Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
Reversible Covalent Inhibition─Desired Covalent Adduct Formation by Mass Action
My Activity
  • Open Access
Review

Reversible Covalent Inhibition─Desired Covalent Adduct Formation by Mass Action
Click to copy article linkArticle link copied!

Open PDF

ACS Chemical Biology

Cite this: ACS Chem. Biol. 2024, 19, 4, 824–838
Click to copy citationCitation copied!
https://doi.org/10.1021/acschembio.3c00805
Published April 3, 2024

Copyright © 2024 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0 .

Abstract

Click to copy section linkSection link copied!

Covalent inhibition has seen a resurgence in the last several years. Although long-plagued by concerns of off-target effects due to nonspecific reactions leading to covalent adducts, there has been success in developing covalent inhibitors, especially within the field of anticancer therapy. Covalent inhibitors can have an advantage over noncovalent inhibitors since the formation of a covalent adduct may serve as an additional mode of selectivity due to the intrinsic reactivity of the target protein that is absent in many other proteins. Unfortunately, many covalent inhibitors form irreversible adducts with off-target proteins, which can lead to considerable side-effects. By designing the inhibitor to form reversible covalent adducts, one can leverage competing on/off kinetics in complex formation by taking advantage of the law of mass action. Although covalent adducts do form with off-target proteins, the reversible nature of inhibition prevents accumulation of the off-target adduct, thus limiting side-effects. In this perspective, we outline important characteristics of reversible covalent inhibitors, including examples and a guide for inhibitor development.

This publication is licensed under

CC-BY-NC-ND 4.0 .
  • cc licence
  • by licence
  • nc licence
  • nd licence
Copyright © 2024 The Authors. Published by American Chemical Society

Special Issue

Published as part of ACS Chemical Biology virtual special issue “Exploring Covalent Modulators in Drug Discovery and Chemical Biology”.

Introduction

Click to copy section linkSection link copied!

Covalent inhibition is an effective way to limit the function of an enzyme. This mechanism has been leveraged by many organisms to limit the growth of competitors vying for resources (e.g., beta-lactams from the fungal genus Penicillium) (1) or to fight infections (e.g., itaconate from mammalian macrophages (2)). The warheads of these covalent inhibitors are sometimes obvious, such as the strained ring of beta-lactams or the α,β-unsaturated carboxylic acid of itaconic acid, while some have subtle mechanisms of covalent inhibition that are not apparent by looking at the functional groups of the inhibitor (e.g., 3-nitropropionate as an inhibitor of isocitrate lyase (3) or wortmannin as a phosphoinositide-3 kinase inhibitor (4)) (Figure 1). The presence of numerous different covalent mechanisms of inhibition in nature suggests that the benefits of producing reactive electrophilic chemicals as a means of survival are, on the whole, more favorable than the potential off-target effects they may cause.

Figure 1

Figure 1. Structures of Penicillin G, 3-nitropropionate, itaconate, and wortmannin are examples of natural product covalent inhibitors with their covalent adducts.

In drug-development, we aim to minimize the extent of side-effects and maximize the therapeutic potential of drugs. Since covalent inhibitors typically contain electrophilic groups, there was considerable concern about indiscriminate reactivity, since that typically meant irreversible inhibition of enzymes (and off-target proteins) which could lead to severe side-effects. (5) Additionally, compounds (or groups within the compound) that upon metabolism bear reactive groups have been avoided for concern of toxicity. (6) Furthermore, concerns about drug–protein adducts or degradation products leading to immune activation (e.g., allergic reaction) have limited the desire to design covalent inhibitors. (7) Consequently, the gold standard for drug-development was in reversible (i.e., noncovalent) inhibitors. (5) Despite this, there have been numerous drugs approved and still in use that are covalent inhibitors, or are prodrugs which are metabolized to become the active inhibitor (such as omeprazole). (8) Additionally, many noncovalent inhibitors have been removed from the market due to toxicity. As such, the well-warranted concerns of covalent inhibition can be overcome with diligence toward expected hurdles and clever design.

Design of Noncovalent and Covalent Inhibitors

Click to copy section linkSection link copied!

The approach for developing noncovalent inhibitors (NCIs) is to design compounds that strongly bind to our target protein, bind significantly weaker to other proteins, and rely on dynamic equilibria leading to the most thermodynamically stable product (i.e., drug binding to intended target). This relies on the law of mass action, which describes the equilibrium of reactants as a function of their concentrations and activities. Indeed, this has been thoroughly described with a discussion of assumptions and models. (9) However, this still leads to unanticipated issues in drug-development for side-effects since small molecules bind to numerous different proteins and enzymes, even if there is little homology in structures. (10) This suggests that there needs to be additional mechanisms of discrimination to prefer binding of the target protein and limit off-target binding. Despite previously addressed concerns, there are advantages available to covalent inhibitors that are not found in NCIs. Indeed, leveraging these advantages has led to a resurgence of covalent inhibitors. (5) One such advantage is the prolonged residence time of the inhibitor in the target protein. (11) A second is the ability to inhibit “undruggable” targets. (12) Some characteristics of “undruggable” targets include those with an “extended or shallow pocket, or no pocket at all” and targets that engage in protein–protein interactions (PPI). (12) RCIs have seen success in inhibiting such “undruggable” targets such as Mcl-1 (which engaged in PPI) wherein a surface lysine residue forms a covalent adduct with the arylboronic acid of the RCI, which interferes in complex formation. (13) Additionally, RCIs have been successful in engaging with targets for which it may not be possible to develop NCIs that discriminate between highly homologous active sites of different enzymes (e.g., KRAS inhibitors). (14) A third is the 2-fold mode for protein selectivity: one as binding in the same fashion as NCIs, and two by forming a bond with a reactive amino acid residue to deactivate the target either by suicide inhibition─irreversible deactivation of the catalytic residues─or increased binding affinity due to forming a covalent adduct resulting in strong competitive inhibition. The incorporation of a covalent warhead improves the “stickiness” of the inhibitor for the target protein. The reaction between the inhibitor and target may be irreversible or reversible. As the name suggests, irreversible covalent inhibitors (ICIs) form covalent adducts that permanently alter the target, which is akin to adhering the inhibitor to the target with glue. Conversely, reversible covalent inhibitors (RCIs) form covalent adducts that do not permanently alter the target, analogous to sticking the inhibitor to the target with Velcro. This is summarized in Figure 2, where NCIs have a single binding equilibrium with on and off kinetics, while ICIs and RCIs have two steps, the second of which is the covalent adduct formation. In ICIs, this second step is inactivation, whereas in RCIs, this second step is also in equilibrium.

Figure 2

Figure 2. Schematic of NCI, ICI, and RCI inhibition with descriptions of the equilibrium or inactivation constants (Mons et al.). Adapted from ref (15). Available under a CC-BY 4.0 license. Copyright 2024 Elma Mons, Sander Roet, Robbert Q. Kim, Monique P. C. Mulder.

Incorporating the law of mass action into drug design

Click to copy section linkSection link copied!

By accepting off-target engagements as inevitable, we can introduce functionality into inhibitors that can address off-target issues. A method of doing this is to deliberately introduce functionality that reduces the level of target binding. Specifically, by designing a covalent inhibitor whose reaction kinetics allow for reversibility, permanent modification of off-target adducts can be mitigated. ICIs will react with unintended targets, which then results in off-target effects. The irreversible nature of ICIs results in permanent modification of the unintended target, which will persist until it is degraded. On the other hand, RCIs contain electrophilic warheads that have good reversible reaction kinetics allows for off-target reactions to occur, but then reverses to liberate the inhibitor. Ideally, an inhibitor would be designed for the target specifically in which it should have strong binding kinetics for the target, and weak binding kinetics for the formation of off-target covalent adducts. It is possible to design an inhibitor to have good on-target engagement, but it is not possible to design an inhibitor that has poor off-target engagement. By incorporating a mechanism of disengagement with off-target proteins, it is expected that off-target covalent inhibition diminishes over time. Conceptually, this suggests considering reaction equilibria, or on/off kinetics which returns us to the law of mass action. By deliberately considering the reaction kinetics with off-target proteins and reaction kinetics with the target protein, we can consider the formation of the desired “product” (inhibitor–target protein adduct) as a result of a series of kinetic equilibria.
Simplified, an RCI may reversibly react with many other substrates, but since the formation of desired product is highly favored, the result of all equilibria eventually leads to the formation of the desired product (Figure 3).

Figure 3

Figure 3. Inhibitor is in equilibrium with many enzymes, including the target enzyme. Since the inhibitor–target complex is greatly favored, this will shift the equilibria of the off-target interactions away from the off-target complexes with the inhibitor as described by Le Chatalier’s principle. Over time, the inhibitor–target complex will be the predominant site of the RCI.

This binding affinity should contribute more to Ki* (* designates a covalent adduct) than the propensity of adduct formation in order to favor formation of the intended inhibitor–target complex over off-target complexes. Additionally, it is worth noting that since Ki* are in terms of enzyme concentration, direct comparison of Ki* between the target protein or enzyme and off-target proteins or enzymes may be misleading since expression levels may differ by orders of magnitude. The overall steady-state equilibrium constant Ki* and target residence time (τ) is described in Figure 4. (15)

Figure 4

Figure 4. The observed equilibrium of a covalent inhibitor is the product of the binding equilibrium constant and the reaction equilibrium constant.

Since the goal in designing an inhibitor is for the compound to interact with the target enzyme to prevent substrate turnover, it is important not only for the inhibitor to bind strongly but to occupy the enzyme for a prolonged period to prevent substrate binding. Additionally, since we are considering steady-state kinetics with all other nucleophiles our inhibitor may form adducts with, the Ki* should be minimized and τ should be maximized. A comprehensive review and tutorial performing kinetic experiments with covalent inhibitors has been described by Mons et al. (15)

Modeling in covalent inhibitor design

Click to copy section linkSection link copied!

As interest in developing covalent inhibitors has increased in recent years, so has computational modeling to better understand and predict reactivity and selectivity. (16) Density Functional Theory (DFT) is a particularly helpful tool to assess the reactivity of electrophile and protein nucleophilicity to predict covalent reactions, such as a reaction between a kinase serine and a covalent inhibitor. (17) It is important to know if the target of interest is amenable to covalent inhibition before spending extensive efforts on synthesis and protein purification to learn that the nucleophilic residue (e.g., a catalytic serine) does not react sufficiently well with the warhead chosen for the inhibitor. Molecular modeling can allow for the incorporation of the appropriate warhead prior to embarking on synthetic efforts. (18) Indeed, Plescia et al., through computational modeling and kinetic experiments, found that a boronic acid warhead was more well-suited for inhibition of prolyl oligopeptidase than the typical nitrile warhead. (18) Recently, there have been increased efforts in designing models to describe the necessary conditions for reversible covalent inhibition to occur. Masuda et al. used the Gibbs free energy of hydrolytic water molecules to interrogate the expected rate of reaction for hydrolysis of the covalent adduct. Despite using training set and test set compounds that would more appropriately be described as competitive substrates, the model still provides useful information demonstrating that (intuitively) both the lower Gibbs free energy of water and the increased activation energy for bond-cleavage lead to a slower reverse reaction of the covalent adduct. In their work, the entropic component of the Gibbs free energy equation (-TΔS) correlates well with kcat, suggesting that their model utilizing the Gibbs free energy of water and the activation energy of covalent–adduct bond cleavage may be effective parameters by which to discriminate covalent inhibitors. (19) Efforts by Chatterjee et al. have been to probe free energy perturbations using quantum mechanics/molecular mechanics (QM/MM) calculations to predict binding affinities and binding kinetics for warheads and their target enzymes. They found that using this approach, when covalent binding was 5.5 kcal/mol stronger than noncovalent binding, selectivity could be predicted by the relative free binding energy of the covalent state. (16) An extensive review on the details of covalent inhibition modeling (irreversible and reversible) has recently been published by Awoonor-Williams et al. (20) Additionally, Scarpino et al. have described detailed protocols for screening large libraries of warheads with selected enzyme targets to model binding mode predictions, single-warhead virtual screening, and multiwarhead virtual screening. (21) For further reading, a detailed review of the application of computational methodologies to covalent inhibitor design has been described by De Cesco et al. (17)

Chemical characteristics of reversible covalent warheads

Click to copy section linkSection link copied!

There have been numerous reversible covalent warheads developed to mediate the off-target effects found in ICIs. (22) The warhead should be selected for its reactivity for the intended amino acid to react with, as different warheads have different reactivities to different amino acids (23) in addition to well-modeled predictions of how reactive the warhead is for the intended amino acids. (24) Jackson et al. have a highly thorough review of α,β-unsaturated carbonyls with thiols. (25) As expected, stronger Michael-acceptors such as α,β-unsaturated aldehydes, ketones, esters, and acid chlorides react quite rapidly, whereas α,β-unsaturated amides and carboxylic acids react significantly less so. The substituents of the amides do greatly affect the reactivity with thiols in which an N-(2-pyridyl) substituent has a half-life with glutathione of approximately 8 min, whereas an N-methyl substituent has a half-life of approximately 17 h. (25) Mechanistic studies performed by the Keillor group on model acrylamide substrates found that the nucleophilicity of the thiolate in question was not a significant contributor to reaction rates. (26,27) Combined, this suggests that when targeting cysteine residues, the intrinsic reactivity is dictated by the electrophile being designed. This is not to advocate neglect of the nature of the cysteine residues being targeted. For example, the reactivity of catalytic cysteine residues can be enhanced through methods such as “desolvation,” which destabilizes the thiolate anion and thus increases its nucleophilicity. (28) Nuances into the reactivity of cysteine residues are thoroughly reviewed by Parvez et al. (29)
Extensive efforts have been made by the Taunton group in tuning the reversibility and the residence times of RCIs. Using the cyano-acrylamide group warhead as a model, Krishnan et al. changed the amide into a variety of heteroaromatic groups. (30) They had then plotted the computed proton affinity of the α-carbanion (relative difference from the parent cyano-acrylamide) versus the rate of elimination. They found a linear relationship in which the greater the proton-affinity, the slower the rate of elimination. (30) This provides evidence of an intuitive assumption in which more acidic protons (lower pKa and lower proton affinity) in the α-position would lead to a more rapid elimination. Therefore, covalent adducts would be less stable, and lead to greater reversibility. (30) Additionally, there can be steric contributions that limit the accessibility of the acidic α-protons, which then reduces the rate of elimination. (31) In efforts to tune residence time, Bradshaw et al. introduced substitutions at the β-position of the cyanoacrylamides. The introduction of bulky β-substitutions did not affect greatly affect inhibition of their enzyme target, but could extend the residence time from minutes to days. (32) By tuning the reactivity of the warhead, the reverse reactivity (elimination), and the residence time, RCIs can mimic the on-target benefits of ICIs while circumventing issues associated with off-target reactivity.
Although not as thoroughly characterized as substituted acrylamides, tunability of boronates and nitriles have been described with postulating that varying the boronate pKa may be optimized for target engagement, (33) and that increasing the electron withdrawing groups bound to the nitrile improves electrophilicity (and therefore reactivity). (34)
Despite many warheads not having as much depth for kinetic and mechanistic studies for tuning reactivity for the purpose of designing RCIs, there are many that have been designed to leverage reactivity toward a particular target amino acid residue, such as cysteine, serine/threonine, or lysine, as seen in Figure 5. The examples include reversible warheads for cysteine such as α-cyanocarbonyls (35) or other α-substituted alkenes with good electron withdrawing groups, (30,36) nitriles, (37) aldehydes, (38) and dihaloacetamides (39) (although the halogens are lost, and the equilibrium is between the free cysteine and 2-oxoacetamide), and thiomethyltetrazines; (40) reversible warheads for serine/threonine include α-oxo-amides (41) and boronic acids; (42) and reactive amines such as lysines and N-terminus amines react with aldehydes to form stabilized imines, (43) either by a neighboring hydrogen bond donor, or through subsequent formation of a stable diazaborine. (44) Example compounds for each of these warheads can be seen in Figure 6. The incorporation of reversible warheads into drug design has led to numerous successes (22) with selected examples highlighting the advantages RCIs have over ICIs, and situations where RCIs are particularly effective.

Figure 5

Figure 5. Nucleophilic residues reacting with electrophilic warheads in equilibrium with their covalent-adduct products, with the electrophilic warheads highlighted in red.

Figure 6

Figure 6. Examples of reversible covalent inhibitors with their electrophilic warheads are highlighted in red.

Biochemical analysis of reversible covalent warheads

Click to copy section linkSection link copied!

After designing and synthesizing the RCIs, validating their reversibility, determining activity, measuring residence time, and evaluating off-target effects require numerous techniques. Confirming reversibility is a relatively easy to address. Using nuclear magnetic resonance (NMR) techniques, the chemical signal for adducts versus the unbound RCI may be quite distinct. By diluting the NMR sample of the covalent adduct, the system equilibrates which reduces the adduct concentration, providing greater unbound RCI. (45) Additionally, the Kd can also be determined through this method if the adduct or RCI has distinct UV/vis spectroscopic signals. (45) Mass spectrometry (MS) can also provide evidence for reversibility in which the covalent adduct is injected into the mass spectrometer, but over the course of the MS experiment, dissociation occurs and the molecular ion of the inhibitor, (31) or the native protein is liberated and observed on the MS spectrum. (46) Competitive inhibition assays utilizing bioluminescent resonance energy transfer (BRET) have also been used in combination with washout experiments in which the protein was saturated with the RCI, then a fluorescent tracer was introduced which replaced the RCI, demonstrating reversibility and binding kinetics of the RCI. (46) Additionally, degradation or denaturation of the RCI-protein adduct followed by recovery of the RCI provides further evidence for reversibility. (32)
The residence time of RCIs can be measured through fluorescence competition assays in vitro or through pharmacokinetic/pharmacodynamic (PK/PD) analyses. Bradshaw et al.─in their efforts to tune residence times of RCIs─they measured the residence time in cells via competition assays in which they incubated Bruton tyrosine kinase (BTK) with an inhibitor followed by washing to removed excess inhibitor, then incubated with a fluorescently labeled inhibitor. Measuring the fluorescence of the labeled inhibitor over time then is used to determine the residence time, and % of BTK bound over time. (32) Residence time measurements in vivo can be performed by dosing an animal model with the RCI, then harvesting the spleen at certain time points (e.g., 1, 12, and 24 h) followed by competition with an irreversible fluorescent probe and measuring the percent occupancy of the RCI. (35)
Evaluating off-target effects is particularly important considering the concerns of toxicity for covalent inhibitors. (6) Classical methods of determining what proteins were forming adducts with the covalent inhibitors involved incubation with a radiolabeled inhibitor, followed by gel chromatography, electrophoresis, excision from the gel and submitted to Edman sequencing to identify the amino acid-inhibitor adduct. (6,47,48) More modern methods include MS methodologies in which a protein of interest is analyzed after isolation (e.g., chromatography, electrophoresis) (49) for covalent modification (without protein digestion if sites of modification are known, after digestion if unknown). (49) Chemical proteomics is another method to probe the off-target reactivity of a covalent inhibitor. By tagging the RCI with a label (e.g., affinity, fluorescent, or radioactive) the adduct may be isolated and analyzed (such as purification from affinity columns using affinity labels, or excising from gels using fluorescent or radioactive labels). (50) These screens have been used to identify target compounds in drug discovery, but could also be applied to identify off-target proteins. (50) Similar principles have been applied to find reactive lysine and cysteine residues. (51−53) It is important to recognize that the presence of a covalent adduct does not necessarily mean that there is an adverse effect associated with it. Identifying off-target adducts can provide evidence to support hypotheses associated with side-effects, but the role of the off-target adduct must be explored further if unknown. (49)
Global stress responses in cells may also result from covalent inhibitors. This may be caused by reactivity with glutathione, which can cause electrophilic stress responses in cells. (54) Performing competition reactions of the protein target and glutathione with RCIs (31) in addition to monitoring for glutathione depletion in cells (55,56) can provide helpful information on the effect of RCIs on electrophilic stress in cells. Furthermore, covalent inhibitors may induce misfolding of proteins which could lead to a global protein misfolding profiles and responses. (57,58)
Additionally, T-cell activation due to chemically reactive intermediates are not necessarily identifiable through MS methods. (49) These can occur via the formation of drug haptens (covalent adducts of proteins) which subsequently induce T-cell-mediated drug-hypersensitivity reactions. (59) Additionally, there may be covalent interactions with the T-cell receptor or human leukocyte antigen (HLA) peptides which ultimately results in a T-cell response. (59) β-lactams are known to form haptens, which are responsible for penicillin allergies. (60) Unfortunately, drug-hypersensitivity reactions cannot be predicted, and remains an active field of research to critical antigens responsible for T-cell activation with respect to drug–protein adducts (59) or the nuances of T-cell activation with major histocompatibility complexes associated with drug-peptide adducts. (61)

Development of highly selective JAK3 inhibitors

Click to copy section linkSection link copied!

Janus kinases (JAK) are critical regulatory enzymes that mediate cell signaling responses, especially within the secretion of cytokine signals which are implicated in immune responses. (62) In humans, there are four subtypes (JAK1, JAK2, JAK3, and TYK2), each having high similarity. (63) Consequently, developing highly selective drugs to modulate the activity of one JAK over another is extremely challenging. Clinical success has been observed in developing pan-JAK inhibitors, with the FDA approval of tofacitinib, but the effects of inhibiting multiple JAKs is unclear, especially since different JAKs play diverse roles in immune health. (63) Goedken et al. aimed to leverage a unique cysteine residue on JAK3 (Cys909), which would confer unique reactivity relative to JAK1, JAK2, and TYR2 (which contain serine at the equivalent position) which makes covalent inhibition a particularly attractive approach. (63,64) Using a small collection of newly developed ICI (13), a known JAK3 inhibitor thought to be an ICI (4), and two previously known noncovalent JAK3 inhibitors (5 and 6) (Figure 7), Goedken et al. characterized and determined that the newly developed ICIs had excellent activity (low nM) and selectivity (>500×) for JAK3 versus other JAK kinases. (63) Additionally, for other highly similar kinases with cysteine residues at the equivalent position as JAK3, there was also high selectivity (>100×). (63) Although compounds 13 were found to have good IC50 values in vitro, this obscures the problem that if the covalent warhead were to react off-target before reaching JAK3, the result would be an off-target, irreversible adduct that could lead to adverse effects. By building off of the work done targeting Cys909 of JAK3, Forster et al. developed new inhibitors using the same tricyclic scaffold (7), but making modifications in which the covalent warhead was replaced with a cyano-acrylamide group, which is a good reversible Michael-acceptor (Figure 6). (46,65) The cyano-acrylamide containing compounds were validated to be reversible inhibitors using washout experiments utilizing BRET in addition to MS where adduct dissociation was observed. (46) Direct comparisons of the Michael-acceptors (9 and 10) versus their saturated equivalents (11 and 12) were made, and it was found that there was a general trend of a higher IC50 (upward of 4× higher for the saturated compounds). (66) This suggests that there are significant noncovalent interactions that the compounds are making with the target enzyme. Forester et al. found that their acrylamide-containing compounds (5 and 8, which form irreversible adducts) had good activity (292 nM and 22 pM), but low selectivity between the different JAK kinases, whereas the cyano-acrylamide-containing compounds (13 and 14, which form reversible adducts) also had good activity (127 pM and 154 pM) in addition to having high selectivity for JAK3 versus JAK1, JAK2, and TYR2 ranging from 400× to 5800× selectivity. (46) In an in vitro assay for selective inhibition of JAK3, the pan-JAK kinase inhibitor (5) was found to inhibit multiple JAK kinases, whereas the RCIs were found to be selective for JAK3. (46) The next generation of the JAK3 RCIs was to optimize the compound for selectivity, activity, and explore structure–activity relationships (SAR). (66) Significant efforts went into modifications at the position of the Michael-acceptor. The next steps for these inhibitors have been to improve pharmacokinetics and metabolic stability utilizing the cyano-acrylamide warhead. (67)

Figure 7

Figure 7. Examples of JAK3 inhibitors with covalent warheads are highlighted in red.

Development of Bruton Tyrosine Kinase inhibitors for immune diseases

Click to copy section linkSection link copied!

BTK is a critical enzyme in immune regulatory responses related to antibody signaling and immune cell recruitment and is expressed in most hematopoietic cells. (68) BTK is critical in signaling cascades in B cells, including the activation of NF-κB, (69) which is essential for regulating B cell activation and development. (70) Dysregulation of the BTK signaling pathway can lead to constitutive activation of NF-κB, and subsequent “addiction” to NF-κB by many cancer types. (71) Consequently, BTK is a particularly important therapeutic target for immune system diseases such as mantle cell lymphoma, chronic lymphocytic leukemia, Waldenstrom macroglobulinemia, marginal zone lymphoma, and chronic graft-versus-host disease, for which BTK inhibitors have been approved or are in development. (35) BTK contains a cysteine residue where analogous enzymes contain a serine, allowing for reactivity-based selectivity, which makes for a good candidate for covalent inhibition. (12) Ibrutinib, acalabrutinib, zanubrutinib, and tirabrutinib (Figure 8) are all approved drugs that inhibit BTK through an irreversible reaction with Cys481. (72−75) These drugs are highly potent inhibitors with low to subnanomolar IC50 values against BTK. (75,76) Despite their high potencies for BTK, kinases that have analogous cysteine residues to Cys481 are also prone to irreversible inhibition. Selectivity indices for ibrutinib show low to modest selectivity for BTK versus a panel of other related tyrosine kinases (0.36–117× selectivity), the second-generation BTK inhibitors acalabrutinib and tirabrutinib have greater selectivity generally, but still modest (0.88 to >8000×). (75) Due to the irreversible nature of the binding, potent off-target inhibition can lead to side-effects including atrial fibrillation, diarrhea, and bleeding significant enough to discontinue treatment (upward of 20–25% of patients). (77−80) Langrish et al. describe the reversible BTK inhibitor rilzabrutinib’s biochemical and safety profile. (35) Rilzabrutinib (Figure 8) contains a cyano-acrylamide group that serves as the reversible warhead and was validated to be an RCI by recovery of the inhibitor following BTK-adduct digestion by trypsin in addition to quantifying the kon (5.1 ± 2.1 × 104 M–1 s–1) and the koff (1.2 ± 0.1 × 10–6 s–1). (35) It was evaluated against 251 kinases from diverse families, with 6 (including BTK) having inhibition at >90% at 1 μM. (35) Residence time of rilzabrutinib was compared for 5 of these kinases, and only BTK was found to have an occupancy rate of over 80% during a 24-h incubation (35) suggesting that despite potent binding of rilzabrutinib to other kinases, it will dissociate from the off-target kinases better than BTK, suggesting that side-effects related to off-target binding may be limited due to low residence time. Langrish et al. performed a series of assays to evaluate the on- and off-target effects of rilzabrutinib. They determined that it had low cytotoxicity (>16,000 nM) in non BTK-expressing cell lines, did not inhibit T-cell receptor-induced activation or calcium flux-induced activation of T-cells (>5000 nM), and did not interfere in EGFR signaling. (35) Additionally, unlike ibrutinib, rilzabrutinib does not reduce normal platelet aggregation (35) which leads to normal clotting (which is a significant problem with ibrutinib). (77,78) Langrish et al. also found that there was durable inhibition of BTK in rats where BTK occupancy was ∼90% after 14 h, while the serum concentration of rilzabrutinib had fallen to <3 ng/mL (compared to nearly 500 ng/mL after 1 h) at 40 mg/kg dosing. This suggests that short exposure time and low systemic exposure is required for effective inhibition which reduces the chance of side-effects. (35) Rilzabrutinib is currently being evaluated in Phase III trials for adults and adolescents with persistent or chronic immune thrombocytopenia, and numerous Phase II clinical trials for immune disorders such as warm autoimmune hemolytic anemia, atopic dermatitis, asthma, IgG4-related disease, chronic spontaneous urticaria, and immune thrombocytopenia. (81)

Figure 8

Figure 8. Clinically approved irreversible covalent inhibitors for BTK: ibrutinib, Zanubrutinib, acalabrutinib, and tirabrutinib, and a reversible covalent inhibitor for BTK in clinical trials: rilzabrutinib. The covalent warhead is highlighted in red.

Development of inhibitors for proteases

Click to copy section linkSection link copied!

Proteases fall into seven categories based on their catalytic residues: aspartic, cysteine, serine, metallo, threonine, glutamic, and asparagine. (82) These proteases are found across all domains of life. As a consequence, the effort to inhibit proteases has been applied in anticancer, antimicrobial, hypertension, type 2 diabetes, Alzheimer’s disease, nonalcoholic fatty liver disease, and fibrosis therapies. (83) Since protease categories are mechanistically discrete due to their catalytic residues, RCIs are an effective tool for selective inhibition. Clinical success has been seen with RCI inhibition (22) including bortezomib and ixazomib (threonine protease inhibitors), (84) boceprevir, saxaglibtin, and telaprevir (serine protease inhibitors), (85−87) and nirmatrelvir (cysteine protease inhibitor). (88) A recent example for the development of a cysteine protease inhibitor was performed by Ma et al. (31) to treat enterovirus 71 (EV 71)─one of the causative agents of hand, foot, and mouth disease. (89) Ma et al. aimed to synthesize an inhibitor for the 3C protease with steady-state kinetics (i.e., rate of reaction and elimination of covalent warhead) residence time of the inhibitor as explicit parameters to tune in designing the optimal cysteine protease inhibitor (Figure 9). (31) Using the scaffold of rupintrivir (an ICI for human rhinovirus, which is a homologue of EV71), modifications were made to improve selectivity for EV71 3C protease, resulting in α-ketoamide (90) (15) and aldehyde-based (91) (16) warheads.

Figure 9

Figure 9. Compound optimization for the EV71 cPro enzyme. The covalent warhead is highlighted in red.

These warheads were likely responsible for poor potency due to off-target reactivity and to address these issues, Ma et al. introduced the cyano-acrylamide group, and modified the amide substitutions to tune reactivity, reversibility, and selectivity. (31) The reversibility of the series of compounds were validated by MS, through fitting kinetic data, and measuring enzyme activity after dialysis. When the covalent adduct was submitted to MS experimentation, the spectra showed both the adduct and the free protease, suggesting reversibility. Different inhibitors were found to have different ratios of the adduct to free enzyme, suggesting different rates of the reverse reaction. The fitting of the kinetic data supported a two-step mechanism, and the dialysis experiments showed that the enzyme would recover activity with increased dialysis time, suggesting that the RCI was disengaging with the protease. (31) All of these supported that the cyano-acrylamide inhibitors behave as RCIs.
To screen the SAR, Ma et al. made many modifications to the warhead. By introducing an α,β-unsaturated ester (17) or amide (18), they found moderate inactivation of 3C protease, and poor EC50 values (>100 000 nM). Introducing halogens as α-substituents to the conjugated esters (19 and 20) led to medium to high-nanomolar Ki* and EC50 values but with highly cytotoxic activity (100% at 100 μM). Substituting the halogens with a nitrile removed cytotoxicity and reduced the Ki* and EC50 values to low to midnanomolar potencies for the cyano-acrylamide (21) and cyano-acrylate ester (22) containing compounds. Interestingly, tertiary amides (such as 21) led to increased inhibitory activity compared to the secondary amides, likely due to steric shielding of the acidic α-proton, which allows for a decreased rate reverse reaction, which would increase residency time. This observation was consistent with the MS data, in which the bulky tertiary amides did not dissociate as much as the secondary amide.
Importantly, competition assays of 21 with 5 mM additions of nonenzymatic nucleophiles such as glutathione, ethanolamine, and lysine were performed. Glutathione modestly affects the in vitro inhibitor activity for cyano-acrylamides. Although the authors did not expect that there would be a significant change in cellular glutathione depletion due to the high potency of the compounds, and their reversible reactivity, the experiments supporting this claim were absent. Finally, in determining selectivity of the inhibitors, the compounds were screened for activity against chymotrypsin (serine protease), cathepsin K (cysteine protease), and calpain-1 (cysteine protease). None of the compounds evaluated had reactivity toward chymotrypsin, suggesting good selectivity for cysteine proteases (as expected considering serine is a poor Michael-donor), and only one compound had >50% inhibition of cathepsin K, while no compounds had >50% inhibition of calpain-1. (31) This is in contrast with analogous inhibitors that utilized aldehyde and cyanohydrin warheads, which had high degrees of inhibition for both cathepsin K and calpain-1 demonstrating that the nature of the warhead significantly reduces off-target activity. Although the screen provided good preliminary evidence of selectivity for the viral cysteine protease, the counterscreen against human proteases is by no means comprehensive, and the experiment would greatly benefit by incorporating more of the other cathepsins, which differ greatly in their amino acid recognition sequences and would therefore differ in their susceptibility to inhibition.

General considerations for developing reversible covalent inhibitors

Click to copy section linkSection link copied!

To develop an effective RCI, some considerations need to be made. There must be a covalent warhead within the designed inhibitor, there must be some selectivity of the covalent warhead for a particular nucleophile, the covalent adduct must be reversible, and the drug must have good binding affinity to the desired target. A framework for designing an optimal reversible covalent inhibitor is described below, in which the assumption is that a good reversible covalent inhibitor has a universal set of characteristics. These steps are summarized in Figure 10.
1)

Before embarking on a program to develop an RCI for your target of choice, perform computational analyses and in silico modeling to ensure that there is a suitable nucleophile to target (e.g., serine, threonine, lysine, cysteine, N-terminus amine). Computational evidence can help support design of the RCI with the appropriate warhead (18) but is not strictly necessary for a successful drug discovery program (especially if there is little high quality structural data with which to work). There have been many works discussing the theoretical frameworks of modeling covalent inhibitors (92) as well as experimental modeling using QM, QM/MM, or QM/MM coupled with molecular dynamics approaches. (16,93−96)

2)

The Law of Mass Action describes the state of products and reactants at equilibrium. Even though the equilibrium may ultimately favor the formation of desired products, the kinetics of desired adduct formation may be prohibitively slow under conditions amenable to therapeutic usage. Conversely, the reverse reaction for off-target effects may be thermodynamically favored, but if the kinetics of dissociation are slow, then the off-target effects may be significant since there may be a long-residence time of the inhibitor and the off-target enzyme, which leads to no functional difference between reversible inhibition and irreversible inhibition on the time scale of cellular stress response. Although slow off-kinetics improve residence time, faster rates may be more suited in certain circumstances. (32) Extensive reviews on slow-binding kinetics and residence time have previously been written concerning drug-design. (11,97−99) Since the intention for developing reversible covalent inhibitors is to reduce off-target effects by taking advantage of reversible kinetics, the residence times of the inhibitor with its on- and off-target enzymes should be optimized. (35,46) These experiments were performed both in the development of the BTK inhibitor rilzabrutinib (35) and JAK3 RCIs. (46)

3)

The binding kinetics of the inhibitor match that of reversible covalent inhibitors. There has been extensive work in characterizing different mechanisms of enzymatic inhibition with kinetic descriptions. (15) A step-by-step guide for the synthesis and biochemical characterization of reversible inhibitors was well-described by Frühauf et al. for histone deacetylase 4 (HDAC4). (100) This should be able to serve as a good starting point for spectroscopic experiments to biochemically characterize the inhibitor. Alternative methods for demonstrating reversible covalent inhibition have been performed by washing followed by tracer treatment (JAK3 inhibitors), (46) using inhibitor recovery after trypsin digest (rilzabrutinib), (35) and combination of steady-state kinetics and biomolecular mass spectrometry (EV71 C3 protease inhibitors). (31)

4)

There should not be a prolonged buildup of the inhibitor–glutathione adduct. This tripeptide is highly abundant in cells (upward of 10 mM) (101) and is intimately involved in redox metabolism (102−104) and electrophilic stress (54) within the cell. Therefore, there must be high reversibility with glutathione in order to not induce a global electrophilic stress response. Assays to be performed to address this issue include competition experiments, in which the intended enzyme and glutathione are coincubated with the reversible covalent inhibitor (31) and glutathione depletion experiments. (55,56) As described above, in the development of the EV71 C3 protease inhibitors, competition with noncovalent nucleophiles such as GSH were performed. (31)

5)

There should be a minimal effect on cellular stress responses. Covalent inhibition of an enzyme may lead to misfolding of the protein. (57,58) If this is nonspecific, this may lead to global protein misfolding stress responses. Examinations of protein profiles can be performed as previously described, (57) and comparisons with efforts to probe reactive residues of the proteome (51−53,105,106) to ensure limited off-target activity. Additionally, the compound should not induce electrophilic stress responses in the cell. (107,108) By incorporating experiments that assess global stress responses, a greater degree of confidence can be made toward knowing that the reversible covalent inhibitor is, indeed, not having obvious off-target effects within the cell.

6)

Beyond effective target engagement, a significant consideration in developing covalent inhibitors is the effect on immune cells. Immune cells are highly sensitive to electrophilic stresses, which can cause either immunostimulatory or immunosuppressive effects. (109) As such, assaying your inhibitor against B cells, T cells, and macrophages to determine whether the inhibitor causes an expression and secretion of pro-inflammatory or anti-inflammatory cytokines is important to avoid potential complications of immune activation.

Figure 10

Figure 10. Summarized activities for each parameter that are important in the design of a reversible covalent inhibitor.

Conclusion and future directions for reversible covalent inhibitors

Click to copy section linkSection link copied!

There has been a resurgence of covalent inhibitor development in recent years. Technology has improved to be able to interrogate many of the concerns that come with covalent inhibition in drug design. In an effort to profit off of the benefits of covalent inhibition (potent on-target activity via chemical deactivation of the target) while allaying concerns about covalent inhibition (indiscriminate reactivity with nucleophiles in serum or cytosol), one can develop RCIs. This approach leverages the law of mass action, in which the equilibrium of a series of reactions depends on their activities and concentrations. The consequence of this is that at steady-state, the concentration of the products (e.g., a covalent adduct of the target protein) will be dependent upon the equilibrium of all other side-reactions. By designing a strongly binding inhibitor, equilibrium will ultimately lead to inhibition of the desired target with off-target inhibition being disfavored. Although off-target binding will occur, the reversibility of the covalent-adduct will limit the phenotypic consequences of off-target binding. We anticipate that like the examples above wherein moving from irreversible covalent inhibition to reversible covalent inhibition, significant off-target effects were circumvented, leading to potentially a significantly safer drug; the adoption of reversible covalent inhibition will lead to promising drugs with few significant side-effects due to off-target reactivity. Although the focus of this perspective has been in drug-design and enzyme inhibition, reversible covalent adducts have been utilized in cell imaging, (110) and in material science (albeit under the name of dynamic covalent bonds). (111) Future directions for the field will include improving modeling to accurately identify sufficiently nucleophilic noncatalytic residues for adduct-formation, improved rules or guidelines for reactivity and reversibility of reactions of nonacrylamide warheads, and develop robust, easy-to-perform assays to detect unintended immune responses from electrophilic stress. Additionally, discovery of new warheads is always welcome, especially if they can be easily modified with fluorophores or radiolabels to track localization either within cells or within the body.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Author
  • Authors
    • Disha Patel - Department of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, Canada
    • Zil E Huma - Department of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, Canada
  • Author Contributions

    Authors contributed equally to this work. D.P, Z.H, and D.D wrote the manuscript; Z.H and D.D. prepared the figures; all the authors proofread and approved the final submitted manuscript.

  • Notes
    The authors declare no competing financial interest.

Acknowledgments

Click to copy section linkSection link copied!

D.D. acknowledges start-up funds provided by Brock University. We thank F. Hammerer and N. Häggman for their discussions and insights.

Keywords

Click to copy section linkSection link copied!

Covalent adduct

a complex that forms from a covalent bond between the inhibitor and a nucleophilic amino acid

Covalent warhead

an electrophilic functional group that reacts with a nucleophilic amino acid

Glutathione

a three-amino-acid molecule (γ-glutamine-cysteine-glycine) that is involved in oxidative stress responses

Irreversible covalent inhibitor

a small molecular that binds with a therapeutic target (such as a protein) which forms a permanent covalent bond

Noncovalent inhibitor

a small molecular that binds with a therapeutic target (such as a protein) which does not form a covalent bond

Nontarget

the unintended biomolecule to which inhibitors bind

Protease

an enzyme that hydrolyses amides of proteins or polypeptides

Residence time

the amount of time that an inhibitor occupies the binding site of a protein

Reversible covalent inhibitor

a small molecular that binds with a therapeutic target (such as a protein) which forms a nonpermanent covalent bond

Target

the intended biomolecule to which inhibitors bind.

References

Click to copy section linkSection link copied!

This article references 111 other publications.

  1. 1
    Fleming, A. On the Antibacterial Action of Cultures of a Penicillium, with Special Reference to Their Use in the Isolation of B. Influenzae. 1929. Bull. World Health Organ. 2001, 79 (8), 780790
  2. 2
    Duncan, D.; Auclair, K. Itaconate: An Antimicrobial Metabolite of Macrophages. Can. J. Chem. 2022, 100 (2), 104113,  DOI: 10.1139/cjc-2021-0117
  3. 3
    Ray, S.; Kreitler, D. F.; Gulick, A. M.; Murkin, A. S. The Nitro Group as a Masked Electrophile in Covalent Enzyme Inhibition. ACS Chem. Biol. 2018, 13 (6), 14701473,  DOI: 10.1021/acschembio.8b00225
  4. 4
    Yuan, H.; Barnes, K. R.; Weissleder, R.; Cantley, L.; Josephson, L. Covalent Reactions of Wortmannin under Physiological Conditions. Chem. Biol. 2007, 14 (3), 321328,  DOI: 10.1016/j.chembiol.2007.02.007
  5. 5
    De Vita, E. 10 Years into the Resurgence of Covalent Drugs. Future Med. Chem. 2021, 13 (2), 193210,  DOI: 10.4155/fmc-2020-0236
  6. 6
    Baillie, T. A. Drug-Protein Adducts: Past, Present, and Future. Med. Chem. Res. 2020, 29 (7), 10931104,  DOI: 10.1007/s00044-020-02567-8
  7. 7
    Potashman, M. H.; Duggan, M. E. Covalent Modifiers: An Orthogonal Approach to Drug Design. J. Med. Chem. 2009, 52 (5), 12311246,  DOI: 10.1021/jm8008597
  8. 8
    Boike, L.; Henning, N. J.; Nomura, D. K. Advances in Covalent Drug Discovery. Nat. Rev. Drug Discovery 2022, 21 (12), 881898,  DOI: 10.1038/s41573-022-00542-z
  9. 9
    Kenakin, T. The Mass Action Equation in Pharmacology. Br. J. Clin. Pharmacol. 2016, 81 (1), 4151,  DOI: 10.1111/bcp.12810
  10. 10
    Pottel, J.; Levit, A.; Korczynska, M.; Fischer, M.; Shoichet, B. K. The Recognition of Unrelated Ligands by Identical Proteins. ACS Chem. Biol. 2018, 13 (9), 25222533,  DOI: 10.1021/acschembio.8b00443
  11. 11
    Knockenhauer, K. E.; Copeland, R. A. The Importance of Binding Kinetics and Drug-Target Residence Time in Pharmacology. Br. J. Pharmacol. 2023, 114,  DOI: 10.1111/bph.16104
  12. 12
    Zhang, G.; Zhang, J.; Gao, Y.; Li, Y.; Li, Y. Strategies for Targeting Undruggable Targets. Expert Opin. Drug Discovery 2022, 17 (1), 5569,  DOI: 10.1080/17460441.2021.1969359
  13. 13
    Akçay, G.; Belmonte, M. A.; Aquila, B.; Chuaqui, C.; Hird, A. W.; Lamb, M. L.; Rawlins, P. B.; Su, N.; Tentarelli, S.; Grimster, N. P.; Su, Q. Inhibition of Mcl-1 through Covalent Modification of a Noncatalytic Lysine Side Chain. Nat. Chem. Biol. 2016, 12 (11), 931936,  DOI: 10.1038/nchembio.2174
  14. 14
    Huang, L.; Guo, Z.; Wang, F.; Fu, L. KRAS Mutation: From Undruggable to Druggable in Cancer. Signal Transduct. Target. Ther. 2021, 6 (1), 386,  DOI: 10.1038/s41392-021-00780-4
  15. 15
    Mons, E.; Roet, S.; Kim, R. Q.; Mulder, M. P. C. A Comprehensive Guide for Assessing Covalent Inhibition in Enzymatic Assays Illustrated with Kinetic Simulations. Curr. Protoc. 2022, 2 (6), e419  DOI: 10.1002/cpz1.419
  16. 16
    Chatterjee, P.; Botello-Smith, W. M.; Zhang, H.; Qian, L.; Alsamarah, A.; Kent, D.; Lacroix, J. J.; Baudry, M.; Luo, Y. Can Relative Binding Free Energy Predict Selectivity of Reversible Covalent Inhibitors?. J. Am. Chem. Soc. 2017, 139 (49), 1794517952,  DOI: 10.1021/jacs.7b08938
  17. 17
    De Cesco, S.; Kurian, J.; Dufresne, C.; Mittermaier, A. K.; Moitessier, N. Covalent Inhibitors Design and Discovery. Eur. J. Med. Chem. 2017, 138, 96114,  DOI: 10.1016/j.ejmech.2017.06.019
  18. 18
    Plescia, J.; De Cesco, S.; Patrascu, M. B.; Kurian, J.; Di Trani, J.; Dufresne, C.; Wahba, A. S.; Janmamode, N.; Mittermaier, A. K.; Moitessier, N. Integrated Synthetic, Biophysical, and Computational Investigations of Covalent Inhibitors of Prolyl Oligopeptidase and Fibroblast Activation Protein α. J. Med. Chem. 2019, 62 (17), 78747884,  DOI: 10.1021/acs.jmedchem.9b00642
  19. 19
    Masuda, Y.; Yoshida, T.; Yamaotsu, N.; Hirono, S. Linear Discriminant Analysis for the in Silico Discovery of Mechanism-Based Reversible Covalent Inhibitors of a Serine Protease: Application of Hydration Thermodynamics Analysis and Semi-Empirical Molecular Orbital Calculation. Chem. Pharm. Bull. (Tokyo) 2018, 66 (4), 399409,  DOI: 10.1248/cpb.c17-00854
  20. 20
    Awoonor-Williams, E.; Walsh, A. G.; Rowley, C. N. Modeling Covalent-Modifier Drugs. Biochim. Biophys. Acta Proteins Proteomics 2017, 1865 (11), 16641675,  DOI: 10.1016/j.bbapap.2017.05.009
  21. 21
    Scarpino, A.; Ferenczy, G. G.; Keserű, G. M. Binding Mode Prediction and Virtual Screening Applications by Covalent Docking. Methods Mol. Biol. Clifton NJ. 2021, 2266, 7388,  DOI: 10.1007/978-1-0716-1209-5_4
  22. 22
    Faridoon; Ng, R.; Zhang, G.; Li, J. J. An Update on the Discovery and Development of Reversible Covalent Inhibitors. Med. Chem. Res. Int. J. Rapid Commun. Des. Mech. Action Biol. Act. Agents 2023, 32 (6), 10391062,  DOI: 10.1007/s00044-023-03065-3
  23. 23
    Martin, J. S.; MacKenzie, C. J.; Fletcher, D.; Gilbert, I. H. Characterising Covalent Warhead Reactivity. Bioorg. Med. Chem. 2019, 27 (10), 20662074,  DOI: 10.1016/j.bmc.2019.04.002
  24. 24
    Péczka, N.; Orgován, Z.; Ábrányi-Balogh, P.; Keserű, G. M. Electrophilic Warheads in Covalent Drug Discovery: An Overview. Expert Opin. Drug Discovery 2022, 17 (4), 413422,  DOI: 10.1080/17460441.2022.2034783
  25. 25
    Jackson, P. A.; Widen, J. C.; Harki, D. A.; Brummond, K. M. Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition Reactions. J. Med. Chem. 2017, 60 (3), 839885,  DOI: 10.1021/acs.jmedchem.6b00788
  26. 26
    Watt, S. K. I.; Charlebois, J. G.; Rowley, C. N.; Keillor, J. W. A Mechanistic Study of Thiol Addition to N-Phenylacrylamide. Org. Biomol. Chem. 2022, 20 (45), 88988906,  DOI: 10.1039/D2OB01369J
  27. 27
    Watt, S. K. I.; Charlebois, J. G.; Rowley, C. N.; Keillor, J. W. A Mechanistic Study of Thiol Addition to N-Acryloylpiperidine. Org. Biomol. Chem. 2023, 21 (10), 22042212,  DOI: 10.1039/D2OB02223K
  28. 28
    Lohman, D. C.; Edwards, D. R.; Wolfenden, R. Catalysis by Desolvation: The Catalytic Prowess of SAM-Dependent Halide-Alkylating Enzymes. J. Am. Chem. Soc. 2013, 135 (39), 1447314475,  DOI: 10.1021/ja406381b
  29. 29
    Parvez, S.; Long, M. J. C.; Poganik, J. R.; Aye, Y. Redox Signaling by Reactive Electrophiles and Oxidants. Chem. Rev. 2018, 118 (18), 87988888,  DOI: 10.1021/acs.chemrev.7b00698
  30. 30
    Krishnan, S.; Miller, R. M.; Tian, B.; Mullins, R. D.; Jacobson, M. P.; Taunton, J. Design of Reversible, Cysteine-Targeted Michael Acceptors Guided by Kinetic and Computational Analysis. J. Am. Chem. Soc. 2014, 136 (36), 1262412630,  DOI: 10.1021/ja505194w
  31. 31
    Ma, Y.; Li, L.; He, S.; Shang, C.; Sun, Y.; Liu, N.; Meek, T. D.; Wang, Y.; Shang, L. Application of Dually Activated Michael Acceptor to the Rational Design of Reversible Covalent Inhibitor for Enterovirus 71 3C Protease. J. Med. Chem. 2019, 62 (13), 61466162,  DOI: 10.1021/acs.jmedchem.9b00387
  32. 32
    Bradshaw, J. M.; McFarland, J. M.; Paavilainen, V. O.; Bisconte, A.; Tam, D.; Phan, V. T.; Romanov, S.; Finkle, D.; Shu, J.; Patel, V.; Ton, T.; Li, X.; Loughhead, D. G.; Nunn, P. A.; Karr, D. E.; Gerritsen, M. E.; Funk, J. O.; Owens, T. D.; Verner, E.; Brameld, K. A.; Hill, R. J.; Goldstein, D. M.; Taunton, J. Prolonged and Tunable Residence Time Using Reversible Covalent Kinase Inhibitors. Nat. Chem. Biol. 2015, 11 (7), 525531,  DOI: 10.1038/nchembio.1817
  33. 33
    Zhou, J.; Stapleton, P.; Xavier-Junior, F. H.; Schatzlein, A.; Haider, S.; Healy, J.; Wells, G. Triazole-Substituted Phenylboronic Acids as Tunable Lead Inhibitors of KPC-2 Antibiotic Resistance. Eur. J. Med. Chem. 2022, 240, 114571,  DOI: 10.1016/j.ejmech.2022.114571
  34. 34
    Ehmke, V.; Quinsaat, J. E. Q.; Rivera-Fuentes, P.; Heindl, C.; Freymond, C.; Rottmann, M.; Brun, R.; Schirmeister, T.; Diederich, F. Tuning and Predicting Biological Affinity: Aryl Nitriles as Cysteine Protease Inhibitors. Org. Biomol. Chem. 2012, 10 (30), 57645768,  DOI: 10.1039/c2ob00034b
  35. 35
    Langrish, C. L.; Bradshaw, J. M.; Francesco, M. R.; Owens, T. D.; Xing, Y.; Shu, J.; LaStant, J.; Bisconte, A.; Outerbridge, C.; White, S. D.; Hill, R. J.; Brameld, K. A.; Goldstein, D. M.; Nunn, P. A. Preclinical Efficacy and Anti-Inflammatory Mechanisms of Action of the Bruton Tyrosine Kinase Inhibitor Rilzabrutinib for Immune-Mediated Disease. J. Immunol. Baltim. Md 1950 2021, 206 (7), 14541468,  DOI: 10.4049/jimmunol.2001130
  36. 36
    Jung, S.; Fuchs, N.; Johe, P.; Wagner, A.; Diehl, E.; Yuliani, T.; Zimmer, C.; Barthels, F.; Zimmermann, R. A.; Klein, P.; Waigel, W.; Meyr, J.; Opatz, T.; Tenzer, S.; Distler, U.; Räder, H.-J.; Kersten, C.; Engels, B.; Hellmich, U. A.; Klein, J.; Schirmeister, T. Fluorovinylsulfones and -Sulfonates as Potent Covalent Reversible Inhibitors of the Trypanosomal Cysteine Protease Rhodesain: Structure-Activity Relationship, Inhibition Mechanism, Metabolism, and In Vivo Studies. J. Med. Chem. 2021, 64 (16), 1232212358,  DOI: 10.1021/acs.jmedchem.1c01002
  37. 37
    Feral, A.; Martin, A. R.; Desfoux, A.; Amblard, M.; Vezenkov, L. L. Covalent-Reversible Peptide-Based Protease Inhibitors. Design, Synthesis, and Clinical Success Stories. Amino Acids 2023, 55, 17751800,  DOI: 10.1007/s00726-023-03286-1
  38. 38
    Fairhurst, R. A.; Knoepfel, T.; Buschmann, N.; Leblanc, C.; Mah, R.; Todorov, M.; Nimsgern, P.; Ripoche, S.; Niklaus, M.; Warin, N.; Luu, V. H.; Madoerin, M.; Wirth, J.; Graus-Porta, D.; Weiss, A.; Kiffe, M.; Wartmann, M.; Kinyamu-Akunda, J.; Sterker, D.; Stamm, C.; Adler, F.; Buhles, A.; Schadt, H.; Couttet, P.; Blank, J.; Galuba, I.; Trappe, J.; Voshol, J.; Ostermann, N.; Zou, C.; Berghausen, J.; Del Rio Espinola, A.; Jahnke, W.; Furet, P. Discovery of Roblitinib (FGF401) as a Reversible-Covalent Inhibitor of the Kinase Activity of Fibroblast Growth Factor Receptor 4. J. Med. Chem. 2020, 63 (21), 1254212573,  DOI: 10.1021/acs.jmedchem.0c01019
  39. 39
    Shindo, N.; Fuchida, H.; Sato, M.; Watari, K.; Shibata, T.; Kuwata, K.; Miura, C.; Okamoto, K.; Hatsuyama, Y.; Tokunaga, K.; Sakamoto, S.; Morimoto, S.; Abe, Y.; Shiroishi, M.; Caaveiro, J. M. M.; Ueda, T.; Tamura, T.; Matsunaga, N.; Nakao, T.; Koyanagi, S.; Ohdo, S.; Yamaguchi, Y.; Hamachi, I.; Ono, M.; Ojida, A. Selective and Reversible Modification of Kinase Cysteines with Chlorofluoroacetamides. Nat. Chem. Biol. 2019, 15 (3), 250258,  DOI: 10.1038/s41589-018-0204-3
  40. 40
    Tallon, A. M.; Xu, Y.; West, G. M.; Am Ende, C. W.; Fox, J. M. Thiomethyltetrazines Are Reversible Covalent Cysteine Warheads Whose Dynamic Behavior Can Be “Switched Off” via Bioorthogonal Chemistry Inside Live Cells. J. Am. Chem. Soc. 2023, 145 (29), 1606916080,  DOI: 10.1021/jacs.3c04444
  41. 41
    Ingiliz, P.; Rockstroh, J. K. HIV-HCV Co-Infection Facing HCV Protease Inhibitor Licensing: Implications for Clinicians. Liver Int. Off. J. Int. Assoc. Study Liver 2012, 32 (8), 11941199,  DOI: 10.1111/j.1478-3231.2012.02796.x
  42. 42
    Teicher, B. A.; Tomaszewski, J. E. Proteasome Inhibitors. Biochem. Pharmacol. 2015, 96 (1), 19,  DOI: 10.1016/j.bcp.2015.04.008
  43. 43
    Metcalf, B.; Chuang, C.; Dufu, K.; Patel, M. P.; Silva-Garcia, A.; Johnson, C.; Lu, Q.; Partridge, J. R.; Patskovska, L.; Patskovsky, Y.; Almo, S. C.; Jacobson, M. P.; Hua, L.; Xu, Q.; Gwaltney, S. L.; Yee, C.; Harris, J.; Morgan, B. P.; James, J.; Xu, D.; Hutchaleelaha, A.; Paulvannan, K.; Oksenberg, D.; Li, Z. Discovery of GBT440, an Orally Bioavailable R-State Stabilizer of Sickle Cell Hemoglobin. ACS Med. Chem. Lett. 2017, 8 (3), 321326,  DOI: 10.1021/acsmedchemlett.6b00491
  44. 44
    Reja, R. M.; Wang, W.; Lyu, Y.; Haeffner, F.; Gao, J. Lysine-Targeting Reversible Covalent Inhibitors with Long Residence Time. J. Am. Chem. Soc. 2022, 144 (3), 11521157,  DOI: 10.1021/jacs.1c12702
  45. 45
    Serafimova, I. M.; Pufall, M. A.; Krishnan, S.; Duda, K.; Cohen, M. S.; Maglathlin, R. L.; McFarland, J. M.; Miller, R. M.; Frödin, M.; Taunton, J. Reversible Targeting of Noncatalytic Cysteines with Chemically Tuned Electrophiles. Nat. Chem. Biol. 2012, 8 (5), 471476,  DOI: 10.1038/nchembio.925
  46. 46
    Forster, M.; Chaikuad, A.; Bauer, S. M.; Holstein, J.; Robers, M. B.; Corona, C. R.; Gehringer, M.; Pfaffenrot, E.; Ghoreschi, K.; Knapp, S.; Laufer, S. A. Selective JAK3 Inhibitors with a Covalent Reversible Binding Mode Targeting a New Induced Fit Binding Pocket. Cell Chem. Biol. 2016, 23 (11), 13351340,  DOI: 10.1016/j.chembiol.2016.10.008
  47. 47
    Dietze, E. C.; Schäfer, A.; Omichinski, J. G.; Nelson, S. D. Inactivation of Glyceraldehyde-3-Phosphate Dehydrogenase by a Reactive Metabolite of Acetaminophen and Mass Spectral Characterization of an Arylated Active Site Peptide. Chem. Res. Toxicol. 1997, 10 (10), 10971103,  DOI: 10.1021/tx970090u
  48. 48
    Rombach, E. M.; Hanzlik, R. P. Identification of a Rat Liver Microsomal Esterase as a Target Protein for Bromobenzene Metabolites. Chem. Res. Toxicol. 1998, 11 (3), 178184,  DOI: 10.1021/tx970076h
  49. 49
    Tailor, A.; Waddington, J. C.; Meng, X.; Park, B. K. Mass Spectrometric and Functional Aspects of Drug-Protein Conjugation. Chem. Res. Toxicol. 2016, 29 (12), 19121935,  DOI: 10.1021/acs.chemrestox.6b00147
  50. 50
    Jeffery, D. A.; Bogyo, M. Chemical Proteomics and Its Application to Drug Discovery. Curr. Opin. Biotechnol. 2003, 14 (1), 8795,  DOI: 10.1016/S0958-1669(02)00010-1
  51. 51
    Weerapana, E.; Wang, C.; Simon, G. M.; Richter, F.; Khare, S.; Dillon, M. B. D.; Bachovchin, D. A.; Mowen, K.; Baker, D.; Cravatt, B. F. Quantitative Reactivity Profiling Predicts Functional Cysteines in Proteomes. Nature 2010, 468 (7325), 790795,  DOI: 10.1038/nature09472
  52. 52
    Backus, K. M.; Correia, B. E.; Lum, K. M.; Forli, S.; Horning, B. D.; González-Páez, G. E.; Chatterjee, S.; Lanning, B. R.; Teijaro, J. R.; Olson, A. J.; Wolan, D. W.; Cravatt, B. F. Proteome-Wide Covalent Ligand Discovery in Native Biological Systems. Nature 2016, 534 (7608), 570574,  DOI: 10.1038/nature18002
  53. 53
    Abbasov, M. E.; Kavanagh, M. E.; Ichu, T.-A.; Lazear, M. R.; Tao, Y.; Crowley, V. M.; Am Ende, C. W.; Hacker, S. M.; Ho, J.; Dix, M. M.; Suciu, R.; Hayward, M. M.; Kiessling, L. L.; Cravatt, B. F. A Proteome-Wide Atlas of Lysine-Reactive Chemistry. Nat. Chem. 2021, 13 (11), 10811092,  DOI: 10.1038/s41557-021-00765-4
  54. 54
    Hayes, J. D.; Flanagan, J. U.; Jowsey, I. R. Glutathione Transferases. Annu. Rev. Pharmacol. Toxicol. 2005, 45, 5188,  DOI: 10.1146/annurev.pharmtox.45.120403.095857
  55. 55
    Jiang, X.; Zhou, Q.; Du, B.; Li, S.; Huang, Y.; Chi, Z.; Lee, W. M.; Yu, M.; Zheng, J. Noninvasive Monitoring of Hepatic Glutathione Depletion through Fluorescence Imaging and Blood Testing. Sci. Adv. 2021, 7 (8), eabd9847  DOI: 10.1126/sciadv.abd9847
  56. 56
    Jeffries, R. E.; Gomez, S. M.; Macdonald, J. M.; Gamcsik, M. P. Direct Detection of Glutathione Biosynthesis, Conjugation, Depletion and Recovery in Intact Hepatoma Cells. Int. J. Mol. Sci. 2022, 23 (9), 4733,  DOI: 10.3390/ijms23094733
  57. 57
    Quanrud, G. M.; Lyu, Z.; Balamurugan, S. V.; Canizal, C.; Wu, H.-T.; Genereux, J. C. Cellular Exposure to Chloroacetanilide Herbicides Induces Distinct Protein Destabilization Profiles. ACS Chem. Biol. 2023, 18 (7), 16611676,  DOI: 10.1021/acschembio.3c00338
  58. 58
    Julio, A. R.; Shikwana, F.; Truong, C.; Burton, N. R.; Dominguez, E.; Turmon, A. C.; Cao, J.; Backus, K. Pervasive Aggregation and Depletion of Host and Viral Proteins in Response to Cysteine-Reactive Electrophilic Compounds. BioRxiv , November 1, 2023.  DOI: 10.1101/2023.10.30.564067 .
  59. 59
    Adair, K.; Meng, X.; Naisbitt, D. J. Drug Hapten-Specific T-Cell Activation: Current Status and Unanswered Questions. Proteomics 2021, 21 (17–18), e2000267  DOI: 10.1002/pmic.202000267
  60. 60
    Weltzien, H. U.; Padovan, E. Molecular Features of Penicillin Allergy. J. Invest. Dermatol. 1998, 110 (3), 203206,  DOI: 10.1046/j.1523-1747.1998.00122.x
  61. 61
    Pirmohamed, M.; Ostrov, D. A.; Park, B. K. New Genetic Findings Lead the Way to a Better Understanding of Fundamental Mechanisms of Drug Hypersensitivity. J. Allergy Clin. Immunol. 2015, 136 (2), 236244,  DOI: 10.1016/j.jaci.2015.06.022
  62. 62
    Agashe, R. P.; Lippman, S. M.; Kurzrock, R. JAK: Not Just Another Kinase. Mol. Cancer Ther. 2022, 21 (12), 17571764,  DOI: 10.1158/1535-7163.MCT-22-0323
  63. 63
    Goedken, E. R.; Argiriadi, M. A.; Banach, D. L.; Fiamengo, B. A.; Foley, S. E.; Frank, K. E.; George, J. S.; Harris, C. M.; Hobson, A. D.; Ihle, D. C.; Marcotte, D.; Merta, P. J.; Michalak, M. E.; Murdock, S. E.; Tomlinson, M. J.; Voss, J. W. Tricyclic Covalent Inhibitors Selectively Target Jak3 through an Active Site Thiol. J. Biol. Chem. 2015, 290 (8), 45734589,  DOI: 10.1074/jbc.M114.595181
  64. 64
    Chen, C.; Lu, D.; Sun, T.; Zhang, T. JAK3 Inhibitors for the Treatment of Inflammatory and Autoimmune Diseases: A Patent Review (2016-Present). Expert Opin. Ther. Pat. 2022, 32 (3), 225242,  DOI: 10.1080/13543776.2022.2023129
  65. 65
    Shindo, N.; Ojida, A. Recent Progress in Covalent Warheads for in Vivo Targeting of Endogenous Proteins. Bioorg. Med. Chem. 2021, 47, 116386,  DOI: 10.1016/j.bmc.2021.116386
  66. 66
    Forster, M.; Chaikuad, A.; Dimitrov, T.; Döring, E.; Holstein, J.; Berger, B.-T.; Gehringer, M.; Ghoreschi, K.; Müller, S.; Knapp, S.; Laufer, S. A. Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4- d]Pyrrolo[2,3- b]Pyridine Scaffold. J. Med. Chem. 2018, 61 (12), 53505366,  DOI: 10.1021/acs.jmedchem.8b00571
  67. 67
    Laux, J.; Forster, M.; Riexinger, L.; Schwamborn, A.; Guezguez, J.; Pokoj, C.; Kudolo, M.; Berger, L. M.; Knapp, S.; Schollmeyer, D.; Guse, J.; Burnet, M.; Laufer, S. A. Pharmacokinetic Optimization of Small Molecule Janus Kinase 3 Inhibitors to Target Immune Cells. ACS Pharmacol. Transl. Sci. 2022, 5 (8), 573602,  DOI: 10.1021/acsptsci.2c00054
  68. 68
    Burger, J. A. Bruton Tyrosine Kinase Inhibitors: Present and Future. Cancer J. 2019, 25 (6), 386393,  DOI: 10.1097/PPO.0000000000000412
  69. 69
    Khan, W. N. Regulation of B Lymphocyte Development and Activation by Bruton’s Tyrosine Kinase. Immunol. Res. 2001, 23 (2–3), 147156,  DOI: 10.1385/IR:23:2-3:147
  70. 70
    Guldenpfennig, C.; Teixeiro, E.; Daniels, M. NF-kB’s Contribution to B Cell Fate Decisions. Front. Immunol. 2023, 14, 1214095,  DOI: 10.3389/fimmu.2023.1214095
  71. 71
    Staudt, L. M. Oncogenic Activation of NF-kappaB. Cold Spring Harb. Perspect. Biol. 2010, 2 (6), a000109,  DOI: 10.1101/cshperspect.a000109
  72. 72
    Small Molecules in Oncology; Martens, U. M., Ed.; Recent Results in Cancer Research Series; Springer: Berlin, Heidelberg, 2014; Vol. 201.  DOI: 10.1007/978-3-642-54490-3 .
  73. 73
    Wu, J.; Zhang, M.; Liu, D. Acalabrutinib (ACP-196): A Selective Second-Generation BTK Inhibitor. J. Hematol. Oncol.J. Hematol Oncol 2016, 9, 21,  DOI: 10.1186/s13045-016-0250-9
  74. 74
    Guo, Y.; Liu, Y.; Hu, N.; Yu, D.; Zhou, C.; Shi, G.; Zhang, B.; Wei, M.; Liu, J.; Luo, L.; Tang, Z.; Song, H.; Guo, Y.; Liu, X.; Su, D.; Zhang, S.; Song, X.; Zhou, X.; Hong, Y.; Chen, S.; Cheng, Z.; Young, S.; Wei, Q.; Wang, H.; Wang, Q.; Lv, L.; Wang, F.; Xu, H.; Sun, H.; Xing, H.; Li, N.; Zhang, W.; Wang, Z.; Liu, G.; Sun, Z.; Zhou, D.; Li, W.; Liu, L.; Wang, L.; Wang, Z. Discovery of Zanubrutinib (BGB-3111), a Novel, Potent, and Selective Covalent Inhibitor of Bruton’s Tyrosine Kinase. J. Med. Chem. 2019, 62 (17), 79237940,  DOI: 10.1021/acs.jmedchem.9b00687
  75. 75
    Liclican, A.; Serafini, L.; Xing, W.; Czerwieniec, G.; Steiner, B.; Wang, T.; Brendza, K. M.; Lutz, J. D.; Keegan, K. S.; Ray, A. S.; Schultz, B. E.; Sakowicz, R.; Feng, J. Y. Biochemical Characterization of Tirabrutinib and Other Irreversible Inhibitors of Bruton’s Tyrosine Kinase Reveals Differences in on - and off - Target Inhibition. Biochim. Biophys. Acta Gen. Subj. 2020, 1864 (4), 129531,  DOI: 10.1016/j.bbagen.2020.129531
  76. 76
    Tam, C. S.; Muñoz, J. L.; Seymour, J. F.; Opat, S. Zanubrutinib: Past, Present, and Future. Blood Cancer J. 2023, 13 (1), 141,  DOI: 10.1038/s41408-023-00902-x
  77. 77
    Estupiñán, H. Y.; Berglöf, A.; Zain, R.; Smith, C. I. E. Comparative Analysis of BTK Inhibitors and Mechanisms Underlying Adverse Effects. Front. Cell Dev. Biol. 2021, 9, 630942,  DOI: 10.3389/fcell.2021.630942
  78. 78
    Mato, A. R.; Nabhan, C.; Thompson, M. C.; Lamanna, N.; Brander, D. M.; Hill, B.; Howlett, C.; Skarbnik, A.; Cheson, B. D.; Zent, C.; Pu, J.; Kiselev, P.; Goy, A.; Claxton, D.; Isaac, K.; Kennard, K. H.; Timlin, C.; Landsburg, D.; Winter, A.; Nasta, S. D.; Bachow, S. H.; Schuster, S. J.; Dorsey, C.; Svoboda, J.; Barr, P.; Ujjani, C. S. Toxicities and Outcomes of 616 Ibrutinib-Treated Patients in the United States: A Real-World Analysis. Haematologica 2018, 103 (5), 874879,  DOI: 10.3324/haematol.2017.182907
  79. 79
    Sharman, J. P.; Black-Shinn, J. L.; Clark, J.; Bitman, B. Understanding Ibrutinib Treatment Discontinuation Patterns for Chronic Lymphocytic Leukemia. Blood 2017, 130 (Supplement 1), 4060,  DOI: 10.1182/blood.V130.Suppl_1.4060.4060
  80. 80
    Winqvist, M.; Andersson, P.-O.; Asklid, A.; Karlsson, K.; Karlsson, C.; Lauri, B.; Lundin, J.; Mattsson, M.; Norin, S.; Sandstedt, A.; Rosenquist, R.; Späth, F.; Hansson, L.; Österborg, A. for the Swedish CLL Group. Long-Term Real-World Results of Ibrutinib Therapy in Patients with Relapsed or Refractory Chronic Lymphocytic Leukemia: 30-Month Follow up of the Swedish Compassionate Use Cohort. Haematologica 2019, 104 (5), e208e210,  DOI: 10.3324/haematol.2018.198820
  81. 81
    ClinicalTrials.gov. Rilzabrutinib. https://clinicaltrials.gov/search?intr=Rilzabrutinib (accessed 2023–11–28).
  82. 82
    Oda, K. New Families of Carboxyl Peptidases: Serine-Carboxyl Peptidases and Glutamic Peptidases. J. Biochem. (Tokyo) 2012, 151 (1), 1325,  DOI: 10.1093/jb/mvr129
  83. 83
    Ćwilichowska, N.; Świderska, K. W.; Dobrzyń, A.; Drąg, M.; Poręba, M. Diagnostic and Therapeutic Potential of Protease Inhibition. Mol. Aspects Med. 2022, 88, 101144,  DOI: 10.1016/j.mam.2022.101144
  84. 84
    Manasanch, E. E.; Orlowski, R. Z. Proteasome Inhibitors in Cancer Therapy. Nat. Rev. Clin. Oncol. 2017, 14 (7), 417433,  DOI: 10.1038/nrclinonc.2016.206
  85. 85
    Flint, M.; Mullen, S.; Deatly, A. M.; Chen, W.; Miller, L. Z.; Ralston, R.; Broom, C.; Emini, E. A.; Howe, A. Y. M. Selection and Characterization of Hepatitis C Virus Replicons Dually Resistant to the Polymerase and Protease Inhibitors HCV-796 and Boceprevir (SCH 503034). Antimicrob. Agents Chemother. 2009, 53 (2), 401411,  DOI: 10.1128/AAC.01081-08
  86. 86
    Augeri, D. J.; Robl, J. A.; Betebenner, D. A.; Magnin, D. R.; Khanna, A.; Robertson, J. G.; Wang, A.; Simpkins, L. M.; Taunk, P.; Huang, Q.; Han, S.-P.; Abboa-Offei, B.; Cap, M.; Xin, L.; Tao, L.; Tozzo, E.; Welzel, G. E.; Egan, D. M.; Marcinkeviciene, J.; Chang, S. Y.; Biller, S. A.; Kirby, M. S.; Parker, R. A.; Hamann, L. G. Discovery and Preclinical Profile of Saxagliptin (BMS-477118): A Highly Potent, Long-Acting, Orally Active Dipeptidyl Peptidase IV Inhibitor for the Treatment of Type 2 Diabetes. J. Med. Chem. 2005, 48 (15), 50255037,  DOI: 10.1021/jm050261p
  87. 87
    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 (5), 18131822,  DOI: 10.1128/AAC.50.5.1813-1822.2006
  88. 88
    Focosi, D.; McConnell, S.; Shoham, S.; Casadevall, A.; Maggi, F.; Antonelli, G. Nirmatrelvir and COVID-19: Development, Pharmacokinetics, Clinical Efficacy, Resistance, Relapse, and Pharmacoeconomics. Int. J. Antimicrob. Agents 2023, 61 (2), 106708,  DOI: 10.1016/j.ijantimicag.2022.106708
  89. 89
    Wen, W.; Qi, Z.; Wang, J. The Function and Mechanism of Enterovirus 71 (EV71) 3C Protease. Curr. Microbiol. 2020, 77 (9), 19681975,  DOI: 10.1007/s00284-020-02082-4
  90. 90
    Zeng, D.; Ma, Y.; Zhang, R.; Nie, Q.; Cui, Z.; Wang, Y.; Shang, L.; Yin, Z. Synthesis and Structure-Activity Relationship of α-Keto Amides as Enterovirus 71 3C Protease Inhibitors. Bioorg. Med. Chem. Lett. 2016, 26 (7), 17621766,  DOI: 10.1016/j.bmcl.2016.02.039
  91. 91
    Zhai, Y.; Ma, Y.; Ma, F.; Nie, Q.; Ren, X.; Wang, Y.; Shang, L.; Yin, Z. Structure-Activity Relationship Study of Peptidomimetic Aldehydes as Enterovirus 71 3C Protease Inhibitors. Eur. J. Med. Chem. 2016, 124, 559573,  DOI: 10.1016/j.ejmech.2016.08.064
  92. 92
    Luo, Y. L. Mechanism-Based and Computational-Driven Covalent Drug Design. J. Chem. Inf. Model. 2021, 61 (11), 53075311,  DOI: 10.1021/acs.jcim.1c01278
  93. 93
    Mihalovits, L. M.; Ferenczy, G. G.; Keserű, G. M. Affinity and Selectivity Assessment of Covalent Inhibitors by Free Energy Calculations. J. Chem. Inf. Model. 2020, 60 (12), 65796594,  DOI: 10.1021/acs.jcim.0c00834
  94. 94
    Mihalovits, L. M.; Ferenczy, G. G.; Keserű, G. M. The Role of Quantum Chemistry in Covalent Inhibitor Design. Int. J. Quantum Chem. 2022, 122 (8), e26768  DOI: 10.1002/qua.26768
  95. 95
    Lonsdale, R.; Burgess, J.; Colclough, N.; Davies, N. L.; Lenz, E. M.; Orton, A. L.; Ward, R. A. Expanding the Armory: Predicting and Tuning Covalent Warhead Reactivity. J. Chem. Inf. Model. 2017, 57 (12), 31243137,  DOI: 10.1021/acs.jcim.7b00553
  96. 96
    Oballa, R. M.; Truchon, J.-F.; Bayly, C. I.; Chauret, N.; Day, S.; Crane, S.; Berthelette, C. A Generally Applicable Method for Assessing the Electrophilicity and Reactivity of Diverse Nitrile-Containing Compounds. Bioorg. Med. Chem. Lett. 2007, 17 (4), 9981002,  DOI: 10.1016/j.bmcl.2006.11.044
  97. 97
    Copeland, R. A.; Pompliano, D. L.; Meek, T. D. Drug-Target Residence Time and Its Implications for Lead Optimization. Nat. Rev. Drug Discovery 2006, 5 (9), 730739,  DOI: 10.1038/nrd2082
  98. 98
    Bernetti, M.; Masetti, M.; Rocchia, W.; Cavalli, A. Kinetics of Drug Binding and Residence Time. Annu. Rev. Phys. Chem. 2019, 70, 143171,  DOI: 10.1146/annurev-physchem-042018-052340
  99. 99
    Ren, T.; Zhu, X.; Jusko, N. M.; Krzyzanski, W.; Jusko, W. J. Pharmacodynamic Model of Slow Reversible Binding and Its Applications in Pharmacokinetic/Pharmacodynamic Modeling: Review and Tutorial. J. Pharmacokinet. Pharmacodyn. 2022, 49 (5), 493510,  DOI: 10.1007/s10928-022-09822-y
  100. 100
    Frühauf, A.; Wolff, B.; Schweipert, M.; Meyer-Almes, F.-J. Synthesis and Characterization of Reversible Covalent HDAC4 Inhibitors. Methods Mol. Biol. Clifton NJ. 2023, 2589, 207221,  DOI: 10.1007/978-1-0716-2788-4_14
  101. 101
    Forman, H. J.; Zhang, H.; Rinna, A. Glutathione: Overview of Its Protective Roles, Measurement, and Biosynthesis. Mol. Aspects Med. 2009, 30 (1–2), 112,  DOI: 10.1016/j.mam.2008.08.006
  102. 102
    Wu, G.; Fang, Y.-Z.; Yang, S.; Lupton, J. R.; Turner, N. D. Glutathione Metabolism and Its Implications for Health. J. Nutr. 2004, 134 (3), 489492,  DOI: 10.1093/jn/134.3.489
  103. 103
    Bansal, A.; Simon, M. C. Glutathione Metabolism in Cancer Progression and Treatment Resistance. J. Cell Biol. 2018, 217 (7), 22912298,  DOI: 10.1083/jcb.201804161
  104. 104
    Bajic, V. P.; Van Neste, C.; Obradovic, M.; Zafirovic, S.; Radak, D.; Bajic, V. B.; Essack, M.; Isenovic, E. R. Glutathione “Redox Homeostasis” and Its Relation to Cardiovascular Disease. Oxid. Med. Cell. Longev. 2019, 2019, 5028181,  DOI: 10.1155/2019/5028181
  105. 105
    Fu, L.; Li, Z.; Liu, K.; Tian, C.; He, J.; He, J.; He, F.; Xu, P.; Yang, J. A Quantitative Thiol Reactivity Profiling Platform to Analyze Redox and Electrophile Reactive Cysteine Proteomes. Nat. Protoc. 2020, 15 (9), 28912919,  DOI: 10.1038/s41596-020-0352-2
  106. 106
    Kuljanin, M.; Mitchell, D. C.; Schweppe, D. K.; Gikandi, A. S.; Nusinow, D. P.; Bulloch, N. J.; Vinogradova, E. V.; Wilson, D. L.; Kool, E. T.; Mancias, J. D.; Cravatt, B. F.; Gygi, S. P. Reimagining High-Throughput Profiling of Reactive Cysteines for Cell-Based Screening of Large Electrophile Libraries. Nat. Biotechnol. 2021, 39 (5), 630641,  DOI: 10.1038/s41587-020-00778-3
  107. 107
    Yamamoto, M.; Kensler, T. W.; Motohashi, H. The KEAP1-NRF2 System: A Thiol-Based Sensor-Effector Apparatus for Maintaining Redox Homeostasis. Physiol. Rev. 2018, 98 (3), 11691203,  DOI: 10.1152/physrev.00023.2017
  108. 108
    Unoki, T.; Akiyama, M.; Kumagai, Y. Nrf2 Activation and Its Coordination with the Protective Defense Systems in Response to Electrophilic Stress. Int. J. Mol. Sci. 2020, 21 (2), 545,  DOI: 10.3390/ijms21020545
  109. 109
    Backus, K. M.; Cao, J.; Maddox, S. M. Opportunities and Challenges for the Development of Covalent Chemical Immunomodulators. Bioorg. Med. Chem. 2019, 27 (15), 34213439,  DOI: 10.1016/j.bmc.2019.05.050
  110. 110
    Lincoln, R.; Zhang, W.; Lovell, T. C.; Jodko-Piórecka, K.; Devlaminck, P. A.; Sakaya, A.; Van Kessel, A.; Cosa, G. Chemically Tuned, Reversible Fluorogenic Electrophile for Live Cell Nanoscopy. ACS Sens. 2022, 7 (1), 166174,  DOI: 10.1021/acssensors.1c01940
  111. 111
    Zheng, S.; Liu, G. Polymeric Emissive Materials Based on Dynamic Covalent Bonds. Mol. Basel Switz. 2022, 27 (19), 6635,  DOI: 10.3390/molecules27196635

Cited By

Click to copy section linkSection link copied!

This article has not yet been cited by other publications.

Open PDF

ACS Chemical Biology

Cite this: ACS Chem. Biol. 2024, 19, 4, 824–838
Click to copy citationCitation copied!
https://doi.org/10.1021/acschembio.3c00805
Published April 3, 2024

Copyright © 2024 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0 .

Article Views

5985

Altmetric

-

Citations

-
Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. Structures of Penicillin G, 3-nitropropionate, itaconate, and wortmannin are examples of natural product covalent inhibitors with their covalent adducts.

    Figure 2

    Figure 2. Schematic of NCI, ICI, and RCI inhibition with descriptions of the equilibrium or inactivation constants (Mons et al.). Adapted from ref (15). Available under a CC-BY 4.0 license. Copyright 2024 Elma Mons, Sander Roet, Robbert Q. Kim, Monique P. C. Mulder.

    Figure 3

    Figure 3. Inhibitor is in equilibrium with many enzymes, including the target enzyme. Since the inhibitor–target complex is greatly favored, this will shift the equilibria of the off-target interactions away from the off-target complexes with the inhibitor as described by Le Chatalier’s principle. Over time, the inhibitor–target complex will be the predominant site of the RCI.

    Figure 4

    Figure 4. The observed equilibrium of a covalent inhibitor is the product of the binding equilibrium constant and the reaction equilibrium constant.

    Figure 5

    Figure 5. Nucleophilic residues reacting with electrophilic warheads in equilibrium with their covalent-adduct products, with the electrophilic warheads highlighted in red.

    Figure 6

    Figure 6. Examples of reversible covalent inhibitors with their electrophilic warheads are highlighted in red.

    Figure 7

    Figure 7. Examples of JAK3 inhibitors with covalent warheads are highlighted in red.

    Figure 8

    Figure 8. Clinically approved irreversible covalent inhibitors for BTK: ibrutinib, Zanubrutinib, acalabrutinib, and tirabrutinib, and a reversible covalent inhibitor for BTK in clinical trials: rilzabrutinib. The covalent warhead is highlighted in red.

    Figure 9

    Figure 9. Compound optimization for the EV71 cPro enzyme. The covalent warhead is highlighted in red.

    Figure 10

    Figure 10. Summarized activities for each parameter that are important in the design of a reversible covalent inhibitor.

  • References


    This article references 111 other publications.

    1. 1
      Fleming, A. On the Antibacterial Action of Cultures of a Penicillium, with Special Reference to Their Use in the Isolation of B. Influenzae. 1929. Bull. World Health Organ. 2001, 79 (8), 780790
    2. 2
      Duncan, D.; Auclair, K. Itaconate: An Antimicrobial Metabolite of Macrophages. Can. J. Chem. 2022, 100 (2), 104113,  DOI: 10.1139/cjc-2021-0117
    3. 3
      Ray, S.; Kreitler, D. F.; Gulick, A. M.; Murkin, A. S. The Nitro Group as a Masked Electrophile in Covalent Enzyme Inhibition. ACS Chem. Biol. 2018, 13 (6), 14701473,  DOI: 10.1021/acschembio.8b00225
    4. 4
      Yuan, H.; Barnes, K. R.; Weissleder, R.; Cantley, L.; Josephson, L. Covalent Reactions of Wortmannin under Physiological Conditions. Chem. Biol. 2007, 14 (3), 321328,  DOI: 10.1016/j.chembiol.2007.02.007
    5. 5
      De Vita, E. 10 Years into the Resurgence of Covalent Drugs. Future Med. Chem. 2021, 13 (2), 193210,  DOI: 10.4155/fmc-2020-0236
    6. 6
      Baillie, T. A. Drug-Protein Adducts: Past, Present, and Future. Med. Chem. Res. 2020, 29 (7), 10931104,  DOI: 10.1007/s00044-020-02567-8
    7. 7
      Potashman, M. H.; Duggan, M. E. Covalent Modifiers: An Orthogonal Approach to Drug Design. J. Med. Chem. 2009, 52 (5), 12311246,  DOI: 10.1021/jm8008597
    8. 8
      Boike, L.; Henning, N. J.; Nomura, D. K. Advances in Covalent Drug Discovery. Nat. Rev. Drug Discovery 2022, 21 (12), 881898,  DOI: 10.1038/s41573-022-00542-z
    9. 9
      Kenakin, T. The Mass Action Equation in Pharmacology. Br. J. Clin. Pharmacol. 2016, 81 (1), 4151,  DOI: 10.1111/bcp.12810
    10. 10
      Pottel, J.; Levit, A.; Korczynska, M.; Fischer, M.; Shoichet, B. K. The Recognition of Unrelated Ligands by Identical Proteins. ACS Chem. Biol. 2018, 13 (9), 25222533,  DOI: 10.1021/acschembio.8b00443
    11. 11
      Knockenhauer, K. E.; Copeland, R. A. The Importance of Binding Kinetics and Drug-Target Residence Time in Pharmacology. Br. J. Pharmacol. 2023, 114,  DOI: 10.1111/bph.16104
    12. 12
      Zhang, G.; Zhang, J.; Gao, Y.; Li, Y.; Li, Y. Strategies for Targeting Undruggable Targets. Expert Opin. Drug Discovery 2022, 17 (1), 5569,  DOI: 10.1080/17460441.2021.1969359
    13. 13
      Akçay, G.; Belmonte, M. A.; Aquila, B.; Chuaqui, C.; Hird, A. W.; Lamb, M. L.; Rawlins, P. B.; Su, N.; Tentarelli, S.; Grimster, N. P.; Su, Q. Inhibition of Mcl-1 through Covalent Modification of a Noncatalytic Lysine Side Chain. Nat. Chem. Biol. 2016, 12 (11), 931936,  DOI: 10.1038/nchembio.2174
    14. 14
      Huang, L.; Guo, Z.; Wang, F.; Fu, L. KRAS Mutation: From Undruggable to Druggable in Cancer. Signal Transduct. Target. Ther. 2021, 6 (1), 386,  DOI: 10.1038/s41392-021-00780-4
    15. 15
      Mons, E.; Roet, S.; Kim, R. Q.; Mulder, M. P. C. A Comprehensive Guide for Assessing Covalent Inhibition in Enzymatic Assays Illustrated with Kinetic Simulations. Curr. Protoc. 2022, 2 (6), e419  DOI: 10.1002/cpz1.419
    16. 16
      Chatterjee, P.; Botello-Smith, W. M.; Zhang, H.; Qian, L.; Alsamarah, A.; Kent, D.; Lacroix, J. J.; Baudry, M.; Luo, Y. Can Relative Binding Free Energy Predict Selectivity of Reversible Covalent Inhibitors?. J. Am. Chem. Soc. 2017, 139 (49), 1794517952,  DOI: 10.1021/jacs.7b08938
    17. 17
      De Cesco, S.; Kurian, J.; Dufresne, C.; Mittermaier, A. K.; Moitessier, N. Covalent Inhibitors Design and Discovery. Eur. J. Med. Chem. 2017, 138, 96114,  DOI: 10.1016/j.ejmech.2017.06.019
    18. 18
      Plescia, J.; De Cesco, S.; Patrascu, M. B.; Kurian, J.; Di Trani, J.; Dufresne, C.; Wahba, A. S.; Janmamode, N.; Mittermaier, A. K.; Moitessier, N. Integrated Synthetic, Biophysical, and Computational Investigations of Covalent Inhibitors of Prolyl Oligopeptidase and Fibroblast Activation Protein α. J. Med. Chem. 2019, 62 (17), 78747884,  DOI: 10.1021/acs.jmedchem.9b00642
    19. 19
      Masuda, Y.; Yoshida, T.; Yamaotsu, N.; Hirono, S. Linear Discriminant Analysis for the in Silico Discovery of Mechanism-Based Reversible Covalent Inhibitors of a Serine Protease: Application of Hydration Thermodynamics Analysis and Semi-Empirical Molecular Orbital Calculation. Chem. Pharm. Bull. (Tokyo) 2018, 66 (4), 399409,  DOI: 10.1248/cpb.c17-00854
    20. 20
      Awoonor-Williams, E.; Walsh, A. G.; Rowley, C. N. Modeling Covalent-Modifier Drugs. Biochim. Biophys. Acta Proteins Proteomics 2017, 1865 (11), 16641675,  DOI: 10.1016/j.bbapap.2017.05.009
    21. 21
      Scarpino, A.; Ferenczy, G. G.; Keserű, G. M. Binding Mode Prediction and Virtual Screening Applications by Covalent Docking. Methods Mol. Biol. Clifton NJ. 2021, 2266, 7388,  DOI: 10.1007/978-1-0716-1209-5_4
    22. 22
      Faridoon; Ng, R.; Zhang, G.; Li, J. J. An Update on the Discovery and Development of Reversible Covalent Inhibitors. Med. Chem. Res. Int. J. Rapid Commun. Des. Mech. Action Biol. Act. Agents 2023, 32 (6), 10391062,  DOI: 10.1007/s00044-023-03065-3
    23. 23
      Martin, J. S.; MacKenzie, C. J.; Fletcher, D.; Gilbert, I. H. Characterising Covalent Warhead Reactivity. Bioorg. Med. Chem. 2019, 27 (10), 20662074,  DOI: 10.1016/j.bmc.2019.04.002
    24. 24
      Péczka, N.; Orgován, Z.; Ábrányi-Balogh, P.; Keserű, G. M. Electrophilic Warheads in Covalent Drug Discovery: An Overview. Expert Opin. Drug Discovery 2022, 17 (4), 413422,  DOI: 10.1080/17460441.2022.2034783
    25. 25
      Jackson, P. A.; Widen, J. C.; Harki, D. A.; Brummond, K. M. Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition Reactions. J. Med. Chem. 2017, 60 (3), 839885,  DOI: 10.1021/acs.jmedchem.6b00788
    26. 26
      Watt, S. K. I.; Charlebois, J. G.; Rowley, C. N.; Keillor, J. W. A Mechanistic Study of Thiol Addition to N-Phenylacrylamide. Org. Biomol. Chem. 2022, 20 (45), 88988906,  DOI: 10.1039/D2OB01369J
    27. 27
      Watt, S. K. I.; Charlebois, J. G.; Rowley, C. N.; Keillor, J. W. A Mechanistic Study of Thiol Addition to N-Acryloylpiperidine. Org. Biomol. Chem. 2023, 21 (10), 22042212,  DOI: 10.1039/D2OB02223K
    28. 28
      Lohman, D. C.; Edwards, D. R.; Wolfenden, R. Catalysis by Desolvation: The Catalytic Prowess of SAM-Dependent Halide-Alkylating Enzymes. J. Am. Chem. Soc. 2013, 135 (39), 1447314475,  DOI: 10.1021/ja406381b
    29. 29
      Parvez, S.; Long, M. J. C.; Poganik, J. R.; Aye, Y. Redox Signaling by Reactive Electrophiles and Oxidants. Chem. Rev. 2018, 118 (18), 87988888,  DOI: 10.1021/acs.chemrev.7b00698
    30. 30
      Krishnan, S.; Miller, R. M.; Tian, B.; Mullins, R. D.; Jacobson, M. P.; Taunton, J. Design of Reversible, Cysteine-Targeted Michael Acceptors Guided by Kinetic and Computational Analysis. J. Am. Chem. Soc. 2014, 136 (36), 1262412630,  DOI: 10.1021/ja505194w
    31. 31
      Ma, Y.; Li, L.; He, S.; Shang, C.; Sun, Y.; Liu, N.; Meek, T. D.; Wang, Y.; Shang, L. Application of Dually Activated Michael Acceptor to the Rational Design of Reversible Covalent Inhibitor for Enterovirus 71 3C Protease. J. Med. Chem. 2019, 62 (13), 61466162,  DOI: 10.1021/acs.jmedchem.9b00387
    32. 32
      Bradshaw, J. M.; McFarland, J. M.; Paavilainen, V. O.; Bisconte, A.; Tam, D.; Phan, V. T.; Romanov, S.; Finkle, D.; Shu, J.; Patel, V.; Ton, T.; Li, X.; Loughhead, D. G.; Nunn, P. A.; Karr, D. E.; Gerritsen, M. E.; Funk, J. O.; Owens, T. D.; Verner, E.; Brameld, K. A.; Hill, R. J.; Goldstein, D. M.; Taunton, J. Prolonged and Tunable Residence Time Using Reversible Covalent Kinase Inhibitors. Nat. Chem. Biol. 2015, 11 (7), 525531,  DOI: 10.1038/nchembio.1817
    33. 33
      Zhou, J.; Stapleton, P.; Xavier-Junior, F. H.; Schatzlein, A.; Haider, S.; Healy, J.; Wells, G. Triazole-Substituted Phenylboronic Acids as Tunable Lead Inhibitors of KPC-2 Antibiotic Resistance. Eur. J. Med. Chem. 2022, 240, 114571,  DOI: 10.1016/j.ejmech.2022.114571
    34. 34
      Ehmke, V.; Quinsaat, J. E. Q.; Rivera-Fuentes, P.; Heindl, C.; Freymond, C.; Rottmann, M.; Brun, R.; Schirmeister, T.; Diederich, F. Tuning and Predicting Biological Affinity: Aryl Nitriles as Cysteine Protease Inhibitors. Org. Biomol. Chem. 2012, 10 (30), 57645768,  DOI: 10.1039/c2ob00034b
    35. 35
      Langrish, C. L.; Bradshaw, J. M.; Francesco, M. R.; Owens, T. D.; Xing, Y.; Shu, J.; LaStant, J.; Bisconte, A.; Outerbridge, C.; White, S. D.; Hill, R. J.; Brameld, K. A.; Goldstein, D. M.; Nunn, P. A. Preclinical Efficacy and Anti-Inflammatory Mechanisms of Action of the Bruton Tyrosine Kinase Inhibitor Rilzabrutinib for Immune-Mediated Disease. J. Immunol. Baltim. Md 1950 2021, 206 (7), 14541468,  DOI: 10.4049/jimmunol.2001130
    36. 36
      Jung, S.; Fuchs, N.; Johe, P.; Wagner, A.; Diehl, E.; Yuliani, T.; Zimmer, C.; Barthels, F.; Zimmermann, R. A.; Klein, P.; Waigel, W.; Meyr, J.; Opatz, T.; Tenzer, S.; Distler, U.; Räder, H.-J.; Kersten, C.; Engels, B.; Hellmich, U. A.; Klein, J.; Schirmeister, T. Fluorovinylsulfones and -Sulfonates as Potent Covalent Reversible Inhibitors of the Trypanosomal Cysteine Protease Rhodesain: Structure-Activity Relationship, Inhibition Mechanism, Metabolism, and In Vivo Studies. J. Med. Chem. 2021, 64 (16), 1232212358,  DOI: 10.1021/acs.jmedchem.1c01002
    37. 37
      Feral, A.; Martin, A. R.; Desfoux, A.; Amblard, M.; Vezenkov, L. L. Covalent-Reversible Peptide-Based Protease Inhibitors. Design, Synthesis, and Clinical Success Stories. Amino Acids 2023, 55, 17751800,  DOI: 10.1007/s00726-023-03286-1
    38. 38
      Fairhurst, R. A.; Knoepfel, T.; Buschmann, N.; Leblanc, C.; Mah, R.; Todorov, M.; Nimsgern, P.; Ripoche, S.; Niklaus, M.; Warin, N.; Luu, V. H.; Madoerin, M.; Wirth, J.; Graus-Porta, D.; Weiss, A.; Kiffe, M.; Wartmann, M.; Kinyamu-Akunda, J.; Sterker, D.; Stamm, C.; Adler, F.; Buhles, A.; Schadt, H.; Couttet, P.; Blank, J.; Galuba, I.; Trappe, J.; Voshol, J.; Ostermann, N.; Zou, C.; Berghausen, J.; Del Rio Espinola, A.; Jahnke, W.; Furet, P. Discovery of Roblitinib (FGF401) as a Reversible-Covalent Inhibitor of the Kinase Activity of Fibroblast Growth Factor Receptor 4. J. Med. Chem. 2020, 63 (21), 1254212573,  DOI: 10.1021/acs.jmedchem.0c01019
    39. 39
      Shindo, N.; Fuchida, H.; Sato, M.; Watari, K.; Shibata, T.; Kuwata, K.; Miura, C.; Okamoto, K.; Hatsuyama, Y.; Tokunaga, K.; Sakamoto, S.; Morimoto, S.; Abe, Y.; Shiroishi, M.; Caaveiro, J. M. M.; Ueda, T.; Tamura, T.; Matsunaga, N.; Nakao, T.; Koyanagi, S.; Ohdo, S.; Yamaguchi, Y.; Hamachi, I.; Ono, M.; Ojida, A. Selective and Reversible Modification of Kinase Cysteines with Chlorofluoroacetamides. Nat. Chem. Biol. 2019, 15 (3), 250258,  DOI: 10.1038/s41589-018-0204-3
    40. 40
      Tallon, A. M.; Xu, Y.; West, G. M.; Am Ende, C. W.; Fox, J. M. Thiomethyltetrazines Are Reversible Covalent Cysteine Warheads Whose Dynamic Behavior Can Be “Switched Off” via Bioorthogonal Chemistry Inside Live Cells. J. Am. Chem. Soc. 2023, 145 (29), 1606916080,  DOI: 10.1021/jacs.3c04444
    41. 41
      Ingiliz, P.; Rockstroh, J. K. HIV-HCV Co-Infection Facing HCV Protease Inhibitor Licensing: Implications for Clinicians. Liver Int. Off. J. Int. Assoc. Study Liver 2012, 32 (8), 11941199,  DOI: 10.1111/j.1478-3231.2012.02796.x
    42. 42
      Teicher, B. A.; Tomaszewski, J. E. Proteasome Inhibitors. Biochem. Pharmacol. 2015, 96 (1), 19,  DOI: 10.1016/j.bcp.2015.04.008
    43. 43
      Metcalf, B.; Chuang, C.; Dufu, K.; Patel, M. P.; Silva-Garcia, A.; Johnson, C.; Lu, Q.; Partridge, J. R.; Patskovska, L.; Patskovsky, Y.; Almo, S. C.; Jacobson, M. P.; Hua, L.; Xu, Q.; Gwaltney, S. L.; Yee, C.; Harris, J.; Morgan, B. P.; James, J.; Xu, D.; Hutchaleelaha, A.; Paulvannan, K.; Oksenberg, D.; Li, Z. Discovery of GBT440, an Orally Bioavailable R-State Stabilizer of Sickle Cell Hemoglobin. ACS Med. Chem. Lett. 2017, 8 (3), 321326,  DOI: 10.1021/acsmedchemlett.6b00491
    44. 44
      Reja, R. M.; Wang, W.; Lyu, Y.; Haeffner, F.; Gao, J. Lysine-Targeting Reversible Covalent Inhibitors with Long Residence Time. J. Am. Chem. Soc. 2022, 144 (3), 11521157,  DOI: 10.1021/jacs.1c12702
    45. 45
      Serafimova, I. M.; Pufall, M. A.; Krishnan, S.; Duda, K.; Cohen, M. S.; Maglathlin, R. L.; McFarland, J. M.; Miller, R. M.; Frödin, M.; Taunton, J. Reversible Targeting of Noncatalytic Cysteines with Chemically Tuned Electrophiles. Nat. Chem. Biol. 2012, 8 (5), 471476,  DOI: 10.1038/nchembio.925
    46. 46
      Forster, M.; Chaikuad, A.; Bauer, S. M.; Holstein, J.; Robers, M. B.; Corona, C. R.; Gehringer, M.; Pfaffenrot, E.; Ghoreschi, K.; Knapp, S.; Laufer, S. A. Selective JAK3 Inhibitors with a Covalent Reversible Binding Mode Targeting a New Induced Fit Binding Pocket. Cell Chem. Biol. 2016, 23 (11), 13351340,  DOI: 10.1016/j.chembiol.2016.10.008
    47. 47
      Dietze, E. C.; Schäfer, A.; Omichinski, J. G.; Nelson, S. D. Inactivation of Glyceraldehyde-3-Phosphate Dehydrogenase by a Reactive Metabolite of Acetaminophen and Mass Spectral Characterization of an Arylated Active Site Peptide. Chem. Res. Toxicol. 1997, 10 (10), 10971103,  DOI: 10.1021/tx970090u
    48. 48
      Rombach, E. M.; Hanzlik, R. P. Identification of a Rat Liver Microsomal Esterase as a Target Protein for Bromobenzene Metabolites. Chem. Res. Toxicol. 1998, 11 (3), 178184,  DOI: 10.1021/tx970076h
    49. 49
      Tailor, A.; Waddington, J. C.; Meng, X.; Park, B. K. Mass Spectrometric and Functional Aspects of Drug-Protein Conjugation. Chem. Res. Toxicol. 2016, 29 (12), 19121935,  DOI: 10.1021/acs.chemrestox.6b00147
    50. 50
      Jeffery, D. A.; Bogyo, M. Chemical Proteomics and Its Application to Drug Discovery. Curr. Opin. Biotechnol. 2003, 14 (1), 8795,  DOI: 10.1016/S0958-1669(02)00010-1
    51. 51
      Weerapana, E.; Wang, C.; Simon, G. M.; Richter, F.; Khare, S.; Dillon, M. B. D.; Bachovchin, D. A.; Mowen, K.; Baker, D.; Cravatt, B. F. Quantitative Reactivity Profiling Predicts Functional Cysteines in Proteomes. Nature 2010, 468 (7325), 790795,  DOI: 10.1038/nature09472
    52. 52
      Backus, K. M.; Correia, B. E.; Lum, K. M.; Forli, S.; Horning, B. D.; González-Páez, G. E.; Chatterjee, S.; Lanning, B. R.; Teijaro, J. R.; Olson, A. J.; Wolan, D. W.; Cravatt, B. F. Proteome-Wide Covalent Ligand Discovery in Native Biological Systems. Nature 2016, 534 (7608), 570574,  DOI: 10.1038/nature18002
    53. 53
      Abbasov, M. E.; Kavanagh, M. E.; Ichu, T.-A.; Lazear, M. R.; Tao, Y.; Crowley, V. M.; Am Ende, C. W.; Hacker, S. M.; Ho, J.; Dix, M. M.; Suciu, R.; Hayward, M. M.; Kiessling, L. L.; Cravatt, B. F. A Proteome-Wide Atlas of Lysine-Reactive Chemistry. Nat. Chem. 2021, 13 (11), 10811092,  DOI: 10.1038/s41557-021-00765-4
    54. 54
      Hayes, J. D.; Flanagan, J. U.; Jowsey, I. R. Glutathione Transferases. Annu. Rev. Pharmacol. Toxicol. 2005, 45, 5188,  DOI: 10.1146/annurev.pharmtox.45.120403.095857
    55. 55
      Jiang, X.; Zhou, Q.; Du, B.; Li, S.; Huang, Y.; Chi, Z.; Lee, W. M.; Yu, M.; Zheng, J. Noninvasive Monitoring of Hepatic Glutathione Depletion through Fluorescence Imaging and Blood Testing. Sci. Adv. 2021, 7 (8), eabd9847  DOI: 10.1126/sciadv.abd9847
    56. 56
      Jeffries, R. E.; Gomez, S. M.; Macdonald, J. M.; Gamcsik, M. P. Direct Detection of Glutathione Biosynthesis, Conjugation, Depletion and Recovery in Intact Hepatoma Cells. Int. J. Mol. Sci. 2022, 23 (9), 4733,  DOI: 10.3390/ijms23094733
    57. 57
      Quanrud, G. M.; Lyu, Z.; Balamurugan, S. V.; Canizal, C.; Wu, H.-T.; Genereux, J. C. Cellular Exposure to Chloroacetanilide Herbicides Induces Distinct Protein Destabilization Profiles. ACS Chem. Biol. 2023, 18 (7), 16611676,  DOI: 10.1021/acschembio.3c00338
    58. 58
      Julio, A. R.; Shikwana, F.; Truong, C.; Burton, N. R.; Dominguez, E.; Turmon, A. C.; Cao, J.; Backus, K. Pervasive Aggregation and Depletion of Host and Viral Proteins in Response to Cysteine-Reactive Electrophilic Compounds. BioRxiv , November 1, 2023.  DOI: 10.1101/2023.10.30.564067 .
    59. 59
      Adair, K.; Meng, X.; Naisbitt, D. J. Drug Hapten-Specific T-Cell Activation: Current Status and Unanswered Questions. Proteomics 2021, 21 (17–18), e2000267  DOI: 10.1002/pmic.202000267
    60. 60
      Weltzien, H. U.; Padovan, E. Molecular Features of Penicillin Allergy. J. Invest. Dermatol. 1998, 110 (3), 203206,  DOI: 10.1046/j.1523-1747.1998.00122.x
    61. 61
      Pirmohamed, M.; Ostrov, D. A.; Park, B. K. New Genetic Findings Lead the Way to a Better Understanding of Fundamental Mechanisms of Drug Hypersensitivity. J. Allergy Clin. Immunol. 2015, 136 (2), 236244,  DOI: 10.1016/j.jaci.2015.06.022
    62. 62
      Agashe, R. P.; Lippman, S. M.; Kurzrock, R. JAK: Not Just Another Kinase. Mol. Cancer Ther. 2022, 21 (12), 17571764,  DOI: 10.1158/1535-7163.MCT-22-0323
    63. 63
      Goedken, E. R.; Argiriadi, M. A.; Banach, D. L.; Fiamengo, B. A.; Foley, S. E.; Frank, K. E.; George, J. S.; Harris, C. M.; Hobson, A. D.; Ihle, D. C.; Marcotte, D.; Merta, P. J.; Michalak, M. E.; Murdock, S. E.; Tomlinson, M. J.; Voss, J. W. Tricyclic Covalent Inhibitors Selectively Target Jak3 through an Active Site Thiol. J. Biol. Chem. 2015, 290 (8), 45734589,  DOI: 10.1074/jbc.M114.595181
    64. 64
      Chen, C.; Lu, D.; Sun, T.; Zhang, T. JAK3 Inhibitors for the Treatment of Inflammatory and Autoimmune Diseases: A Patent Review (2016-Present). Expert Opin. Ther. Pat. 2022, 32 (3), 225242,  DOI: 10.1080/13543776.2022.2023129
    65. 65
      Shindo, N.; Ojida, A. Recent Progress in Covalent Warheads for in Vivo Targeting of Endogenous Proteins. Bioorg. Med. Chem. 2021, 47, 116386,  DOI: 10.1016/j.bmc.2021.116386
    66. 66
      Forster, M.; Chaikuad, A.; Dimitrov, T.; Döring, E.; Holstein, J.; Berger, B.-T.; Gehringer, M.; Ghoreschi, K.; Müller, S.; Knapp, S.; Laufer, S. A. Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4- d]Pyrrolo[2,3- b]Pyridine Scaffold. J. Med. Chem. 2018, 61 (12), 53505366,  DOI: 10.1021/acs.jmedchem.8b00571
    67. 67
      Laux, J.; Forster, M.; Riexinger, L.; Schwamborn, A.; Guezguez, J.; Pokoj, C.; Kudolo, M.; Berger, L. M.; Knapp, S.; Schollmeyer, D.; Guse, J.; Burnet, M.; Laufer, S. A. Pharmacokinetic Optimization of Small Molecule Janus Kinase 3 Inhibitors to Target Immune Cells. ACS Pharmacol. Transl. Sci. 2022, 5 (8), 573602,  DOI: 10.1021/acsptsci.2c00054
    68. 68
      Burger, J. A. Bruton Tyrosine Kinase Inhibitors: Present and Future. Cancer J. 2019, 25 (6), 386393,  DOI: 10.1097/PPO.0000000000000412
    69. 69
      Khan, W. N. Regulation of B Lymphocyte Development and Activation by Bruton’s Tyrosine Kinase. Immunol. Res. 2001, 23 (2–3), 147156,  DOI: 10.1385/IR:23:2-3:147
    70. 70
      Guldenpfennig, C.; Teixeiro, E.; Daniels, M. NF-kB’s Contribution to B Cell Fate Decisions. Front. Immunol. 2023, 14, 1214095,  DOI: 10.3389/fimmu.2023.1214095
    71. 71
      Staudt, L. M. Oncogenic Activation of NF-kappaB. Cold Spring Harb. Perspect. Biol. 2010, 2 (6), a000109,  DOI: 10.1101/cshperspect.a000109
    72. 72
      Small Molecules in Oncology; Martens, U. M., Ed.; Recent Results in Cancer Research Series; Springer: Berlin, Heidelberg, 2014; Vol. 201.  DOI: 10.1007/978-3-642-54490-3 .
    73. 73
      Wu, J.; Zhang, M.; Liu, D. Acalabrutinib (ACP-196): A Selective Second-Generation BTK Inhibitor. J. Hematol. Oncol.J. Hematol Oncol 2016, 9, 21,  DOI: 10.1186/s13045-016-0250-9
    74. 74
      Guo, Y.; Liu, Y.; Hu, N.; Yu, D.; Zhou, C.; Shi, G.; Zhang, B.; Wei, M.; Liu, J.; Luo, L.; Tang, Z.; Song, H.; Guo, Y.; Liu, X.; Su, D.; Zhang, S.; Song, X.; Zhou, X.; Hong, Y.; Chen, S.; Cheng, Z.; Young, S.; Wei, Q.; Wang, H.; Wang, Q.; Lv, L.; Wang, F.; Xu, H.; Sun, H.; Xing, H.; Li, N.; Zhang, W.; Wang, Z.; Liu, G.; Sun, Z.; Zhou, D.; Li, W.; Liu, L.; Wang, L.; Wang, Z. Discovery of Zanubrutinib (BGB-3111), a Novel, Potent, and Selective Covalent Inhibitor of Bruton’s Tyrosine Kinase. J. Med. Chem. 2019, 62 (17), 79237940,  DOI: 10.1021/acs.jmedchem.9b00687
    75. 75
      Liclican, A.; Serafini, L.; Xing, W.; Czerwieniec, G.; Steiner, B.; Wang, T.; Brendza, K. M.; Lutz, J. D.; Keegan, K. S.; Ray, A. S.; Schultz, B. E.; Sakowicz, R.; Feng, J. Y. Biochemical Characterization of Tirabrutinib and Other Irreversible Inhibitors of Bruton’s Tyrosine Kinase Reveals Differences in on - and off - Target Inhibition. Biochim. Biophys. Acta Gen. Subj. 2020, 1864 (4), 129531,  DOI: 10.1016/j.bbagen.2020.129531
    76. 76
      Tam, C. S.; Muñoz, J. L.; Seymour, J. F.; Opat, S. Zanubrutinib: Past, Present, and Future. Blood Cancer J. 2023, 13 (1), 141,  DOI: 10.1038/s41408-023-00902-x
    77. 77
      Estupiñán, H. Y.; Berglöf, A.; Zain, R.; Smith, C. I. E. Comparative Analysis of BTK Inhibitors and Mechanisms Underlying Adverse Effects. Front. Cell Dev. Biol. 2021, 9, 630942,  DOI: 10.3389/fcell.2021.630942
    78. 78
      Mato, A. R.; Nabhan, C.; Thompson, M. C.; Lamanna, N.; Brander, D. M.; Hill, B.; Howlett, C.; Skarbnik, A.; Cheson, B. D.; Zent, C.; Pu, J.; Kiselev, P.; Goy, A.; Claxton, D.; Isaac, K.; Kennard, K. H.; Timlin, C.; Landsburg, D.; Winter, A.; Nasta, S. D.; Bachow, S. H.; Schuster, S. J.; Dorsey, C.; Svoboda, J.; Barr, P.; Ujjani, C. S. Toxicities and Outcomes of 616 Ibrutinib-Treated Patients in the United States: A Real-World Analysis. Haematologica 2018, 103 (5), 874879,  DOI: 10.3324/haematol.2017.182907
    79. 79
      Sharman, J. P.; Black-Shinn, J. L.; Clark, J.; Bitman, B. Understanding Ibrutinib Treatment Discontinuation Patterns for Chronic Lymphocytic Leukemia. Blood 2017, 130 (Supplement 1), 4060,  DOI: 10.1182/blood.V130.Suppl_1.4060.4060
    80. 80
      Winqvist, M.; Andersson, P.-O.; Asklid, A.; Karlsson, K.; Karlsson, C.; Lauri, B.; Lundin, J.; Mattsson, M.; Norin, S.; Sandstedt, A.; Rosenquist, R.; Späth, F.; Hansson, L.; Österborg, A. for the Swedish CLL Group. Long-Term Real-World Results of Ibrutinib Therapy in Patients with Relapsed or Refractory Chronic Lymphocytic Leukemia: 30-Month Follow up of the Swedish Compassionate Use Cohort. Haematologica 2019, 104 (5), e208e210,  DOI: 10.3324/haematol.2018.198820
    81. 81
      ClinicalTrials.gov. Rilzabrutinib. https://clinicaltrials.gov/search?intr=Rilzabrutinib (accessed 2023–11–28).
    82. 82
      Oda, K. New Families of Carboxyl Peptidases: Serine-Carboxyl Peptidases and Glutamic Peptidases. J. Biochem. (Tokyo) 2012, 151 (1), 1325,  DOI: 10.1093/jb/mvr129
    83. 83
      Ćwilichowska, N.; Świderska, K. W.; Dobrzyń, A.; Drąg, M.; Poręba, M. Diagnostic and Therapeutic Potential of Protease Inhibition. Mol. Aspects Med. 2022, 88, 101144,  DOI: 10.1016/j.mam.2022.101144
    84. 84
      Manasanch, E. E.; Orlowski, R. Z. Proteasome Inhibitors in Cancer Therapy. Nat. Rev. Clin. Oncol. 2017, 14 (7), 417433,  DOI: 10.1038/nrclinonc.2016.206
    85. 85
      Flint, M.; Mullen, S.; Deatly, A. M.; Chen, W.; Miller, L. Z.; Ralston, R.; Broom, C.; Emini, E. A.; Howe, A. Y. M. Selection and Characterization of Hepatitis C Virus Replicons Dually Resistant to the Polymerase and Protease Inhibitors HCV-796 and Boceprevir (SCH 503034). Antimicrob. Agents Chemother. 2009, 53 (2), 401411,  DOI: 10.1128/AAC.01081-08
    86. 86
      Augeri, D. J.; Robl, J. A.; Betebenner, D. A.; Magnin, D. R.; Khanna, A.; Robertson, J. G.; Wang, A.; Simpkins, L. M.; Taunk, P.; Huang, Q.; Han, S.-P.; Abboa-Offei, B.; Cap, M.; Xin, L.; Tao, L.; Tozzo, E.; Welzel, G. E.; Egan, D. M.; Marcinkeviciene, J.; Chang, S. Y.; Biller, S. A.; Kirby, M. S.; Parker, R. A.; Hamann, L. G. Discovery and Preclinical Profile of Saxagliptin (BMS-477118): A Highly Potent, Long-Acting, Orally Active Dipeptidyl Peptidase IV Inhibitor for the Treatment of Type 2 Diabetes. J. Med. Chem. 2005, 48 (15), 50255037,  DOI: 10.1021/jm050261p
    87. 87
      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 (5), 18131822,  DOI: 10.1128/AAC.50.5.1813-1822.2006
    88. 88
      Focosi, D.; McConnell, S.; Shoham, S.; Casadevall, A.; Maggi, F.; Antonelli, G. Nirmatrelvir and COVID-19: Development, Pharmacokinetics, Clinical Efficacy, Resistance, Relapse, and Pharmacoeconomics. Int. J. Antimicrob. Agents 2023, 61 (2), 106708,  DOI: 10.1016/j.ijantimicag.2022.106708
    89. 89
      Wen, W.; Qi, Z.; Wang, J. The Function and Mechanism of Enterovirus 71 (EV71) 3C Protease. Curr. Microbiol. 2020, 77 (9), 19681975,  DOI: 10.1007/s00284-020-02082-4
    90. 90
      Zeng, D.; Ma, Y.; Zhang, R.; Nie, Q.; Cui, Z.; Wang, Y.; Shang, L.; Yin, Z. Synthesis and Structure-Activity Relationship of α-Keto Amides as Enterovirus 71 3C Protease Inhibitors. Bioorg. Med. Chem. Lett. 2016, 26 (7), 17621766,  DOI: 10.1016/j.bmcl.2016.02.039
    91. 91
      Zhai, Y.; Ma, Y.; Ma, F.; Nie, Q.; Ren, X.; Wang, Y.; Shang, L.; Yin, Z. Structure-Activity Relationship Study of Peptidomimetic Aldehydes as Enterovirus 71 3C Protease Inhibitors. Eur. J. Med. Chem. 2016, 124, 559573,  DOI: 10.1016/j.ejmech.2016.08.064
    92. 92
      Luo, Y. L. Mechanism-Based and Computational-Driven Covalent Drug Design. J. Chem. Inf. Model. 2021, 61 (11), 53075311,  DOI: 10.1021/acs.jcim.1c01278
    93. 93
      Mihalovits, L. M.; Ferenczy, G. G.; Keserű, G. M. Affinity and Selectivity Assessment of Covalent Inhibitors by Free Energy Calculations. J. Chem. Inf. Model. 2020, 60 (12), 65796594,  DOI: 10.1021/acs.jcim.0c00834
    94. 94
      Mihalovits, L. M.; Ferenczy, G. G.; Keserű, G. M. The Role of Quantum Chemistry in Covalent Inhibitor Design. Int. J. Quantum Chem. 2022, 122 (8), e26768  DOI: 10.1002/qua.26768
    95. 95
      Lonsdale, R.; Burgess, J.; Colclough, N.; Davies, N. L.; Lenz, E. M.; Orton, A. L.; Ward, R. A. Expanding the Armory: Predicting and Tuning Covalent Warhead Reactivity. J. Chem. Inf. Model. 2017, 57 (12), 31243137,  DOI: 10.1021/acs.jcim.7b00553
    96. 96
      Oballa, R. M.; Truchon, J.-F.; Bayly, C. I.; Chauret, N.; Day, S.; Crane, S.; Berthelette, C. A Generally Applicable Method for Assessing the Electrophilicity and Reactivity of Diverse Nitrile-Containing Compounds. Bioorg. Med. Chem. Lett. 2007, 17 (4), 9981002,  DOI: 10.1016/j.bmcl.2006.11.044
    97. 97
      Copeland, R. A.; Pompliano, D. L.; Meek, T. D. Drug-Target Residence Time and Its Implications for Lead Optimization. Nat. Rev. Drug Discovery 2006, 5 (9), 730739,  DOI: 10.1038/nrd2082
    98. 98
      Bernetti, M.; Masetti, M.; Rocchia, W.; Cavalli, A. Kinetics of Drug Binding and Residence Time. Annu. Rev. Phys. Chem. 2019, 70, 143171,  DOI: 10.1146/annurev-physchem-042018-052340
    99. 99
      Ren, T.; Zhu, X.; Jusko, N. M.; Krzyzanski, W.; Jusko, W. J. Pharmacodynamic Model of Slow Reversible Binding and Its Applications in Pharmacokinetic/Pharmacodynamic Modeling: Review and Tutorial. J. Pharmacokinet. Pharmacodyn. 2022, 49 (5), 493510,  DOI: 10.1007/s10928-022-09822-y
    100. 100
      Frühauf, A.; Wolff, B.; Schweipert, M.; Meyer-Almes, F.-J. Synthesis and Characterization of Reversible Covalent HDAC4 Inhibitors. Methods Mol. Biol. Clifton NJ. 2023, 2589, 207221,  DOI: 10.1007/978-1-0716-2788-4_14
    101. 101
      Forman, H. J.; Zhang, H.; Rinna, A. Glutathione: Overview of Its Protective Roles, Measurement, and Biosynthesis. Mol. Aspects Med. 2009, 30 (1–2), 112,  DOI: 10.1016/j.mam.2008.08.006
    102. 102
      Wu, G.; Fang, Y.-Z.; Yang, S.; Lupton, J. R.; Turner, N. D. Glutathione Metabolism and Its Implications for Health. J. Nutr. 2004, 134 (3), 489492,  DOI: 10.1093/jn/134.3.489
    103. 103
      Bansal, A.; Simon, M. C. Glutathione Metabolism in Cancer Progression and Treatment Resistance. J. Cell Biol. 2018, 217 (7), 22912298,  DOI: 10.1083/jcb.201804161
    104. 104
      Bajic, V. P.; Van Neste, C.; Obradovic, M.; Zafirovic, S.; Radak, D.; Bajic, V. B.; Essack, M.; Isenovic, E. R. Glutathione “Redox Homeostasis” and Its Relation to Cardiovascular Disease. Oxid. Med. Cell. Longev. 2019, 2019, 5028181,  DOI: 10.1155/2019/5028181
    105. 105
      Fu, L.; Li, Z.; Liu, K.; Tian, C.; He, J.; He, J.; He, F.; Xu, P.; Yang, J. A Quantitative Thiol Reactivity Profiling Platform to Analyze Redox and Electrophile Reactive Cysteine Proteomes. Nat. Protoc. 2020, 15 (9), 28912919,  DOI: 10.1038/s41596-020-0352-2
    106. 106
      Kuljanin, M.; Mitchell, D. C.; Schweppe, D. K.; Gikandi, A. S.; Nusinow, D. P.; Bulloch, N. J.; Vinogradova, E. V.; Wilson, D. L.; Kool, E. T.; Mancias, J. D.; Cravatt, B. F.; Gygi, S. P. Reimagining High-Throughput Profiling of Reactive Cysteines for Cell-Based Screening of Large Electrophile Libraries. Nat. Biotechnol. 2021, 39 (5), 630641,  DOI: 10.1038/s41587-020-00778-3
    107. 107
      Yamamoto, M.; Kensler, T. W.; Motohashi, H. The KEAP1-NRF2 System: A Thiol-Based Sensor-Effector Apparatus for Maintaining Redox Homeostasis. Physiol. Rev. 2018, 98 (3), 11691203,  DOI: 10.1152/physrev.00023.2017
    108. 108
      Unoki, T.; Akiyama, M.; Kumagai, Y. Nrf2 Activation and Its Coordination with the Protective Defense Systems in Response to Electrophilic Stress. Int. J. Mol. Sci. 2020, 21 (2), 545,  DOI: 10.3390/ijms21020545
    109. 109
      Backus, K. M.; Cao, J.; Maddox, S. M. Opportunities and Challenges for the Development of Covalent Chemical Immunomodulators. Bioorg. Med. Chem. 2019, 27 (15), 34213439,  DOI: 10.1016/j.bmc.2019.05.050
    110. 110
      Lincoln, R.; Zhang, W.; Lovell, T. C.; Jodko-Piórecka, K.; Devlaminck, P. A.; Sakaya, A.; Van Kessel, A.; Cosa, G. Chemically Tuned, Reversible Fluorogenic Electrophile for Live Cell Nanoscopy. ACS Sens. 2022, 7 (1), 166174,  DOI: 10.1021/acssensors.1c01940
    111. 111
      Zheng, S.; Liu, G. Polymeric Emissive Materials Based on Dynamic Covalent Bonds. Mol. Basel Switz. 2022, 27 (19), 6635,  DOI: 10.3390/molecules27196635