74. Xue, G.; Gong, L.; Yuan, C.; Xu, M.; Wang, X.; Jiang, L.; Huang, M. A structural mechanism of flavonoids in inhibiting serine proteases. Food Funct. 2017, 8, 2437–2443. [Google Scholar] [CrossRef]
  75. Jiang, L.; Zhang, X.; Zhou, Y.; Chen, Y.; Luo, Z.; Li, J.; Yuan, C.; Huang, M. Halogen bonding for the design of inhibitors by targeting the S1 pocket of serine proteases. RSC Adv. 2018, 8, 28189–28197. [Google Scholar] [CrossRef]
  76. Visser, M.E.; Witztum, J.L.; Stroes, E.S.; Kastelein, J.J. Antisense oligonucleotides for the treatment of dyslipidaemia. Eur. Heart J. 2012, 33, 1451–1458. [Google Scholar] [CrossRef]
  77. Lippi, G.; Harenberg, J.; Mattiuzzi, C.; Favaloro, E.J. Next generation antithrombotic therapy: Focus on antisense therapy against coagulation factor XI. Semin. Thromb. Hemost. 2015, 41, 255–262. [Google Scholar] [CrossRef]
  78. Liu, C.; Chen, J.; Chen, H.; Zhang, T.; He, D.; Luo, Q.; Chi, J.; Hong, Z.; Liao, Y.; Zhang, S.; et al. PCSK9 Inhibition: From Current Advances to Evolving Future. Cells 2022, 11, 2972. [Google Scholar] [CrossRef]
  79. Bethune, C.; Walsh, M.; Jung, B.; Yu, R.; Geary, R.S.; Bhanot, S. Pharmacokinetics and Pharmacodynamics of Ionis-FXIRx, an Antisense Inhibitor of Factor XI, in Patients with End-Stage Renal Disease on Hemodialysis. Blood 2017, 130, 1116. [Google Scholar] [CrossRef]
  80. Fredenburgh, J.C.; Weitz, J.I. News at XI: Moving beyond factor Xa inhibitors. J. Thromb. Haemost. 2023, 21, 1692–1702. [Google Scholar] [CrossRef]
  81. Frampton, J.E. Inclisiran: A Review in Hypercholesterolemia. Am. J. Cardiovasc. Drugs 2023, 23, 219–230. [Google Scholar] [CrossRef] [PubMed]
  82. Fan, F.S. Small-interfering RNA targeting proprotein convertase subtilisin/kexin type 9 might promote fatty liver disease and hepatocellular carcinoma through upregulation of CD36. Tumor. Biol. 2023, 45, 73–80. [Google Scholar] [CrossRef] [PubMed]
  83. Liu, H.; Chen, J.; Wang, X.; Yan, S.; Xu, Y.; San, M.; Tang, W.; Yang, F.; Cao, Z.; Li, W.; et al. Functional characterization of a new non-Kunitz serine protease inhibitor from the scorpion Lychas mucronatus. Int. J. Biol. Macromol. 2015, 72, 158–162. [Google Scholar] [CrossRef] [PubMed]
  84. Jouiaei, M.; Casewell, N.R.; Yanagihara, A.A.; Nouwens, A.; Cribb, B.W.; Whitehead, D.; Jackson, T.N.; Ali, S.A.; Wagstaff, S.C.; Koludarov, I.; et al. Firing the sting: Chemically induced discharge of cnidae reveals novel proteins and peptides from box jellyfish (Chironex fleckeri) venom. Toxins 2015, 7, 936–950. [Google Scholar] [CrossRef]
  85. Pomin, V.H.; Mourão, P.A. Specific sulfation and glycosylation-a structural combination for the anticoagulation of marine carbohydrates. Front. Cell. Infect. Microbiol. 2014, 4, 33. [Google Scholar] [CrossRef]
  86. Sabotič, J.; Bleuler-Martinez, S.; Renko, M.; Avanzo Caglič, P.; Kallert, S.; Štrukelj, B.; Turk, D.; Aebi, M.; Kos, J.; Künzler, M. Structural basis of trypsin inhibition and entomotoxicity of cospin, serine protease inhibitor involved in defense of Coprinopsis cinerea fruiting bodies. J. Biol. Chem. 2012, 287, 3898–3907. [Google Scholar] [CrossRef]
  87. Harish, B.S.; Uppuluri, K.B. Microbial serine protease inhibitors and their therapeutic applications. Int. J. Biol. Macromol. 2018, 107, 1373–1387. [Google Scholar] [CrossRef]
  88. Mosolov, V.V.; Valueva, T.A. Proteinase inhibitors and their function in plants: A review. Appl. Biochem. Microbiol. 2005, 41, 227–246. [Google Scholar] [CrossRef]
  89. Speranskaya, A.S.; Krinitsina, A.A.; Kudryavtseva, A.V.; Poltronieri, P.; Santino, A.; Oparina, N.Y.; Dmitriev, A.A.; Belenikin, M.S.; Guseva, M.A.; Shevelev, A.B. Impact of recombination on polymorphism of genes encoding Kunitz-type protease inhibitors in the genus Solanum. Biochimie 2012, 94, 1687–1696. [Google Scholar] [CrossRef]
  90. Cid-Gallegos, M.S.; Corzo-Ríos, L.J.; Jiménez-Martínez, C.; Sánchez-Chino, X.M. Protease Inhibitors from Plants as Therapeutic Agents- A Review. Plant Foods Hum. Nutr. 2022, 77, 20–29. [Google Scholar] [CrossRef]
  91. Kanost, M.R. Serine proteinase inhibitors in arthropod immunity. Dev. Comp. Immunol. 1999, 23, 291–301. [Google Scholar] [CrossRef]
  92. Mishra, M. Evolutionary Aspects of the Structural Convergence and Functional Diversification of Kunitz-Domain Inhibitors. J. Mol. Evol. 2020, 88, 537–548. [Google Scholar] [CrossRef] [PubMed]
  93. Royston, D. The current place of aprotinin in the management of bleeding. Anaesthesia 2015, 70 (Suppl. 1), 46-e17. [Google Scholar] [CrossRef] [PubMed]
  94. Himbert, S.; D’Alessandro, A.; Qadri, S.M.; Majcher, M.J.; Hoare, T.; Sheffield, W.P.; Nagao, M.; Nagle, J.F.; Rheinstädter, M.C. The bending rigidity of the red blood cell cytoplasmic membrane. PLoS ONE 2022, 17, e0269619. [Google Scholar] [CrossRef] [PubMed]
  95. Ivachtchenko, A.V.; Ivashchenko, A.A.; Shkil, D.O.; Ivashchenko, I.A. Aprotinin-Drug against Respiratory Diseases. Int. J. Mol. Sci. 2023, 24, 11173. [Google Scholar] [CrossRef]
  96. Padín, J.F.; Pérez-Ortiz, J.M.; Redondo-Calvo, F.J. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. Int. J. Mol. Sci. 2024, 25, 7553. [Google Scholar] [CrossRef]
  97. Padín, J.F.; Pérez-Ortiz, J.M.; Redondo-Calvo, F.J. Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions. Int. J. Mol. Sci. 2024, 25, 7209. [Google Scholar] [CrossRef]
  98. Ma, D.; Mizurini, D.M.; Assumpção, T.C.; Li, Y.; Qi, Y.; Kotsyfakis, M.; Ribeiro, J.M.; Monteiro, R.Q.; Francischetti, I.M. Desmolaris, a novel factor XIa anticoagulant from the salivary gland of the vampire bat (Desmodus rotundus) inhibits inflammation and thrombosis in vivo. Blood 2013, 122, 4094–4106. [Google Scholar] [CrossRef]
  99. Decrem, Y.; Rath, G.; Blasioli, V.; Cauchie, P.; Robert, S.; Beaufays, J.; Frère, J.M.; Feron, O.; Dogné, J.M.; Dessy, C.; et al. Ir-CPI, a coagulation contact phase inhibitor from the tick Ixodes ricinus, inhibits thrombus formation without impairing hemostasis. J. Exp. Med. 2009, 206, 2381–2395. [Google Scholar] [CrossRef]
  100. Kuhar, K.; Kansal, R.; Subrahmanyam, B.; Koundal, K.R.; Miglani, K.; Gupta, V.K. A Bowman–Birk protease inhibitor with antifeedant and antifungal activity from Dolichos biflorus. Acta Physiol. Plant. 2013, 35, 1887–1903. [Google Scholar] [CrossRef]
  101. Clemente, A.; Arques Mdel, C. Bowman-Birk inhibitors from legumes as colorectal chemopreventive agents. World J. Gastroenterol. 2014, 20, 10305–10315. [Google Scholar] [CrossRef] [PubMed]
  102. Kennedy, A.R. Cancer prevention by protease inhibitors. Prev. Med. 1993, 22, 796–811. [Google Scholar] [CrossRef] [PubMed]
  103. Kennedy, A.R.; Wan, X.S. Effects of the Bowman-Birk inhibitor on growth, invasion, and clonogenic survival of human prostate epithelial cells and prostate cancer cells. Prostate 2002, 50, 125–133. [Google Scholar] [CrossRef] [PubMed]
  104. Kennedy, A.R. The Bowman-Birk inhibitor from soybeans as an anticarcinogenic agent. Am. J. Clin. Nutr. 1998, 68, 1406s–1412s. [Google Scholar] [CrossRef] [PubMed]
  105. Friedrich, T.; Kröger, B.; Bialojan, S.; Lemaire, H.G.; Höffken, H.W.; Reuschenbach, P.; Otte, M.; Dodt, J. A Kazal-type inhibitor with thrombin specificity from Rhodnius prolixus. J. Biol. Chem. 1993, 268, 16216–16222. [Google Scholar] [CrossRef]
  106. Mägert, H.J.; Ständker, L.; Kreutzmann, P.; Zucht, H.D.; Reinecke, M.; Sommerhoff, C.P.; Fritz, H.; Forssmann, W.G. LEKTI, a novel 15-domain type of human serine proteinase inhibitor. J. Biol. Chem. 1999, 274, 21499–21502. [Google Scholar] [CrossRef]
  107. Augustin, R.; Siebert, S.; Bosch, T.C. Identification of a kazal-type serine protease inhibitor with potent anti-staphylococcal activity as part of Hydra’s innate immune system. Dev. Comp. Immunol. 2009, 33, 830–837. [Google Scholar] [CrossRef]
  108. Dietrich, M.A.; Słowińska, M.; Karol, H.; Adamek, M.; Steinhagen, D.; Hejmej, A.; Bilińska, B.; Ciereszko, A. Serine protease inhibitor Kazal-type 2 is expressed in the male reproductive tract of carp with a possible role in antimicrobial protection. Fish Shellfish Immunol. 2017, 60, 150–163. [Google Scholar] [CrossRef]
  109. Kim, B.Y.; Lee, K.S.; Zou, F.M.; Wan, H.; Choi, Y.S.; Yoon, H.J.; Kwon, H.W.; Je, Y.H.; Jin, B.R. Antimicrobial activity of a honeybee (Apis cerana) venom Kazal-type serine protease inhibitor. Toxicon 2013, 76, 110–117. [Google Scholar] [CrossRef]
  110. Mende, K.; Petoukhova, O.; Koulitchkova, V.; Schaub, G.A.; Lange, U.; Kaufmann, R.; Nowak, G. Dipetalogastin, a potent thrombin inhibitor from the blood-sucking insect. Dipetalogaster maximus cDNA cloning, expression and characterization. Eur. J. Biochem. 1999, 266, 583–590. [Google Scholar] [CrossRef]
  111. Campos, I.T.; Amino, R.; Sampaio, C.A.; Auerswald, E.A.; Friedrich, T.; Lemaire, H.G.; Schenkman, S.; Tanaka, A.S. Infestin, a thrombin inhibitor presents in Triatoma infestans midgut, a Chagas’ disease vector: Gene cloning, expression and characterization of the inhibitor. Insect. Biochem. Mol. Biol. 2002, 32, 991–997. [Google Scholar] [CrossRef] [PubMed]
  112. Campos, I.T.; Tanaka-Azevedo, A.M.; Tanaka, A.S. Identification and characterization of a novel factor XIIa inhibitor in the hematophagous insect, Triatoma infestans (Hemiptera: Reduviidae). FEBS Lett. 2004, 577, 512–516. [Google Scholar] [CrossRef] [PubMed]
  113. Watanabe, R.M.; Tanaka-Azevedo, A.M.; Araujo, M.S.; Juliano, M.A.; Tanaka, A.S. Characterization of thrombin inhibitory mechanism of rAaTI, a Kazal-type inhibitor from Aedes aegypti with anticoagulant activity. Biochimie 2011, 93, 618–623. [Google Scholar] [CrossRef] [PubMed]
  114. Bartelt, D.C.; Shapanka, R.; Greene, L.J. The primary structure of the human pancreatic secretory trypsin inhibitor. Amino acid sequence of the reduced S-aminoethylated protein. Arch. Biochem. Biophys. 1977, 179, 189–199. [Google Scholar] [CrossRef]
  115. Horii, A.; Kobayashi, T.-A.; Tomita, N.; Yamamoto, T.; Fukushige, S.; Murotsu, T.; Ogawa, M.; Mori, T.; Matsubara, K.i. Primary structure of human pancreatic secretory trypsin inhibitor (PSTI) gene. Biochem. Biophys. Res. Commun. 1987, 149, 635–641. [Google Scholar] [CrossRef]
  116. Mehner, C.; Radisky, E.S. Bad Tumors Made Worse: SPINK1. Front. Cell. Dev. Biol. 2019, 7, 10. [Google Scholar] [CrossRef]
  117. Lin, T.C. Functional Roles of SPINK1 in Cancers. Int. J. Mol. Sci. 2021, 22, 3814. [Google Scholar] [CrossRef]
  118. Guo, M.; Zhou, X.; Han, X.; Zhang, Y.; Jiang, L. SPINK1 is a prognosis predicting factor of non-small cell lung cancer and regulates redox homeostasis. Oncol. Lett. 2019, 18, 6899–6908. [Google Scholar] [CrossRef]
  119. Lu, F.; Shah, P.A.; Rao, A.; Gifford-Hollingsworth, C.; Chen, A.; Trey, G.; Soryal, M.; Talat, A.; Aslam, A.; Nasir, B.; et al. Liver Cancer-Specific Serine Protease Inhibitor Kazal Is a Potentially Novel Biomarker for the Early Detection of Hepatocellular Carcinoma. Clin. Transl. Gastroenterol. 2020, 11, e00271. [Google Scholar] [CrossRef]
  120. Jia, J.; Ga, L.; Liu, Y.; Yang, Z.; Wang, Y.; Guo, X.; Ma, R.; Liu, R.; Li, T.; Tang, Z.; et al. Serine Protease Inhibitor Kazal Type 1, A Potential Biomarker for the Early Detection, Targeting, and Prediction of Response to Immune Checkpoint Blockade Therapies in Hepatocellular Carcinoma. Front. Immunol. 2022, 13, 923031. [Google Scholar] [CrossRef]
  121. Appleyard, G.; Tisdale, M. Inhibition of the growth of human coronavirus 229E by leupeptin. J. Gen. Virol. 1985, 66 Pt 2, 363–366. [Google Scholar] [CrossRef] [PubMed]
  122. Ramakrishnan, J.; Kandasamy, S.; Iruthayaraj, A.; Magudeeswaran, S.; Chinnasamy, K.; Poomani, K. Strong Binding of Leupeptin with TMPRSS2 Protease May Be an Alternative to Camostat and Nafamostat for SARS-CoV-2 Repurposed Drug: Evaluation from Molecular Docking and Molecular Dynamics Simulations. Appl. Biochem. Biotechnol. 2021, 193, 1909–1923. [Google Scholar] [CrossRef] [PubMed]
  123. Di Nisio, M.; Middeldorp, S.; Büller, H.R. Direct thrombin inhibitors. N. Engl. J. Med. 2005, 353, 1028–1040. [Google Scholar] [CrossRef] [PubMed]
  124. Esslinger, H.U.; Haas, S.; Maurer, R.; Lassmann, A.; Dübbers, K.; Müller-Peltzer, H. Pharmacodynamic and safety results of PEG-Hirudin in healthy volunteers. Thromb. Haemost. 1997, 77, 911–919. [Google Scholar] [CrossRef]
  125. Bossavy, J.P.; Sakariassen, K.S.; Rübsamen, K.; Thalamas, C.; Boneu, B.; Cadroy, Y. Comparison of the antithrombotic effect of PEG-hirudin and heparin in a human ex vivo model of arterial thrombosis. Arterioscler. Thromb. Vasc. Biol. 1999, 19, 1348–1353. [Google Scholar] [CrossRef]
  126. Johns, D.G.; Campeau, L.C.; Banka, P.; Bautmans, A.; Bueters, T.; Bianchi, E.; Branca, D.; Bulger, P.G.; Crevecoeur, I.; Ding, F.X.; et al. Orally Bioavailable Macrocyclic Peptide That Inhibits Binding of PCSK9 to the Low Density Lipoprotein Receptor. Circulation 2023, 148, 144–158. [Google Scholar] [CrossRef]
  127. Zhang, Z.; Shen, C.; Fang, M.; Han, Y.; Long, C.; Liu, W.; Yang, M.; Liu, M.; Zhang, D.; Cao, Q.; et al. Novel contact-kinin inhibitor sylvestin targets thromboinflammation and ameliorates ischemic stroke. Cell Mol. Life Sci. 2022, 79, 240. [Google Scholar] [CrossRef]
  128. Lin, K.; Perni, R.B.; Kwong, A.D.; Lin, C. VX-950, a novel hepatitis C virus (HCV) NS3-4A protease inhibitor, exhibits potent antiviral activities in HCv replicon cells. Antimicrob. Agents Chemother. 2006, 50, 1813–1822. [Google Scholar] [CrossRef]
  129. Xu, P.; Andreasen, P.A.; Huang, M. Structural Principles in the Development of Cyclic Peptidic Enzyme Inhibitors. Int. J. Biol. Sci. 2017, 13, 1222–1233. [Google Scholar] [CrossRef]
  130. Huntington, J.A. Serpin structure, function and dysfunction. J. Thromb. Haemost. 2011, 9 (Suppl. 1), 26–34. [Google Scholar] [CrossRef]
  131. Kellici, T.F.; Pilka, E.S.; Bodkin, M.J. Small-molecule modulators of serine protease inhibitor proteins (serpins). Drug Discov. Today 2021, 26, 442–454. [Google Scholar] [CrossRef] [PubMed]
  132. Matamala, N.; Martínez, M.T.; Lara, B.; Pérez, L.; Vázquez, I.; Jimenez, A.; Barquín, M.; Ferrarotti, I.; Blanco, I.; Janciauskiene, S.; et al. Alternative transcripts of the SERPINA1 gene in alpha-1 antitrypsin deficiency. J. Transl. Med. 2015, 13, 211. [Google Scholar] [CrossRef] [PubMed]
  133. Sharp, H.L. The current status of alpha-1-antityrpsin, a protease inhibitor, in gastrointestinal disease. Gastroenterology 1976, 70, 611–621. [Google Scholar] [CrossRef] [PubMed]
  134. Jonigk, D.; Al-Omari, M.; Maegel, L.; Müller, M.; Izykowski, N.; Hong, J.; Hong, K.; Kim, S.H.; Dorsch, M.; Mahadeva, R.; et al. Anti-inflammatory and immunomodulatory properties of α1-antitrypsin without inhibition of elastase. Proc. Natl. Acad. Sci. USA 2013, 110, 15007–15012. [Google Scholar] [CrossRef]
  135. Connolly, B.; Isaacs, C.; Cheng, L.; Asrani, K.H.; Subramanian, R.R. SERPINA1 mRNA as a Treatment for Alpha-1 Antitrypsin Deficiency. J. Nucleic Acids 2018, 2018, 8247935. [Google Scholar] [CrossRef]
  136. Herman, W.A.; Seńko, A.; Korczowska, I.; Lacka, K. Assessment of selected serum inflammatory markers of acute phase response and their correlations with adrenal androgens and metabolic syndrome in a population of men over the age of 40. Pol. Arch. Med. Wewn. 2009, 119, 704–711. [Google Scholar] [CrossRef]
  137. Kalsheker, N.A. Alpha 1-antichymotrypsin. Int. J. Biochem. Cell Biol. 1996, 28, 961–964. [Google Scholar] [CrossRef]
  138. Forsyth, S.; Horvath, A.; Coughlin, P. A review and comparison of the murine alpha1-antitrypsin and alpha1-antichymotrypsin multigene clusters with the human clade A serpins. Genomics 2003, 81, 336–345. [Google Scholar] [CrossRef]
  139. Duranton, J.; Boudier, C.; Belorgey, D.; Mellet, P.; Bieth, J.G. DNA strongly impairs the inhibition of cathepsin G by alpha(1)-antichymotrypsin and alpha(1)-proteinase inhibitor. J. Biol. Chem. 2000, 275, 3787–3792. [Google Scholar] [CrossRef]
  140. Heit, C.; Jackson, B.C.; McAndrews, M.; Wright, M.W.; Thompson, D.C.; Silverman, G.A.; Nebert, D.W.; Vasiliou, V. Update of the human and mouse SERPIN gene superfamily. Hum. Genom. 2013, 7, 22. [Google Scholar] [CrossRef]
  141. Björk, I.; Olson, S.T. Antithrombin. A bloody important serpin. Adv. Exp. Med. Biol. 1997, 425, 17–33. [Google Scholar] [PubMed]
  142. Horie, S.; Ishii, H.; Kazama, M. Heparin-like glycosaminoglycan is a receptor for antithrombin III-dependent but not for thrombin-dependent prostacyclin production in human endothelial cells. Thromb. Res. 1990, 59, 895–904. [Google Scholar] [CrossRef] [PubMed]
  143. Levy, J.H.; Sniecinski, R.M.; Welsby, I.J.; Levi, M. Antithrombin: Anti-inflammatory properties and clinical applications. Thromb. Haemost. 2016, 115, 712–728. [Google Scholar] [CrossRef] [PubMed]
  144. García Merino, A. Monoclonal antibodies. Basic features. Neurologia 2011, 26, 301–306. [Google Scholar] [CrossRef]
  145. Sounni, N.E.; Noel, A. Targeting the tumor microenvironment for cancer therapy. Clin. Chem. 2013, 59, 85–93. [Google Scholar] [CrossRef]
  146. Gouni-Berthold, I.; Berthold, H.K. PCSK9 antibodies for the treatment of hypercholesterolemia. Nutrients 2014, 6, 5517–5533. [Google Scholar] [CrossRef]
  147. Blom, D.J.; Hala, T.; Bolognese, M.; Lillestol, M.J.; Toth, P.D.; Burgess, L.; Ceska, R.; Roth, E.; Koren, M.J.; Ballantyne, C.M.; et al. A 52-week placebo-controlled trial of evolocumab in hyperlipidemia. N. Engl. J. Med. 2014, 370, 1809–1819. [Google Scholar] [CrossRef]
  148. Lorentz, C.U.; Verbout, N.G.; Wallisch, M.; Hagen, M.W.; Shatzel, J.J.; Olson, S.R.; Puy, C.; Hinds, M.T.; McCarty, O.J.T.; Gailani, D.; et al. Contact Activation Inhibitor and Factor XI Antibody, AB023, Produces Safe, Dose-Dependent Anticoagulation in a Phase 1 First-In-Human Trial. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 799–809. [Google Scholar] [CrossRef]
  149. Thomas, D.; Thelen, K.; Kraff, S.; Schwers, S.; Schiffer, S.; Unger, S.; Yassen, A.; Boxnick, S. BAY 1213790, a fully human IgG1 antibody targeting coagulation factor XIa: First evaluation of safety, pharmacodynamics, and pharmacokinetics. Res. Pract. Thromb. Haemost. 2019, 3, 242–253. [Google Scholar] [CrossRef]
  150. Jiang, L.; Botkjaer, K.A.; Andersen, L.M.; Yuan, C.; Andreasen, P.A.; Huang, M. Rezymogenation of active urokinase induced by an inhibitory antibody. Biochem. J. 2013, 449, 161–166. [Google Scholar] [CrossRef]
  151. Kromann-Hansen, T.; Louise Lange, E.; Peter Sørensen, H.; Hassanzadeh-Ghassabeh, G.; Huang, M.; Jensen, J.K.; Muyldermans, S.; Declerck, P.J.; Komives, E.A.; Andreasen, P.A. Discovery of a novel conformational equilibrium in urokinase-type plasminogen activator. Sci. Rep. 2017, 7, 3385. [Google Scholar] [CrossRef]
  152. Lindner, T.; Loktev, A.; Altmann, A.; Giesel, F.; Kratochwil, C.; Debus, J.; Jäger, D.; Mier, W.; Haberkorn, U. Development of Quinoline-Based Theranostic Ligands for the Targeting of Fibroblast Activation Protein. J. Nucl. Med. 2018, 59, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
  153. Ibrahim, A.A.; Fujimura, T.; Uno, T.; Terada, T.; Hirano, K.I.; Hosokawa, H.; Ohta, A.; Miyata, T.; Ando, K.; Yahata, T. Plasminogen activator inhibitor-1 promotes immune evasion in tumors by facilitating the expression of programmed cell death-ligand 1. Front. Immunol. 2024, 15, 1365894. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, S.; Sun, Y.; Shao, D.; Pan, Y.; Gao, X.; Zhao, P.; Liu, Q.; Shang, G.; Shang, W.; Fu, Z.; et al. High expression of serine protease inhibitor kazal type 1 predicts poor prognosis and promotes the progression and invasion of oral tongue squamous cell carcinoma. Arch. Oral Biol. 2024, 164, 106003. [Google Scholar] [CrossRef]
  155. Enriquez, A.; Baranchuk, A.; Redfearn, D.; Simpson, C.; Abdollah, H.; Michael, K. Dabigatran for the prevention and treatment of thromboembolic disorders. Expert. Rev. Cardiovasc. Ther. 2015, 13, 529–540. [Google Scholar] [CrossRef]
  156. Ricca Gonçalves, L.; Robalo Nunes, A. Dabigatran Reversal: A Practical Approach. Acta Med. Port. 2022, 35, 394–398. [Google Scholar] [CrossRef]
  157. Paik, J. Dabigatran Etexilate: A Review in Pediatric Venous Thromboembolism. Paediatr. Drugs 2022, 24, 423–431. [Google Scholar] [CrossRef]
  158. Lin, S.; Wang, Y.; Zhang, L.; Guan, W. Dabigatran must be used carefully: Literature review and recommendations for management of adverse events. Drug Des. Devel. Ther. 2019, 13, 1527–1533. [Google Scholar] [CrossRef]
  159. Turpie, A.G.G.; Purdham, D.; Ciaccia, A. Nonvitamin K antagonist oral anticoagulant use in patients with renal impairment. Ther. Adv. Cardiovasc. Dis. 2017, 11, 243–256. [Google Scholar] [CrossRef]
  160. Cortese, F.; Calculli, G.; Gesualdo, M.; Cecere, A.; Zito, A.; De Vito, F.; Carbonara, R.; Carbonara, S.; Cortese, A.M.; Ciccone, M.M. Idarucizumab: What Should We Know? Curr. Drug Targets 2018, 19, 81–88. [Google Scholar] [CrossRef]
  161. Eriksson, B.I. Clinical experience of melagatran/ximelagatran in major orthopaedic surgery. Thromb. Res. 2003, 109 (Suppl. 1), S23–S29. [Google Scholar] [CrossRef] [PubMed]
  162. Francis, C.W. Ximelagatran: A new oral anticoagulant. Best Pract. Res. Clin. Haematol. 2004, 17, 139–152. [Google Scholar] [CrossRef] [PubMed]
  163. Koehl, J.L.; Hayes, B.D.; Al-Samkari, H.; Rosovsky, R. A comprehensive evaluation of apixaban in the treatment of venous thromboembolism. Expert Rev. Hematol. 2020, 13, 155–173. [Google Scholar] [CrossRef]
  164. Murphy, G.; Grace, Y.; Chaudry, S.; Chamoun, R. Betrixaban: A Novel Oral Anticoagulant With a New Niche. J. Pharm. Technol. 2018, 34, 123–133. [Google Scholar] [CrossRef]
  165. Eisen, A.; Ruff, C.T. Edoxaban in patients with atrial fibrillation. Ther. Adv. Cardiovasc. Dis. 2017, 11, 81–90. [Google Scholar] [CrossRef]
  166. Raskob, G.E.; van Es, N.; Verhamme, P.; Carrier, M.; Di Nisio, M.; Garcia, D.; Grosso, M.A.; Kakkar, A.K.; Kovacs, M.J.; Mercuri, M.F.; et al. Edoxaban for the Treatment of Cancer-Associated Venous Thromboembolism. N. Engl. J. Med. 2018, 378, 615–624. [Google Scholar] [CrossRef]
  167. Kruger, P.C.; Eikelboom, J.W.; Douketis, J.D.; Hankey, G.J. Deep vein thrombosis: Update on diagnosis and management. Med. J. Aust. 2019, 210, 516–524. [Google Scholar] [CrossRef]
  168. Male, C.; Lensing, A.W.A.; Palumbo, J.S.; Kumar, R.; Nurmeev, I.; Hege, K.; Bonnet, D.; Connor, P.; Hooimeijer, H.L.; Torres, M.; et al. Rivaroxaban compared with standard anticoagulants for the treatment of acute venous thromboembolism in children: A randomised, controlled, phase 3 trial. Lancet Haematol. 2020, 7, e18–e27. [Google Scholar] [CrossRef]
  169. Ahrens, I.; Peter, K.; Lip, G.Y.; Bode, C. Development and clinical applications of novel oral anticoagulants. Part II. Drugs under clinical investigation. Discov. Med. 2012, 13, 445–450. [Google Scholar]
  170. Guertin, K.R.; Choi, Y.M. The discovery of the Factor Xa inhibitor otamixaban: From lead identification to clinical development. Curr. Med. Chem. 2007, 14, 2471–2481. [Google Scholar] [CrossRef]
  171. Goldstein, S.; Bates, E.R.; Bhatt, D.L.; Cao, C.; Holmes, D.; Kupfer, S.; Martinez, F.; Spaeder, J.; Weitz, J.I.; Ye, Z.; et al. Phase 2 study of TAK-442, an oral factor Xa inhibitor, in patients following acute coronary syndrome. Thromb. Haemost. 2014, 111, 1141–1152. [Google Scholar] [CrossRef] [PubMed]
  172. Agnelli, G.; Haas, S.; Ginsberg, J.S.; Krueger, K.A.; Dmitrienko, A.; Brandt, J.T. A phase II study of the oral factor Xa inhibitor LY517717 for the prevention of venous thromboembolism after hip or knee replacement. J. Thromb. Haemost. 2007, 5, 746–753. [Google Scholar] [CrossRef] [PubMed]
  173. Jang, J.H.; Wong, L.; Ko, B.S.; Yoon, S.S.; Li, K.; Baltcheva, I.; Nidamarthy, P.K.; Chawla, R.; Junge, G.; Yap, E.S. Iptacopan monotherapy in patients with paroxysmal nocturnal hemoglobinuria: A 2-cohort open-label proof-of-concept study. Blood Adv. 2022, 6, 4450–4460. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, H.; Rizk, D.V.; Perkovic, V.; Maes, B.; Kashihara, N.; Rovin, B.; Trimarchi, H.; Sprangers, B.; Meier, M.; Kollins, D.; et al. Results of a randomized double-blind placebo-controlled Phase 2 study propose iptacopan as an alternative complement pathway inhibitor for IgA nephropathy. Kidney Int. 2024, 105, 189–199. [Google Scholar] [CrossRef] [PubMed]
  175. Breining, P.; Frølund, A.L.; Højen, J.F.; Gunst, J.D.; Staerke, N.B.; Saedder, E.; Cases-Thomas, M.; Little, P.; Nielsen, L.P.; Søgaard, O.S.; et al. Camostat mesylate against SARS-CoV-2 and COVID-19-Rationale, dosing and safety. Basic Clin. Pharmacol. Toxicol. 2021, 128, 204–212. [Google Scholar] [CrossRef]
  176. Froriep, D.; Clement, B.; Bittner, F.; Mendel, R.R.; Reichmann, D.; Schmalix, W.; Havemeyer, A. Activation of the anti-cancer agent upamostat by the mARC enzyme system. Xenobiotica 2013, 43, 780–784. [Google Scholar] [CrossRef]
  177. Beierlein, W.; Scheule, A.M.; Dietrich, W.; Ziemer, G. Forty years of clinical aprotinin use: A review of 124 hypersensitivity reactions. Ann. Thorac. Surg. 2005, 79, 741–748. [Google Scholar] [CrossRef]
  178. Dietrich, W.; Späth, P.; Ebell, A.; Richter, J.A. Prevalence of anaphylactic reactions to aprotinin: Analysis of two hundred forty-eight reexposures to aprotinin in heart operations. J. Thorac. Cardiovasc. Surg. 1997, 113, 194–201. [Google Scholar] [CrossRef]
  179. Suda, H.; Aoyagi, T.; Hamada, M.; Takeuchi, T.; Umezawa, H. Antipain, a new protease inhibitor isolated from actinomycetes. J. Antibiot. 1972, 25, 263–266. [Google Scholar] [CrossRef]
  180. Raphaelis, S.; Frommlet, F.; Mayer, H.; Koller, A. Implementation of a nurse-led self-management support intervention for patients with cancer-related pain: A cluster randomized phase-IV study with a stepped wedge design (EvANtiPain). BMC Cancer 2020, 20, 559. [Google Scholar] [CrossRef]
  181. Buijs, N.; Vlaming, H.C.; van Haren, M.J.; Martin, N.I. Synthetic Studies with Bacitracin A and Preparation of Analogues Containing Alternative Zinc Binding Groups. Chembiochem 2022, 23, e202200547. [Google Scholar] [CrossRef] [PubMed]
  182. Gennemark, P.; Walter, K.; Clemmensen, N.; Rekić, D.; Nilsson, C.A.M.; Knöchel, J.; Hölttä, M.; Wernevik, L.; Rosengren, B.; Kakol-Palm, D.; et al. An oral antisense oligonucleotide for PCSK9 inhibition. Sci. Transl. Med. 2021, 13, eabe9117. [Google Scholar] [CrossRef] [PubMed]
  183. Graetz, T.J.; Tellor, B.R.; Smith, J.R.; Avidan, M.S. Desirudin: A review of the pharmacology and clinical application for the prevention of deep vein thrombosis. Expert Rev. Cardiovasc. Ther. 2011, 9, 1101–1109. [Google Scholar] [CrossRef]
  184. Ballantyne, C.M.; Banka, P.; Mendez, G.; Garcia, R.; Rosenstock, J.; Rodgers, A.; Mendizabal, G.; Mitchel, Y.; Catapano, A.L. Phase 2b Randomized Trial of the Oral PCSK9 Inhibitor MK-0616. J. Am. Coll. Cardiol. 2023, 81, 1553–1564. [Google Scholar] [CrossRef] [PubMed]
  185. Lin, C.; Kwong, A.D.; Perni, R.B. Discovery and development of VX-950, a novel, covalent, and reversible inhibitor of hepatitis C virus NS3.4A serine protease. Infect. Disord. Drug Targets 2006, 6, 3–16. [Google Scholar] [CrossRef]
  186. Roujeau, J.C.; Mockenhaupt, M.; Tahan, S.R.; Henshaw, J.; Martin, E.C.; Harding, M.; van Baelen, B.; Bengtsson, L.; Singhal, P.; Kauffman, R.S.; et al. Telaprevir-related dermatitis. JAMA Dermatol. 2013, 149, 152–158. [Google Scholar] [CrossRef]
  187. Garcias-Ladaria, J.; Pérez-Ferriols, A.; Ortega-García, P.; Diago, M. Management of refractory telaprevir-induced dermatitis using oral corticosteroids. Actas Dermosifiliogr. 2014, 105, e55–e60. [Google Scholar] [CrossRef]
  188. Bentounes, N.K.; Melicine, S.; Martin, A.C.; Smadja, D.M.; Gendron, N. Development of new anticoagulant in 2023: Prime time for anti-factor XI and XIa inhibitors. J. Med. Vasc. 2023, 48, 69–80. [Google Scholar] [CrossRef]
  189. Ray, K.K.; Wright, R.S.; Kallend, D.; Koenig, W.; Leiter, L.A.; Raal, F.J.; Bisch, J.A.; Richardson, T.; Jaros, M.; Wijngaard, P.L.J.; et al. Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. N. Engl. J. Med. 2020, 382, 1507–1519. [Google Scholar] [CrossRef]
  190. Scott, A.M.; Wiseman, G.; Welt, S.; Adjei, A.; Lee, F.T.; Hopkins, W.; Divgi, C.R.; Hanson, L.H.; Mitchell, P.; Gansen, D.N.; et al. A Phase I dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clin. Cancer Res. 2003, 9, 1639–1647. [Google Scholar]
  191. Hao, Y.; Yang, Y.L.; Wang, Y.C.; Li, J. Effect of the Early Application of Evolocumab on Blood Lipid Profile and Cardiovascular Prognosis in Patients with Extremely High-Risk Acute Coronary Syndrome. Int. Heart J. 2022, 63, 669–677. [Google Scholar] [CrossRef] [PubMed]
  192. Pérez de Isla, L.; Díaz-Díaz, J.L.; Romero, M.J.; Muñiz-Grijalvo, O.; Mediavilla, J.D.; Argüeso, R.; Sánchez Muñoz-Torrero, J.F.; Rubio, P.; Álvarez-Baños, P.; Ponte, P.; et al. Alirocumab and Coronary Atherosclerosis in Asymptomatic Patients with Familial Hypercholesterolemia: The ARCHITECT Study. Circulation 2023, 147, 1436–1443. [Google Scholar] [CrossRef] [PubMed]
  193. Lafayette, R.A.; Rovin, B.H.; Reich, H.N.; Tumlin, J.A.; Floege, J.; Barratt, J. Safety, Tolerability and Efficacy of Narsoplimab, a Novel MASP-2 Inhibitor for the Treatment of IgA Nephropathy. Kidney Int. Rep. 2020, 5, 2032–2041. [Google Scholar] [CrossRef] [PubMed]
  194. Weitz, J.I.; Bauersachs, R.; Becker, B.; Berkowitz, S.D.; Freitas, M.C.S.; Lassen, M.R.; Metzig, C.; Raskob, G.E. Effect of Osocimab in Preventing Venous Thromboembolism Among Patients Undergoing Knee Arthroplasty: The FOXTROT Randomized Clinical Trial. JAMA 2020, 323, 130–139. [Google Scholar] [CrossRef]
  195. Poulsen, A.; Kang, C.; Keller, T.H. Drug design for flavivirus proteases: What are we missing? Curr. Pharm. Des. 2014, 20, 3422–3427. [Google Scholar] [CrossRef]
  196. Kang, C.; Keller, T.H.; Luo, D. Zika Virus Protease: An Antiviral Drug Target. Trends Microbiol. 2017, 25, 797–808. [Google Scholar] [CrossRef]
  197. Saha, N.; Robev, D.; Himanen, J.P.; Nikolov, D.B. ADAM proteases: Emerging role and targeting of the non-catalytic domains. Cancer Lett. 2019, 467, 50–57. [Google Scholar] [CrossRef]
  198. Xie, X.; Guo, N.; Xue, G.; Xie, D.; Yuan, C.; Harrison, J.; Li, J.; Jiang, L.; Huang, M. Solution Structure of SpoIVB Reveals Mechanism of PDZ Domain-Regulated Protease Activity. Front. Microbiol. 2019, 10, 1232. [Google Scholar] [CrossRef]
  199. Oberst, M.D.; Williams, C.A.; Dickson, R.B.; Johnson, M.D.; Lin, C.Y. The activation of matriptase requires its noncatalytic domains, serine protease domain, and its cognate inhibitor. J. Biol. Chem. 2003, 278, 26773–26779. [Google Scholar] [CrossRef]
  200. Al-Horani, R.A.; Afosah, D.K. Recent advances in the discovery and development of factor XI/XIa inhibitors. Med. Res. Rev. 2018, 38, 1974–2023. [Google Scholar] [CrossRef]
  201. Fjällskog, M.L.; Frii, L.; Bergh, J. Is Cremophor EL, solvent for paclitaxel, cytotoxic? Lancet 1993, 342, 873. [Google Scholar] [CrossRef] [PubMed]
  202. Fader, A.N.; Rose, P.G. Abraxane for the treatment of gynecologic cancer patients with severe hypersensitivity reactions to paclitaxel. Int. J. Gynecol. Cancer 2009, 19, 1281–1283. [Google Scholar] [CrossRef] [PubMed]
  203. Borensztajn, K.; Spek, C.A. Blood coagulation factor Xa as an emerging drug target. Expert Opin. Ther. Targets 2011, 15, 341–349. [Google Scholar] [CrossRef] [PubMed]
  204. Polack, B. Thrombin: A multifunctional enzyme. Ann. Biol. Clin. 2003, 61, 23–31. [Google Scholar]
  205. Ding, L.; Shu, Z.; Hao, J.; Luo, X.; Ye, X.; Zhu, W.; Duan, W.; Chen, Z. Schixator, a new FXa inhibitor from Schistosoma japonicum with antithrombotic effect and low bleeding risk. Biochem. Biophys. Res. Commun. 2022, 603, 138–143. [Google Scholar] [CrossRef]
  206. Narayanan, S. Multifunctional roles of thrombin. Ann. Clin. Lab. Sci. 1999, 29, 275–280. [Google Scholar]
  207. Laboissière, M.C.; Young, M.M.; Pinho, R.G.; Todd, S.; Fletterick, R.J.; Kuntz, I.; Craik, C.S. Computer-assisted mutagenesis of ecotin to engineer its secondary binding site for urokinase inhibition. J. Biol. Chem. 2002, 277, 26623–26631. [Google Scholar] [CrossRef]
  208. Mackman, R.L.; Katz, B.A.; Breitenbucher, J.G.; Hui, H.C.; Verner, E.; Luong, C.; Liu, L.; Sprengeler, P.A. Exploiting subsite S1 of trypsin-like serine proteases for selectivity: Potent and selective inhibitors of urokinase-type plasminogen activator. J. Med. Chem. 2001, 44, 3856–3871. [Google Scholar] [CrossRef]
  209. Sperl, S.; Jacob, U.; Arroyo de Prada, N.; Stürzebecher, J.; Wilhelm, O.G.; Bode, W.; Magdolen, V.; Huber, R.; Moroder, L. (4-aminomethyl)phenylguanidine derivatives as nonpeptidic highly selective inhibitors of human urokinase. Proc. Natl. Acad. Sci. USA 2000, 97, 5113–5118. [Google Scholar] [CrossRef]
  210. Spencer, J.R.; McGee, D.; Allen, D.; Katz, B.A.; Luong, C.; Sendzik, M.; Squires, N.; Mackman, R.L. 4-Aminoarylguanidine and 4-aminobenzamidine derivatives as potent and selective urokinase-type plasminogen activator inhibitors. Bioorg. Med. Chem. Lett. 2002, 12, 2023–2026. [Google Scholar] [CrossRef]
  211. Duffy, F.J.; Devocelle, M.; Shields, D.C. Computational approaches to developing short cyclic peptide modulators of protein-protein interactions. Methods Mol. Biol. 2015, 1268, 241–271. [Google Scholar] [CrossRef] [PubMed]
  212. Guerra, Y.; Armijos-Jaramillo, V.; Pons, T.; Tejera, E.; Berry, C. Canonical or noncanonical? Structural plasticity of serine protease-binding loops in Kunitz-STI protease inhibitors. Protein Sci. 2023, 32, e4570. [Google Scholar] [CrossRef] [PubMed]
  213. Gryaznov, S.; Skorski, T.; Cucco, C.; Nieborowska-Skorska, M.; Chiu, C.Y.; Lloyd, D.; Chen, J.K.; Koziolkiewicz, M.; Calabretta, B. Oligonucleotide N3’-->P5’ phosphoramidates as antisense agents. Nucleic Acids Res. 1996, 24, 1508–1514. [Google Scholar] [CrossRef] [PubMed]
  214. Summerton, J. Morpholino antisense oligomers: The case for an RNase H-independent structural type. Biochim. Biophys. Acta 1999, 1489, 141–158. [Google Scholar] [CrossRef]
  215. Addison, M.L.; Ranasinghe, P.; Webb, D.J. Novel Pharmacological Approaches in the Treatment of Hypertension: A Focus on RNA-Based Therapeutics. Hypertension 2023, 80, 2243–2254. [Google Scholar] [CrossRef]
  216. Brown, D.A.; Kang, S.H.; Gryaznov, S.M.; DeDionisio, L.; Heidenreich, O.; Sullivan, S.; Xu, X.; Nerenberg, M.I. Effect of phosphorothioate modification of oligodeoxynucleotides on specific protein binding. J. Biol. Chem. 1994, 269, 26801–26805. [Google Scholar] [CrossRef]
  217. Teplova, M.; Minasov, G.; Tereshko, V.; Inamati, G.B.; Cook, P.D.; Manoharan, M.; Egli, M. Crystal structure and improved antisense properties of 2’-O-(2-methoxyethyl)-RNA. Nat. Struct. Biol. 1999, 6, 535–539. [Google Scholar] [CrossRef]
  218. Koshkin, A.A.; Singh, S.K.; Nielsen, P.; Rajwanshi, V.K.; Kumar, R.; Meldgaard, M.; Olsen, C.E.; Wengel, J. LNA (Locked Nucleic Acids): Synthesis of the adenine, cytosine, guanine, 5-methylcytosine, thymine and uracil bicyclonucleoside monomers, oligomerisation, and unprecedented nucleic acid recognition. Tetrahedron 1998, 54, 3607–3630. [Google Scholar] [CrossRef]
  219. Swayze, E.E.; Siwkowski, A.M.; Wancewicz, E.V.; Migawa, M.T.; Wyrzykiewicz, T.K.; Hung, G.; Monia, B.P.; Bennett, C.F. Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals. Nucleic Acids Res. 2007, 35, 687–700. [Google Scholar] [CrossRef]
  220. Koizumi, M. ENA oligonucleotides as therapeutics. Curr. Opin. Mol. Ther. 2006, 8, 144–149. [Google Scholar]
  221. Seth, P.P.; Siwkowski, A.; Allerson, C.R.; Vasquez, G.; Lee, S.; Prakash, T.P.; Wancewicz, E.V.; Witchell, D.; Swayze, E.E. Short antisense oligonucleotides with novel 2’-4’ conformationaly restricted nucleoside analogues show improved potency without increased toxicity in animals. J. Med. Chem. 2009, 52, 10–13. [Google Scholar] [CrossRef] [PubMed]
  222. Jason, T.L.; Koropatnick, J.; Berg, R.W. Toxicology of antisense therapeutics. Toxicol. Appl. Pharmacol. 2004, 201, 66–83. [Google Scholar] [CrossRef] [PubMed]
  223. Chan, J.H.; Lim, S.; Wong, W.S. Antisense oligonucleotides: From design to therapeutic application. Clin. Exp. Pharmacol. Physiol. 2006, 33, 533–540. [Google Scholar] [CrossRef] [PubMed]
  224. Wurster, C.D.; Ludolph, A.C. Antisense oligonucleotides in neurological disorders. Ther. Adv. Neurol. Disord. 2018, 11, 1756286418776932. [Google Scholar] [CrossRef]
  225. Bhalani, D.V.; Nutan, B.; Kumar, A.; Singh Chandel, A.K. Bioavailability Enhancement Techniques for Poorly Aqueous Soluble Drugs and Therapeutics. Biomedicines 2022, 10, 2055. [Google Scholar] [CrossRef]
  226. Li, H.; Wang, Z.; Yu, S.; Chen, S.; Zhou, Y.; Qu, Y.; Xu, P.; Jiang, L.; Yuan, C.; Huang, M. Albumin-based drug carrier targeting urokinase receptor for cancer therapy. Int. J. Pharm. 2023, 634, 122636. [Google Scholar] [CrossRef]
  227. Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and challenges of extracellular vesicle-based drug delivery system: Considering cell source. Drug Deliv. 2020, 27, 585–598. [Google Scholar] [CrossRef]
  228. Vandooren, J.; Opdenakker, G.; Loadman, P.M.; Edwards, D.R. Proteases in cancer drug delivery. Adv. Drug Deliv. Rev. 2016, 97, 144–155. [Google Scholar] [CrossRef]
  229. Liu, S.; Aaronson, H.; Mitola, D.J.; Leppla, S.H.; Bugge, T.H. Potent antitumor activity of a urokinase-activated engineered anthrax toxin. Proc. Natl. Acad. Sci. USA 2003, 100, 657–662. [Google Scholar] [CrossRef]
  230. Liu, S.; Netzel-Arnett, S.; Birkedal-Hansen, H.; Leppla, S.H. Tumor cell-selective cytotoxicity of matrix metalloproteinase-activated anthrax toxin. Cancer Res. 2000, 60, 6061–6067. [Google Scholar]
  231. Devy, L.; de Groot, F.M.; Blacher, S.; Hajitou, A.; Beusker, P.H.; Scheeren, H.W.; Foidart, J.M.; Noël, A. Plasmin-activated doxorubicin prodrugs containing a spacer reduce tumor growth and angiogenesis without systemic toxicity. FASEB J. 2004, 18, 565–567. [Google Scholar] [CrossRef] [PubMed]
  232. Fielding, A.K.; Maurice, M.; Morling, F.J.; Cosset, F.L.; Russell, S.J. Inverse targeting of retroviral vectors: Selective gene transfer in a mixed population of hematopoietic and nonhematopoietic cells. Blood 1998, 91, 1802–1809. [Google Scholar] [CrossRef] [PubMed]
  233. Zhao, Y.; Li, Y.; Wang, F.; Gan, X.; Zheng, T.; Chen, M.; Wei, L.; Chen, J.; Yu, C. CES1-Triggered Liver-Specific Cargo Release of CRISPR/Cas9 Elements by Cationic Triadic Copolymeric Nanoparticles Targeting Gene Editing of PCSK9 for Hyperlipidemia Amelioration. Adv. Sci. 2023, 10, e2300502. [Google Scholar] [CrossRef] [PubMed]
  234. Prakash, T.P.; Graham, M.J.; Yu, J.; Carty, R.; Low, A.; Chappell, A.; Schmidt, K.; Zhao, C.; Aghajan, M.; Murray, H.F.; et al. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res. 2014, 42, 8796–8807. [Google Scholar] [CrossRef] [PubMed]
  235. Wohlfart, S.; Gelperina, S.; Kreuter, J. Transport of drugs across the blood-brain barrier by nanoparticles. J. Control Release 2012, 161, 264–273. [Google Scholar] [CrossRef]
  236. Jang, W.D.; Jeon, S.; Kim, S.; Lee, S.Y. Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assay. Proc. Natl. Acad. Sci. USA 2021, 118, e2024302118. [Google Scholar] [CrossRef]
  237. Yarden, Y.; Sliwkowski, M.X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2001, 2, 127–137. [Google Scholar] [CrossRef]
  238. Peters, C.; Brown, S. Antibody–drug conjugates as novel anti-cancer chemotherapeutics. Biosci. Rep. 2015, 35, e00225. [Google Scholar] [CrossRef]
  239. Baah, S.; Laws, M.; Rahman, K.M. Antibody-Drug Conjugates-A Tutorial Review. Molecules 2021, 26, 2943. [Google Scholar] [CrossRef]
  240. LeBeau, A.M.; Duriseti, S.; Murphy, S.T.; Pepin, F.; Hann, B.; Gray, J.W.; VanBrocklin, H.F.; Craik, C.S. Targeting uPAR with Antagonistic Recombinant Human Antibodies in Aggressive Breast Cancer. Cancer Res. 2013, 73, 2070–2081. [Google Scholar] [CrossRef]
  241. Li, R.; Zheng, K.; Hu, P.; Chen, Z.; Zhou, S.; Chen, J.; Yuan, C.; Chen, S.; Zheng, W.; Ma, E.; et al. A novel tumor targeting drug carrier for optical imaging and therapy. Theranostics 2014, 4, 642–659. [Google Scholar] [CrossRef] [PubMed]
  242. Zheng, K.; Li, R.; Zhou, X.; Hu, P.; Zhang, Y.; Huang, Y.; Chen, Z.; Huang, M. Dual actions of albumin packaging and tumor targeting enhance the antitumor efficacy and reduce the cardiotoxicity of doxorubicin in vivo. Int. J. Nanomed. 2015, 10, 5327–5342. [Google Scholar] [CrossRef]
  243. Zhou, Y.; Yu, S.; Chen, D.; Li, H.; Xu, P.; Yuan, C.; Jiang, L.; Huang, M. Nafamostat Mesylate in Combination with the Mouse Amino-Terminal Fragment of Urokinase-Human Serum Albumin Improves the Treatment Outcome of Triple-Negative Breast Cancer Therapy. Mol. Pharm. 2023, 20, 905–917. [Google Scholar] [CrossRef] [PubMed]
  244. Zhou, X.; Zheng, K.; Li, R.; Chen, Z.; Yuan, C.; Hu, P.; Chen, J.; Xue, J.; Huang, M. A drug carrier targeting murine uPAR for photodynamic therapy and tumor imaging. Acta Biomater. 2015, 23, 116–126. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Simplified diagram of serine protease catalysis hydrolysis mechanism. The -OH group of Ser acts as a nucleophile, attacking the carbonyl of the substrate peptide bond, leading to peptide bond hydrolysis. A pair of electrons from the nitrogen in the imidazole ring of His can accept a hydrogen from the Ser-OH group. The carboxyl group of Asp stabilizes the tetrahedral intermediate structure through hydrogen bonding with His, followed by the formation of an acyl-enzyme intermediate. A water molecule then attacks the acyl-enzyme intermediate, producing a second tetrahedral intermediate, which releases the new product from the S1 pocket.
Figure 1. Simplified diagram of serine protease catalysis hydrolysis mechanism. The -OH group of Ser acts as a nucleophile, attacking the carbonyl of the substrate peptide bond, leading to peptide bond hydrolysis. A pair of electrons from the nitrogen in the imidazole ring of His can accept a hydrogen from the Ser-OH group. The carboxyl group of Asp stabilizes the tetrahedral intermediate structure through hydrogen bonding with His, followed by the formation of an acyl-enzyme intermediate. A water molecule then attacks the acyl-enzyme intermediate, producing a second tetrahedral intermediate, which releases the new product from the S1 pocket.
Catalysts 14 00787 g001
Figure 2. Schematic diagram of serine protease functions. Injury to the body, whether internal or external, activates coagulation factors, triggering the coagulation cascade, and ultimately converting prothrombin into thrombin. Thrombin activates fibrinogen, which is then transformed into fibrin, precipitating at the wound site to form a thrombus. This process promotes wound healing or can lead to thrombotic diseases. The presence of tPA and uPA in the body activates plasminogen to convert into plasmin, which dissolves fibrin and facilitates thrombus clearance, playing a crucial role in the clinical treatment of thrombosis. However, excessive activation of plasmin can lead to severe bleeding. Additionally, uPA-induced excessive plasmin activation can alter the tumor microenvironment and decrease the extracellular matrix (ECM). Therefore, high levels of uPA are positively correlated with various cancers. This indicates that uPA can be a biomarker for highly invasive cancers [22].
Figure 2. Schematic diagram of serine protease functions. Injury to the body, whether internal or external, activates coagulation factors, triggering the coagulation cascade, and ultimately converting prothrombin into thrombin. Thrombin activates fibrinogen, which is then transformed into fibrin, precipitating at the wound site to form a thrombus. This process promotes wound healing or can lead to thrombotic diseases. The presence of tPA and uPA in the body activates plasminogen to convert into plasmin, which dissolves fibrin and facilitates thrombus clearance, playing a crucial role in the clinical treatment of thrombosis. However, excessive activation of plasmin can lead to severe bleeding. Additionally, uPA-induced excessive plasmin activation can alter the tumor microenvironment and decrease the extracellular matrix (ECM). Therefore, high levels of uPA are positively correlated with various cancers. This indicates that uPA can be a biomarker for highly invasive cancers [22].
Catalysts 14 00787 g002
Figure 3. Mechanisms of action of serine protease inhibitors. (A) The mechanism of action of classic serine protease inhibitors [27]. The P1 group inserts into the interior of the S1 pocket of the serine protease. (B) Mechanism of action of non-standard inhibitors. Non-canonical inhibitors interact with the active site of serine proteases through their N-terminus, occupying the active site without forming new peptide bonds. This leads to the “closure” of the S1 pocket, preventing substrates from entering the S1 pocket.
Figure 3. Mechanisms of action of serine protease inhibitors. (A) The mechanism of action of classic serine protease inhibitors [27]. The P1 group inserts into the interior of the S1 pocket of the serine protease. (B) Mechanism of action of non-standard inhibitors. Non-canonical inhibitors interact with the active site of serine proteases through their N-terminus, occupying the active site without forming new peptide bonds. This leads to the “closure” of the S1 pocket, preventing substrates from entering the S1 pocket.
Catalysts 14 00787 g003
Figure 4. Three possible mechanisms for inhibiting autolysis [34]. (A) The binding of the allosteric inhibitor on the autolysis loop leads to the movement of the oxyanion stabilizing loop and the distortion of the oxyanion hole. (B) Occlusion of the S1 specificity pocket caused by the binding of the antibody to the autolysis loop. (C) The salt bridge between residue Asp194 and the N-terminal residue 16 is disrupted, which causes the N-terminal residue 16 to fall out of the protease and leads to an inactive and zymogen-like protease.
Figure 4. Three possible mechanisms for inhibiting autolysis [34]. (A) The binding of the allosteric inhibitor on the autolysis loop leads to the movement of the oxyanion stabilizing loop and the distortion of the oxyanion hole. (B) Occlusion of the S1 specificity pocket caused by the binding of the antibody to the autolysis loop. (C) The salt bridge between residue Asp194 and the N-terminal residue 16 is disrupted, which causes the N-terminal residue 16 to fall out of the protease and leads to an inactive and zymogen-like protease.
Catalysts 14 00787 g004
Table 1. Summary of the structure, molecular weight, and targets of small-molecule inhibitors.
Table 1. Summary of the structure, molecular weight, and targets of small-molecule inhibitors.
NumberCompound NamesStructuresMolecular Weight g/molTargets
1DabigatranCatalysts 14 00787 i001471.5Thrombin
2XimelagatranCatalysts 14 00787 i002473.6Thrombin
3MelagatranCatalysts 14 00787 i003429.5Thrombin
4AZD0837Catalysts 14 00787 i004496.9Thrombin
5ApixabanCatalysts 14 00787 i005459.5FXa
6BetrixabanCatalysts 14 00787 i006451.9FXa
7EdoxabanCatalysts 14 00787 i007548.1FXa
8RivaroxabanCatalysts 14 00787 i008435.9FXa
9DarexabanCatalysts 14 00787 i009476.4FXa
10EribaxabanCatalysts 14 00787 i010484.9FXa
11OtamixabanCatalysts 14 00787 i011446.5FXa
12TAK-442Catalysts 14 00787 i012480FXa
13LY517717Catalysts 14 00787 i013459.6FXa
14MilvexianCatalysts 14 00787 i014626.4FXIa
15LNP023Catalysts 14 00787 i015422.5CFB
16NafamostatCatalysts 14 00787 i016347.4broad-spectrum
17CamostatCatalysts 14 00787 i017398.4broad-spectrum
18UpamostatCatalysts 14 00787 i018629.8broad-spectrum
19QuercetinCatalysts 14 00787 i019302.23broad-spectrum
20MyricetinCatalysts 14 00787 i020318.23broad-spectrum
Table 2. Serine protease inhibitors have entered the clinical trial stage.
Table 2. Serine protease inhibitors have entered the clinical trial stage.
NumberCompoundTherapyTarget PhaseIndicationsUsage Limitations Solutions
1DabigatranSmall moleculeThrombinIV [155]NVAF [38,156], VTE [157]Emergency surgery, advanced age [158], pregnant women, severe liver and kidney dysfunction, mechanical heart valve, concomitant administration of drug classes that are strong inhibitors of CYP3A4 and P-glycoprotein [159]Idarucizumab [160], reduce the dosage, switch to safer medications [158]
2XimelagatranSmall moleculeThrombinIIIVTE [161]Liver dysfunction [162]No findings at this stage
3Melagatran Small moleculeThrombinIIIVTE [161]Liver dysfunction [162]No findings at this stage
4AZD0837Small moleculeThrombinIIAF [45], Chronic ventricular fibrillation [46]No findings at this stageNo findings at this stage
5ApixabanSmall moleculeThrombinIIIDVT, PE [163]Dialysis patients, severely impaired liver function, active pathological bleeding [54], pregnant women, severe liver and kidney dysfunction, mechanical heart valve, concomitant administration of drug classes that are strong inhibitors of CYP3A4 and P-glycoprotein [159]Switch to safer medications
6BetrixabanSmall moleculeThrombinIIIVTE [164]No findings at this stageNo findings at this stage
7EdoxabanSmall moleculeThrombinIIIAF [165], VTE [166]Pregnant women, severe liver and kidney dysfunction, mechanical heart valve, concomitant administration of drug classes that are strong inhibitors of CYP3A4 and P-glycoprotein [159]Anticoagulation with intravenous unfractionated heparin, low molecular weight heparin (LMWH), or warfarin [167]
8RivaroxabanSmall moleculeThrombinIIIVTE [168]Pregnant women, severe liver and kidney dysfunction, mechanical heart valve, concomitant administration of drug classes that are strong inhibitors of CYP3A4 and P-glycoprotein [159]Anticoagulation with intravenous unfractionated heparin, low molecular weight heparin (LMWH), or warfarin [167]
9DarexabanSmall moleculeThrombinIIIACS, AF, DVT, PE [169]No findings at this stageNo findings at this stage
10OtamixabanSmall moleculeThrombinIIACS, Non-urgent PCI [170]No findings at this stageNo findings at this stage
11TAK-442Small moleculeThrombinIIACS [171]No findings at this stageNo findings at this stage
12LY517717Small moleculeThrombinIIDVT, PE [172]No findings at this stageNo findings at this stage
13MilvexianSmall moleculeFXIaIINo findings at this stage [63]No findings at this stageNo findings at this stage
14LNP023Small moleculeComplement Factor BIIIPNH [173], IgAN [174]No findings at this stageNo findings at this stage
15NafamostatSmall moleculebroad-spectrumIVAcute pancreatitis, AKI, various malignant tumors [65]No findings at this stageNo findings at this stage
16CamostatSmall moleculebroad-spectrumIVChronic pancreatitis, post-operative reflux esophagitis, COVID-19 [175] No findings at this stageNo findings at this stage
17UpamostatSmall moleculebroad-spectrumIIPancreatic cancer, breast cancer, COVID-19 [176] No findings at this stageNo findings at this stage
18AprotininPolypeptidebroad-spectrumIVAnti-inflammatory and antithrombotic during operation [95] Sensitive to Aprotinin [177,178]Switch to safer medications
19AntipainPolypeptidebroad-spectrumIVAnalgesia [179,180]No findings at this stageNo findings at this stage
20Bacitracin APolypeptidebroad-spectrumIVBacterial infection [181]No findings at this stageNo findings at this stage
21leupeptinPolypeptidebroad-spectrumIVCOVID-19 [182] No findings at this stageNo findings at this stage
22DesirudinPolypeptideThrombinIIIVTE [44]Renal dysfunction [183]Dose adjustment [183]
23PEG-HirudinPolypeptideThrombinIINo findings at this stageNo findings at this stageNo findings at this stage
24MK-0616PolypeptidePCSK9IIHypercholesterolemia [184]No findings at this stageNo findings at this stage
25VX-950PolypeptideNS3/4A proteaseIChronic hepatitis C [185]Skin disease [186]Discontinue and take oral corticosteroids [187]
26AZD8233ASOPCSK9IIHypercholesterolemiaNo findings at this stageNo findings at this stage
27FesomersenASOFXIaIINo findings at this stage [188] No findings at this stageNo findings at this stage
28IONIS-FXIRXASOFXIaIINo findings at this stage [188]No findings at this stageNo findings at this stage
29InclisiranSiRNAPCSK9IIIHypercholesterolemia [81,189]No findings at this stageNo findings at this stage
30SibrotuzumabAntibodyFAPIFibroblast activation protein-positive cancer [190]No findings at this stageNo findings at this stage
31EvolocumabAntibodyPCSK9IIIHyperlipidemia [147]Liver function impairment [191]No findings at this stage
32AlirocumabAntibodyPCSK9IVHypercholesterolemia [192]No findings at this stageNo findings at this stage
33NarsoplimabAntibodyMASP-2IIIIgAN [193]No findings at this stageNo findings at this stage
34XIsomab 3G3AntibodyFXIaIINo findings at this stage [188]No findings at this stageNo findings at this stage
35OsocimabAntibodyFXIaIIVTE [194]No findings at this stageNo findings at this stage
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wei, Y.; Huang, M.; Jiang, L. Advancements in Serine Protease Inhibitors: From Mechanistic Insights to Clinical Applications. Catalysts 2024, 14, 787. https://doi.org/10.3390/catal14110787

AMA Style

Wei Y, Huang M, Jiang L. Advancements in Serine Protease Inhibitors: From Mechanistic Insights to Clinical Applications. Catalysts. 2024; 14(11):787. https://doi.org/10.3390/catal14110787

Chicago/Turabian Style

Wei, Yang, Mingdong Huang, and Longguang Jiang. 2024. "Advancements in Serine Protease Inhibitors: From Mechanistic Insights to Clinical Applications" Catalysts 14, no. 11: 787. https://doi.org/10.3390/catal14110787

APA Style

Wei, Y., Huang, M., & Jiang, L. (2024). Advancements in Serine Protease Inhibitors: From Mechanistic Insights to Clinical Applications. Catalysts, 14(11), 787. https://doi.org/10.3390/catal14110787

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop