Abstract
Traditional biochemical target-based and phenotypic cell-based screening approaches to drug discovery have produced the current covalent and non-covalent pharmacopoeia. Strategies to expand the druggable proteome include Inverse Drug Discovery, which involves incubating one weak organic electrophile at a time with the proteins of a living cell to identify the conjugates formed. An alkyne substructure in each organic electrophile enables affinity chromatography–mass spectrometry, which produces a list of proteins that each distinct compound reacts with. Herein, we review Inverse Drug Discovery in the context of organic compounds of intermediate complexity harboring Sulfur(VI)-fluoride exchange (SuFEx) electrophiles used to expand the cellular proteins that can be targeted covalently.
Introduction
Drug discovery programs seek to identify molecules capable of alleviating the impact of diseases. Through iterative refinement, this effort produces a lead molecule, which engages a specific target to produce disease-relevant functional changes [1]. The most common approaches to drug discovery are in vitro screens against a biochemical target or cell-based phenotypic screens–both employing large collections of structurally diverse small molecules (Fig. 1A, B). Traditional drug discovery approaches are not without their pitfalls. A target-based biochemical screening approach requires extensive knowledge about the target and its relevance to pathophysiology to properly transform screening hits into potent and selective lead compounds. Moreover, targeting a particular protein in vitro might not directly translate to clinically relevant outcomes [2]. Phenotypic screening relies on examining observable changes in cellular phenotype mediated by small molecule treatment. Target identification and mechanistic validation still prove to be non-trivial, although recent chemoproteomic and genetic advancements help address these challenges associated with phenotypic screening [3,4].
Figure 1. Screening Methodologies for Drug Discovery.

A. A target-based approach first involves identifying a small-molecule-amenable causative protein target involved in the pathophysiology of a given disease. A library of compounds is screened against the given target to identify high affinity-binding molecules, which can be developed into a lead molecule primed for validation and optimization in vivo. B. A phenotypic-based approach employs a large library of compounds screened for eliciting a desired phenotypic outcome in a model organism or cell line. Target deconvolution and validation is necessary to determine the mechanism of action of initial hits before medicinal chemistry. C. Inverse Drug Discovery leverages a small library of alkyne-containing latent electrophiles that are screened in various cell lines, followed by identification of the disease-relevant target(s) and optimization of a lead molecule.
Despite the history of using covalent protein ligands in pharmacology to demonstrate proof-of-principle, trepidation remains over their use as drugs because of possible off-target reactivity and toxicity concerns [5,6]. Most covalent drugs gained approval from regulatory agencies before their covalent nature was known. However, the recently FDA-approved drugs ibrutinib, a BTK inhibitor [7], and telaprevir, a serine protease inhibitor [8], were established to be covalent prior to their approval, reflecting a change in attitude in the pharmaceutical industry and within regulatory agencies. Covalent drugs form a chemical bond with a proteinogenic nucleophile as a means to modulate protein activity [9]. A common strategy to minimize reactivity towards closely related protein family members is through using covalent inhibitors that target a distal nucleophile rather than the catalytic nucleophile [10,11].
Activity-based protein profiling (ABPP) exploits active-site directed chemical probes to survey enzymatic family function, which can be leveraged for the discovery of unique ligands against a particular protein [12–14]. Reactivity-based probes extend this approach to survey the global proteome reactivity of a given amino-acid side chain (e.g., the thiol of cysteine), and can similarly provide a successful pathway towards discovery of novel covalent ligands that target a particular protein [15–18]. Quantitative mass spectrometric methods are critical for assessing conjugate formation between proteins and broadly reactive probes harboring a reporter tag such as an alkyne. New covalent ligands can be discovered by monitoring decreases in probe labeling intensity for a given protein when co-treated with an inhibitor molecule exhibiting increased selectivity. Activity-Based Protein Profiling provides valuable information about protein families using highly reactive electrophiles. In contrast, utilizing less reactive electrophiles, or electrophiles that need to be activated by protein binding prior to becoming reactive (latent electrophiles), may allow greater selectivity towards protein nucleophiles in a target-agnostic manner. In this regard, latent electrophiles, i.e., electrophilic species exhibiting environment-dependent protein reactivity towards a single protein or a small number of proteins, but inert towards the majority of the cellular proteome, provide another option to minimize possible off-target activity and subsequent toxicity [19]. Herein, we review an alternate screening strategy, hereafter referred to as Inverse Drug Discovery, that has its conceptual origins in ABPP, and employs latent electrophiles for covalent modification of disease-relevant protein targets (Fig. 1C).
Inverse drug discovery rationale and workflow
Inverse Drug Discovery (IDD) matches structurally unique compounds of intermediate complexity harboring a weak electrophile with proteins capable of undergoing reaction in the context of a living cell or cell lysate. As reversible binding precedes conjugate formation, utilizing structures of intermediate complexity with distinct three-dimensional structures is necessary to ensure reasonable binding affinity (KD’s in the μM range) with only a subset of the human proteome having an appropriate arrangement of side chain functionality to allow reaction with the latent electrophile. In IDD, the scaffolds of intermediate structural complexity are compounds with a higher molecular weight than a typical fragment (>230 Da) often adhering to Lipinski’s rule of 5 also incorporating a conjugation handle (e.g., an alkyne), which facilitates affinity chromatography–mass spectrometric analysis [20,21]. This methodology generates a list of the proteins reacting with each chemically unique structure harboring a latent electrophile and an alkyne. The designation “inverse” derives from the fact that we are screening the proteome of a living cell against one small molecule. This contrasts with traditional drug discovery approaches, which screen thousands to millions of compounds individually, seeking activity against a distinct target or alteration of a specific cellular phenotype.
An IDD campaign begins with the synthesis of a small collection of alkyne-containing small molecules of intermediate complexity that harbor weak, but activatable electrophiles. Novel synthetic routes to SuFEx-containing electrophiles have dramatically expanded the available scaffolds for IDD [22]. Preequilibrium binding allows for a proximity-driven reaction between the latent electrophile and proteinogenic side chain nucleophiles (i.e., cysteine, lysine, tyrosine) [23]. Reaction of the alkyne tag with biotin azide via a CuAAC “click reaction” or Azide-Alkyne cycloaddition reaction allows affinity enrichment of liganded proteins and subsequent identification of protein targets. After prioritizing the small molecule-protein conjugates for pathobiological relevance, x-ray crystallography allows validation of bond-forming reactions between a given small molecule and a recombinantly-produced protein target, often identifying the amino acid side chain involved in the conjugation reaction [19,24]. As crystallization is not possible for some proteins, peptide mass fingerprinting or cryogenic electron microscopy often proves useful for identifying the conjugation bond formed [25–27].
Mass spectrometry (MS) is critical to the IDD workflow, which harnesses quantitative proteomics to identify the proteins covalently targeted. Following treatment of cells with a small molecule latent electrophile, biotin attachment, affinity enrichment, and tryptic digestion, the peptides afforded are subjected to two or three MS measurements done in tandem, where the mass-to-charge (m/z) ratio of digested and fragmented peptides is detected [28,29]. The m/z ratio and fragmentation pattern allow for identification of the peptide sequences, thus leading to final identification of the target proteins [28]. Knowledge of the modification site is critical for future optimization of the lead molecule; however, the increased hydrophobicity and altered charge state of probe-modified peptides can hinder identification. Novel methods leveraging predictable probe-modified peptide gas-phase fragmentation pathways or electron-transfer dissociation fragmentation have proven useful for identification of the site of modification for cysteine, tyrosine, and lysine side chains, respectively [30–32].
Previous IDD Probes based on Arylfluorosulfates and Sulfuramidimidoyl Fluorides
Sulfur(VI)-fluoride exchange (SuFEx)-functional groups are a prototypical class of latent electrophiles [33]. In the context of an appropriate preequilibrium binding step to a protein having the right constellation of side chains, SuFEx chemistry ensues, which involves nucleophilic attack at the Sulfur(VI) atom followed by concomitant loss of the fluoride ion. Nearby arginine and lysine side chains in the protein undergoing reaction appear to catalyze fluoride displacement [19,34]. SuFEx functional groups are relatively stable towards buffer, and, more importantly, inert towards denatured proteins, and most other macromolecules, including most of the folded proteome lacking the ability to bind and undergo reaction with the latent electrophile [23]. The most-recognized class of SuFEx-derived electrophiles are sulfonyl fluorides, which have historically been used to target serine, lysine, or tyrosine residues [33–39]. However, sulfonyl fluorides are highly reactive and are not ideally suited for IDD [38]. Thus far, IDD has utilized less electrophilic arylfluorosulfates and sulfuramidimidoyl fluorides as weakly-reactive latent electrophiles [19,24] (Fig. 2).
Figure 2. SuFEx chemistry applied to Inverse Drug Discovery.

A. Structure of SuFEx hubs used for proteome reactivity studies with the SuFEx hub highlighted in red and sites of structural diversity denoted as colored shapes. B. Key to SuFEx-derived electrophile●protein conjugate formation is hydrogen-bond donor mediated activation-stabilization of the fluoride ion, and sufficient molecular complementarity between protein and scaffold to allow a proximal nucleophilic residue to attack. C. X-Ray structure of GST01 conjugated to Probe 1 at Tyr229, with Lys57 and Lys59 likely mediating conjugate formation through pKa perturbation, visualized using UCSF Chimera [85].
The Kelly and Sharpless labs identified arylfluorosulfates as having reactivity towards only a small subset of the human cellular proteome, including intracellular lipid-binding proteins (Table 1) using organic substructures that can bind in such a way as to facilitate the SuFEx conjugation reaction [23,34]. Moreover, the excellent cell permeability and relative buffer stability of arylfluorosulfates—exemplified by stability in basic phosphate buffer for two weeks and neutral buffers for months—highlight their utility as pharmacological tools [23,34,36,40]. Accordingly, arylfluorosulfate probes attached to different organic structures of intermediate structural complexity were found to react with different protein targets (Table 1) [19]. Notably, arylfluorosulfate Probe 1, a quinolinic acid-based molecule, was found to react with GSTO1, a stress responsive protein which has been shown to be upregulated in drug resistance cancer (Fig. 2C), as well as HSDL2, a potential therapeutic target for glioma [19,41,42]. The orientations of side chain residues favorable for SuFEx reactions appear to be a tyrosine nucleophile flanked by adjacent arginine/lysine side chains, as exemplified by GSTO1 wherein Tyr229 is adjacent to Lys 57/Lys59 [19].
Table 1.
Structures of SuFEx-derived electrophiles and corresponding disease-relevant protein targets they selectively react with in whole proteome experiments. Probe numbers in “Name” are those used in the cited paper.
| Structure | Name | Proteins Targeted | Disease Indication | Paper |
|---|---|---|---|---|
|
|
Biphenyl Arylfluorosulfate Probe 4 | CRABP2 | Cancer/Osteoarthritis | Chen et al, 2016 |
| FBP4 | Diabetes/Atherosclerosis | |||
|
Quinolinic Acid-Based Arylfluorosulfate Probe 1 | GSTO1 | Drug Resistant Cancer | Mortenson et al, 2018 |
| HSDL2 | Glioma | |||
| TIGAR | Glioma | |||
|
Imidazole-Based Arylfluorosulfate Probe 2 | GSTP1 | Drug Resistant Cancer | |
| NME1 | Cancer | |||
|
Imidazole-Based Arylfluorosulfate Probe 3 | TTR | TTR Amyloidosis | |
| CRABP2 | Cancer/Osteoarthritis | |||
|
Thymidine-Based SAF Probe 2 | PARP1 | BRCA1/BRCA2-Deficient Cancer | Brighty et al., 2020 |
|
Nitropyridine-Based SAF Probe 13 | MIF | Atherosclerosis |
Brighty and coworkers recently employed terminal acetylene-containing sulfuramidimidoyl fluorides (SAFs) to extend the capacity of SuFEx-derived electrophiles used in IDD [24]. SAFs are less electrophilic than their arylfluorosulfate counterparts and possess chirality from replacement of the arylfluorosulfates’ S=O bond with a S=NR bond (Fig. 2A). While a small subset of conjugates identified overlapped with previously described arylfluorosulfate SuFEx-reactive proteins, many of the SAF-reactive proteins were unique (Table 1) [19,24,34,43–46]. Of note, a thymidine-based SAF small molecule reacted with the catalytic domain of PARP1, a key DNA repair enzyme commonly targeted by BRCA1/BRCA2-deficient cancer therapies [47,48]. This conjugation led to reduced PARylation in HeLa cells, i.e., inhibition of the enzyme [24]. Coupling multiple organic substructures of intermediate complexity through a SAF connection is an attractive option for developing promising ligands for other therapeutically relevant targets, while minimizing off-target reactivity owing to the inherently lower SAF reactivity.
New Classes of Latent Electrophiles
Electrophile class often dictates the sector of the proteome amenable to covalent labeling, and this has been reviewed elsewhere [11,39]. However, novel chemoproteomic methods have shown that some electrophiles that were thought to be amino-acid-selective are quite promiscuous [49]. Further confounding matters, electrophiles targeting a given amino acid often have differential proteome reactivity, indicating that local stereoelectronic factors intrinsic to a particular protein influence selectivity. As such, recently discovered latent electrophiles may be utilized for IDD applications, as discussed here (Fig. 3).
Figure 3. Novel Classes of Latent Electrophiles.

A. Representative novel classes of recently discovered latent electrophiles. Site of reactivity highlighted in red (cysteine reactive), yellow (tyrosine reactive), and orange (aspartate/glutamate reactive). B. Electrophiles that react through nucleophilic aromatic substitution elicit dramatically different proteome reactivity. C. Metabolically activatable electrophiles, such as 147 (top) formation of quinone methide, and ML210 (bottom) formation of nitrile oxide, react with small subset of the proteome.
Novel latent electrophiles have been recently developed to circumvent off-target reactivity associated with traditional cysteine-targeted electrophiles, which have been extensively reviewed elsewhere [50,51]. Examples include α-chlorofluoroacetamides (reversible covalency), bicyclobutane carboxylic acid (strain release), and thiol-alkyne/methacrylamide reaction (proximity-driven) [52–56] (Fig. 3A). Many of these electrophiles may be amenable to IDD-screening efforts on the appropriate organic substructure of intermediate complexity. Proximity-driven covalent chemistry has been extended to lysine-selective electrophiles, where employing the N-acyl-N-alkyl-sulfonamide (NASA) warhead allows conjugate formation only with sufficient ligand-protein interaction [57] (Fig. 3A). Ligand-directed NASA warheads have found utility for HSP90 inhibition and blocking the HDM2/p53 protein-protein interaction, with other applications surely to come [57,58].
Tyrosine is among the least abundant of all amino acids, and its local environment within a protein has substantial effects on its reactivity [59–61]. In analogous fashion to SuFEx chemistry, the generally more reactive Sulfur(VI)-triazole exchange (SuTEx) chemistry utilizes 1,2,4-triazoles as a leaving group from the Sulfur(VI) hub for reaction with specific tyrosine residues [62] (Fig. 3A). This technology allowed for quantification of both phosphotyrosine sites upon pervanadate treatment and annotation of hyperreactive tyrosine residues, with enhanced proteome coverage compared with traditional sulfonyl-fluoride-based approaches [63]. Importantly, SuTEx ligands could be amenable to the IDD platform, as modulating electronics of the adduct group and leaving group imbues unique proteome reactivity [64]. Exemplifying this is the discovery of a recent SuTEx ligand for modulating prostaglandin reductase 2 [65]. Another underexplored mode of reactivity is through nucleophilic aromatic substitution (SNAr) [66]. Some electrophiles, such as 2-sulfonylpyridines, are predominantly cysteine selective, whereas others, such as triazine-based scaffolds, can target tyrosines or lysines [67–69] (Fig. 3B). The varied amino-acid selectivity of SNAr-based electrophiles indicates that protein microenvironment plays a larger role in conjugate formation than nucleophilicity of a given residue. As a result of this promiscuity, SNAr-based scaffolds may be amenable for IDD.
Covalent ligands directed at carboxylate-containing side chains in proteins have hitherto been relatively unexplored, due to the lack of inherent nucleophilicity. Nevertheless, functionally important glutamate and aspartate may play a critical role in human health and disease, and thus warrant interest for covalent ligand development. The 3-phenyl-2H-azirine moiety covalently labels reactive carboxylic acid residues in vitro and in vivo, such as annotated active site residues [70]. Similarly, alkyl diazirines have distinct reactivity towards carboxylate side chains acids residing on negative electrostatic surfaces [71,72]. Finally, unbiased chemoproteomic methods uncovered novel carboxylic acid-directed ligands, such as hydrazonyl chlorides [73]. Elaborating these electrophiles into more complex scaffolds, and tuning their reactivity may allow target-agnostic discovery of novel ligandable proteins, as has been previously accomplished with fragment-based approaches [74].
A final class of latent electrophiles warranting discussion are those which are metabolically activated to generate reactive electrophilic species capable of protein-conjugate formation proximal to the site of metabolite generation. Some examples include the masked nitrile oxide ferroptosis inducer, ML210, and the quinone methide producing ATF6 activator, 147, among others [75–78] (Fig. 3C). While it is difficult to predict a priori whether electrophilic metabolite generation will occur, understanding the selectivity of these reactive moieties, and others yet-to-be discovered, will surely facilitate expansion of the chemically tractable proteome.
Conclusion and Future Directions
The IDD strategy is a method to connect weak electrophiles attached to organic substructures of intermediate complexity to the human proteins in a cell they react with. Using latent electrophiles to discover covalent ligands for various proteins should reduce off-target reactivity inherent to more reactive functional groups. The target-agnostic nature of the IDD workflow delays decisions about which disease etiology to focus on until after conjugate identification. In addition, choosing organic substructures with a high degree of modularity eases requisite diversification of the initial hit into more potent lead molecules. IDD is conceptually similar to activity-based protein profiling, which typically employs more reactive electrophiles and is related to fragment-based drug discovery efforts. Work in this arena has dramatically expanded the chemical space surrounding the ligandable proteome [15,74,79–82]. While the organic substructure drives proximity-induced conjugate formation through equilibrium binding, the chosen electrophile can also bias proteome reactivity and chemoselectivity, thus both components can be altered to enhance conjugate selectivity [53]. Swapping the sulfonyl fluoride for an arylfluorosulfate on the dcpS targeting-diaminoquinazoline scaffold led to reaction with Ser272 versus Tyr 113/Tyr143, and such a change may abolish reactivity completely in other cases [36]. The gold standard for an IDD-derived pharmaceutical would be an electrophile-harboring scaffold that reacts with only one cellular protein. While some novel electrophiles possess reactivity towards a single target, this selectivity originates from chemical reactivity of the nucleophilic residue, rather than proximity-driven reactivity linked to the 3-dimensional side chain orientation within a protein [83,84]. Such a scenario hinders further medicinal chemistry optimization. Future medicinal chemistry efforts aim to design structures harboring weakly activatable electrophiles capable of reaction with only a single member of the entire human proteome. Advances in understanding the reactivity of unique latent electrophiles will hopefully aid in this endeavor.
Acknowledgements
This review was made possible by support from the Skaggs Institute for Chemical Biology, the Lita Annenberg Hazen Foundation, and by the National Institutes of Health grant DK046335 (J.W.K.). Molecular graphics and analyses were performed with UCSF Chimera, developed by the Resource for Biocomputing, Visualization, and Informatics at the University of California, San Francisco, with support from NIH P41-GM103311. We acknowledge Dr. Evan T. Powers, Lynée A. Massey, Dr. Colleen Fearns, Dr. Kyunga Lee, and Leonard Yoon for comments and editing of the manuscript. Figures were created with BioRender.com
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- 1.Hughes JP, Rees S, Kalindjian SB, Philpott KL: Principles of early drug discovery Br J Pharmacol, 162 (2011), pp. 1239–1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Swinney DC, Lee JA: Recent advances in phenotypic drug discovery F1000Res, 9 (2020), pp. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mullard A: The phenotypic screening pendulum swings Nat Rev Drug Discov, 14 (2015), pp. 807–809. [DOI] [PubMed] [Google Scholar]
- 4.Nijman SM: Functional genomics to uncover drug mechanism of action Nat Chem Biol, 11 (2015), pp. 942–948. [DOI] [PubMed] [Google Scholar]
- 5.Zhou S, Chan E, Duan W, Huang M, Chen YZ: Drug bioactivation, covalent binding to target proteins and toxicity relevance Drug Metab Rev, 37 (2005), pp. 41–213. [DOI] [PubMed] [Google Scholar]
- 6.Lanning BR, Whitby LR, Dix MM, Douhan J, Gilbert AM, Hett EC, Johnson TO, Joslyn C, Kath JC, Niessen S, et al. : A road map to evaluate the proteome-wide selectivity of covalent kinase inhibitors Nat Chem Biol, 10 (2014), pp. 760–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Brown JR: Ibrutinib (PCI-32765), the first BTK (Bruton’s tyrosine kinase) inhibitor in clinical trials Curr Hematol Malig Rep, 8 (2013), pp. 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lin C, Kwong AD, Perni RB: Discovery and development of VX-950, a novel, covalent, and reversible inhibitor of hepatitis C virus NS3.4A serine protease Infect Disord Drug Targets, 6 (2006), pp. 3–16. [DOI] [PubMed] [Google Scholar]
- 9.Singh J, Petter RC, Baillie TA, Whitty A: The resurgence of covalent drugs Nat Rev Drug Discov, 10 (2011), pp. 307–317. [DOI] [PubMed] [Google Scholar]
- 10.De Vita E: 10 years into the resurgence of covalent drugs Future Med Chem, 13 (2021), pp. 193–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.*.Zhang T, Hatcher JM, Teng M, Gray NS, Kostic M: Recent Advances in Selective and Irreversible Covalent Ligand Development and Validation Cell Chem Biol, 26 (2019), pp. 1486–1500. [DOI] [PMC free article] [PubMed] [Google Scholar]; This review outlines approaches to validate novel covalent ligands from a chemical biology viewpoint, advances in finding new amino-acid selective chemotypes, and screening approaches for finding novel covalent ligands
- 12.Cravatt BF, Wright AT, Kozarich JW: Activity-based protein profiling: from enzyme chemistry to proteomic chemistry Annu Rev Biochem, 77 (2008), pp. 383–414. [DOI] [PubMed] [Google Scholar]
- 13.Liu Y, Patricelli MP, Cravatt BF: Activity-based protein profiling: the serine hydrolases Proc Natl Acad Sci U S A, 96 (1999), pp. 14694–14699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kidd D, Liu Y, Cravatt BF: Profiling serine hydrolase activities in complex proteomes Biochemistry, 40 (2001), pp. 4005–4015. [DOI] [PubMed] [Google Scholar]
- 15.**.Backus KM, Correia BE, Lum KM, Forli S, Horning BD, Gonzalez-Paez GE, Chatterjee S, Lanning BR, Teijaro JR, Olson AJ, et al. : Proteome-wide covalent ligand discovery in native biological systems Nature, 534 (2016), pp. 570–574. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper utilizes chemoproteomic methods for fragment-based ligand discovery through screening of cysteine-reactive electrophilic fragments. This approach allows the discovery of covalent ligands for numerous relatively untargeted protein classes
- 16.Hacker SM, Backus KM, Lazear MR, Forli S, Correia BE, Cravatt BF: Global profiling of lysine reactivity and ligandability in the human proteome Nat Chem, 9 (2017), pp. 1181–1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Vinogradova EV, Zhang X, Remillard D, Lazar DC, Suciu RM, Wang Y, Bianco G, Yamashita Y, Crowley VM, Schafroth MA, et al. : An Activity-Guided Map of Electrophile-Cysteine Interactions in Primary Human T Cells Cell, 182 (2020), pp. 1009–1026 e1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Weerapana E, Wang C, Simon GM, Richter F, Khare S, Dillon MB, Bachovchin DA, Mowen K, Baker D, Cravatt BF: Quantitative reactivity profiling predicts functional cysteines in proteomes Nature, 468 (2010), pp. 790–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.**.Mortenson DE, Brighty GJ, Plate L, Bare G, Chen W, Li S, Wang H, Cravatt BF, Forli S, Powers ET, et al. : “Inverse Drug Discovery” Strategy To Identify Proteins That Are Targeted by Latent Electrophiles As Exemplified by Aryl Fluorosulfates J Am Chem Soc, 140 (2018), pp. 200–210. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper presents the inverse drug discovery approach using arylfluorosulfates to identify ligand-protein conjugates for each unique chemical structure, including proteins for which no known ligand exists
- 20.Jhoti H, Williams G, Rees DC, Murray CW: The ‘rule of three’ for fragment-based drug discovery: where are we now? Nat Rev Drug Discov, 12 (2013), pp. 644–645. [DOI] [PubMed] [Google Scholar]
- 21.Lipinski CA: Rule of five in 2015 and beyond: Target and ligand structural limitations, ligand chemistry structure and drug discovery project decisions Adv Drug Deliv Rev, 101 (2016), pp. 34–41. [DOI] [PubMed] [Google Scholar]
- 22.Liu Z, Meng G, Guo T, Dong J, Wu P: Novel Approaches to Access Arylfluorosulfates and Sulfamoyl Fluorides Based on Sulfur (VI) Fluoride Exchange Curr Protoc Chem Biol, 11 (2019), pp. e64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.**.Martin-Gago P, Olsen CA: Arylfluorosulfate-Based Electrophiles for Covalent Protein Labeling: A New Addition to the Arsenal Angew Chem Int Ed Engl, 58 (2019), pp. 957–966. [DOI] [PMC free article] [PubMed] [Google Scholar]; This succinct review provides excellent background on the use of arylfluorosulfates, and why they are useful tools for interrogating the proteome
- 24.**.Brighty GJ, Botham RC, Li S, Nelson L, Mortenson DE, Li G, Morisseau C, Wang H, Hammock BD, Sharpless KB, et al. : Using sulfuramidimidoyl fluorides that undergo sulfur(VI) fluoride exchange for inverse drug discovery Nat Chem, 12 (2020), pp. 906–913. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper presents SAFs as a new SuFEx-derived electrophile amenable for Inverse Drug Discovery
- 25.Pinch BJ, Doctor ZM, Nabet B, Browne CM, Seo HS, Mohardt ML, Kozono S, Lian X, Manz TD, Chun Y, et al. : Identification of a potent and selective covalent Pin1 inhibitor Nat Chem Biol, 16 (2020), pp. 979–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pettinger J, Le Bihan YV, Widya M, van Montfort RL, Jones K, Cheeseman MD: An Irreversible Inhibitor of HSP72 that Unexpectedly Targets Lysine-56 Angew Chem Int Ed Engl, 56 (2017), pp. 3536–3540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Van Drie JH, Tong L: Cryo-EM as a powerful tool for drug discovery Bioorg Med Chem Lett, 30 (2020), pp. 127524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Schubert OT, Rost HL, Collins BC, Rosenberger G, Aebersold R: Quantitative proteomics: challenges and opportunities in basic and applied research Nat Protoc, 12 (2017), pp. 1289–1294. [DOI] [PubMed] [Google Scholar]
- 29.Aebersold R, Mann M: Mass spectrometry-based proteomics Nature, 422 (2003), pp. 198–207. [DOI] [PubMed] [Google Scholar]
- 30.Ficarro SB, Browne CM, Card JD, Alexander WM, Zhang T, Park E, McNally R, Dhe-Paganon S, Seo HS, Lamberto I, et al. : Leveraging Gas-Phase Fragmentation Pathways for Improved Identification and Selective Detection of Targets Modified by Covalent Probes Anal Chem, 88 (2016), pp. 12248–12254. [DOI] [PubMed] [Google Scholar]
- 31.McCloud RL, Yuan K, Mahoney KE, Bai DL, Shabanowitz J, Ross MM, Hunt DF, Hsu KL: Direct Target Site Identification of a Sulfonyl-Triazole Covalent Kinase Probe by LC-MS Chemical Proteomics Anal Chem, 93 (2021), pp. 11946–11955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Browne CM, Jiang B, Ficarro SB, Doctor ZM, Johnson JL, Card JD, Sivakumaren SC, Alexander WM, Yaron TM, Murphy CJ, et al. : A Chemoproteomic Strategy for Direct and Proteome-Wide Covalent Inhibitor Target-Site Identification J Am Chem Soc, 141 (2019), pp. 191–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.**.Dong J, Krasnova L, Finn MG, Sharpless KB: Sulfur(VI) fluoride exchange (SuFEx): another good reaction for click chemistry Angew Chem Int Ed Engl, 53 (2014), pp. 9430–9448. [DOI] [PubMed] [Google Scholar]; This seminal review provides a background on SuFEx chemistry and its numerous uses in chemical biology, drug discovery, and organic synthesis
- 34.*.Chen W, Dong J, Plate L, Mortenson DE, Brighty GJ, Li S, Liu Y, Galmozzi A, Lee PS, Hulce JJ, et al. : Arylfluorosulfates Inactivate Intracellular Lipid Binding Protein(s) through Chemoselective SuFEx Reaction with a Binding Site Tyr Residue J Am Chem Soc, 138 (2016), pp. 7353–7364. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper introduces aryfluorosulfates as latent electrophiles capable of reacting with a small number of protein residues
- 35.Baker BR: Specific irreversible enzyme inhibitors Annu Rev Pharmacol, 10 (1970), pp. 35–50. [DOI] [PubMed] [Google Scholar]
- 36.Fadeyi OO, Hoth LR, Choi C, Feng X, Gopalsamy A, Hett EC, Kyne RE Jr., Robinson RP, Jones LH: Covalent Enzyme Inhibition through Fluorosulfate Modification of a Noncatalytic Serine Residue ACS Chem Biol, 12 (2017), pp. 2015–2020. [DOI] [PubMed] [Google Scholar]
- 37.Gu C, Shannon DA, Colby T, Wang Z, Shabab M, Kumari S, Villamor JG, McLaughlin CJ, Weerapana E, Kaiser M, et al. : Chemical proteomics with sulfonyl fluoride probes reveals selective labeling of functional tyrosines in glutathione transferases Chem Biol, 20 (2013), pp. 541–548. [DOI] [PubMed] [Google Scholar]
- 38.Narayanan A, Jones LH: Sulfonyl fluorides as privileged warheads in chemical biology Chem Sci, 6 (2015), pp. 2650–2659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Shannon DA, Weerapana E: Covalent protein modification: the current landscape of residue-specific electrophiles Curr Opin Chem Biol, 24 (2015), pp. 18–26. [DOI] [PubMed] [Google Scholar]
- 40.Mukherjee H, Debreczeni J, Breed J, Tentarelli S, Aquila B, Dowling JE, Whitty A, Grimster NP: A study of the reactivity of S((VI))-F containing warheads with nucleophilic amino-acid side chains under physiological conditions Org Biomol Chem, 15 (2017), pp. 9685–9695. [DOI] [PubMed] [Google Scholar]
- 41.Townsend DM, Tew KD: The role of glutathione-S-transferase in anti-cancer drug resistance Oncogene, 22 (2003), pp. 7369–7375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ruokun C, Yake X, Fengdong Y, Xinting W, Laijun S, Xianzhi L: Lentivirus-mediated silencing of HSDL2 suppresses cell proliferation in human gliomas Tumour Biol, 37 (2016), pp. 15065–15077. [DOI] [PubMed] [Google Scholar]
- 43.Zheng Q, Woehl JL, Kitamura S, Santos-Martins D, Smedley CJ, Li G, Forli S, Moses JE, Wolan DW, Sharpless KB: SuFEx-enabled, agnostic discovery of covalent inhibitors of human neutrophil elastase Proc Natl Acad Sci U S A, 116 (2019), pp. 18808–18814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.*.Hett EC, Xu H, Geoghegan KF, Gopalsamy A, Kyne RE Jr., Menard CA, Narayanan A, Parikh MD, Liu S, Roberts L, et al. : Rational targeting of active-site tyrosine residues using sulfonyl fluoride probes ACS Chem Biol, 10 (2015), pp. 1094–1098. [DOI] [PubMed] [Google Scholar]; This paper leverages structure-based design to create a covalent sulfonyl-fluoride containing probe capable of preferentially reacting with the mRNA decapping enzyme DcpS among the entire proteome
- 45.Zhao Q, Ouyang X, Wan X, Gajiwala KS, Kath JC, Jones LH, Burlingame AL, Taunton J: Broad-Spectrum Kinase Profiling in Live Cells with Lysine-Targeted Sulfonyl Fluoride Probes J Am Chem Soc, 139 (2017), pp. 680–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hanoulle X, Van Damme J, Staes A, Martens L, Goethals M, Vandekerckhove J, Gevaert K: A new functional, chemical proteomics technology to identify purine nucleotide binding sites in complex proteomes J Proteome Res, 5 (2006), pp. 3438–3445. [DOI] [PubMed] [Google Scholar]
- 47.Futreal PA, Liu Q, Shattuck-Eidens D, Cochran C, Harshman K, Tavtigian S, Bennett LM, Haugen-Strano A, Swensen J, Miki Y, et al. : BRCA1 mutations in primary breast and ovarian carcinomas Science, 266 (1994), pp. 120–122. [DOI] [PubMed] [Google Scholar]
- 48.Wooster R, Bignell G, Lancaster J, Swift S, Seal S, Mangion J, Collins N, Gregory S, Gumbs C, Micklem G: Identification of the breast cancer susceptibility gene BRCA2 Nature, 378 (1995), pp. 789–792. [DOI] [PubMed] [Google Scholar]
- 49.Zanon PRA, Yu F, Musacchio P, Lewald L, Zollo M, Krauskopf K, Mrdovic D, Raunft P, Maher TE, Cigler M, et al. : Profiling the proeteome-wide selectivity of diverse electrophiles Chemrxiv, (2021), pp. 10.33774/chemrxiv-32021-w33777rss-v33772. [DOI] [Google Scholar]
- 50.Ray S, Murkin AS: New Electrophiles and Strategies for Mechanism-Based and Targeted Covalent Inhibitor Design Biochemistry, 58 (2019), pp. 5234–5244. [DOI] [PubMed] [Google Scholar]
- 51.Ochtrop P, Hackenberger CPR: Recent advances of thiol-selective bioconjugation reactions Curr Opin Chem Biol, 58 (2020), pp. 28–36. [DOI] [PubMed] [Google Scholar]
- 52.Shindo N, Fuchida H, Sato M, Watari K, Shibata T, Kuwata K, Miura C, Okamoto K, Hatsuyama Y, Tokunaga K, et al. : Selective and reversible modification of kinase cysteines with chlorofluoroacetamides Nat Chem Biol, 15 (2019), pp. 250–258. [DOI] [PubMed] [Google Scholar]
- 53.Tokunaga K, Sato M, Kuwata K, Miura C, Fuchida H, Matsunaga N, Koyanagi S, Ohdo S, Shindo N, Ojida A: Bicyclobutane Carboxylic Amide as a Cysteine-Directed Strained Electrophile for Selective Targeting of Proteins J Am Chem Soc, 142 (2020), pp. 18522–18531. [DOI] [PubMed] [Google Scholar]
- 54.Mons E, Jansen IDC, Loboda J, van Doodewaerd BR, Hermans J, Verdoes M, van Boeckel CAA, van Veelen PA, Turk B, Turk D, et al. : The Alkyne Moiety as a Latent Electrophile in Irreversible Covalent Small Molecule Inhibitors of Cathepsin K J Am Chem Soc, 141 (2019), pp. 3507–3514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Mons E, Kim RQ, van Doodewaerd BR, van Veelen PA, Mulder MPC, Ovaa H: Exploring the Versatility of the Covalent Thiol-Alkyne Reaction with Substituted Propargyl Warheads: A Deciding Role for the Cysteine Protease J Am Chem Soc, 143 (2021), pp. 6423–6433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Reddi RN, Resnick E, Rogel A, Rao BV, Gabizon R, Goldenberg K, Gurwicz N, Zaidman D, Plotnikov A, Barr H, et al. : Tunable Methacrylamides for Covalent Ligand Directed Release Chemistry J Am Chem Soc, 143 (2021), pp. 4979–4992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Tamura T, Ueda T, Goto T, Tsukidate T, Shapira Y, Nishikawa Y, Fujisawa A, Hamachi I: Rapid labelling and covalent inhibition of intracellular native proteins using ligand-directed N-acyl-N-alkyl sulfonamide Nat Commun, 9 (2018), pp. 1870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ueda T, Tamura T, Kawano M, Shiono K, Hobor F, Wilson AJ, Hamachi I: Enhanced Suppression of a Protein-Protein Interaction in Cells Using Small-Molecule Covalent Inhibitors Based on an N-Acyl-N-alkyl Sulfonamide Warhead J Am Chem Soc, 143 (2021), pp. 4766–4774. [DOI] [PubMed] [Google Scholar]
- 59.Trader DJ, Carlson EE: Chemoselective hydroxyl group transformation: an elusive target Mol Biosyst, 8 (2012), pp. 2484–2493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Li BX, Kim DK, Bloom S, Huang RY, Qiao JX, Ewing WR, Oblinsky DG, Scholes GD, MacMillan DWC: Site-selective tyrosine bioconjugation via photoredox catalysis for native-to-bioorthogonal protein transformation Nat Chem, 13 (2021), pp. 902–908. [DOI] [PubMed] [Google Scholar]
- 61.Leier S, Richter S, Bergmann R, Wuest M, Wuest F: Radiometal-Containing Aryl Diazonium Salts for Chemoselective Bioconjugation of Tyrosine Residues ACS Omega, 4 (2019), pp. 22101–22107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Borne AL, Brulet JW, Yuan K, Hsu KL: Development and biological applications of sulfur-triazole exchange (SuTEx) chemistry RSC Chem Biol, 2 (2021), pp. 322–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.*.Hahm HS, Toroitich EK, Borne AL, Brulet JW, Libby AH, Yuan K, Ware TB, McCloud RL, Ciancone AM, Hsu KL: Global targeting of functional tyrosines using sulfur-triazole exchange chemistry Nat Chem Biol, 16 (2020), pp. 150–159. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper provides a novel example of Sulfur(VI)-derived electrophiles using a triazole leaving group to profile tyrosine reactivity
- 64.Brulet JW, Borne AL, Yuan K, Libby AH, Hsu KL: Liganding Functional Tyrosine Sites on Proteins Using Sulfur-Triazole Exchange Chemistry J Am Chem Soc, 142 (2020), pp. 8270–8280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Toroitich EK, Ciancone AM, Hahm HS, Brodowski SM, Libby AH, Hsu KL: Discovery of a Cell-Active SuTEx Ligand of Prostaglandin Reductase 2 Chembiochem, 22 (2021), pp. 2134–2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhang C, Vinogradova EV, Spokoyny AM, Buchwald SL, Pentelute BL: Arylation Chemistry for Bioconjugation Angew Chem Int Ed Engl, 58 (2019), pp. 4810–4839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zambaldo C, Vinogradova EV, Qi X, Iaconelli J, Suciu RM, Koh M, Senkane K, Chadwick SR, Sanchez BB, Chen JS, et al. : 2-Sulfonylpyridines as Tunable, Cysteine-Reactive Electrophiles J Am Chem Soc, 142 (2020), pp. 8972–8979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Crawford LA, Weerapana E: A tyrosine-reactive irreversible inhibitor for glutathione S-transferase Pi (GSTP1) Mol Biosyst, 12 (2016), pp. 1768–1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Shannon DA, Banerjee R, Webster ER, Bak DW, Wang C, Weerapana E: Investigating the proteome reactivity and selectivity of aryl halides J Am Chem Soc, 136 (2014), pp. 3330–3333. [DOI] [PubMed] [Google Scholar]
- 70.Ma N, Hu J, Zhang ZM, Liu W, Huang M, Fan Y, Yin X, Wang J, Ding K, Ye W, et al. : 2H-Azirine-Based Reagents for Chemoselective Bioconjugation at Carboxyl Residues Inside Live Cells J Am Chem Soc, 142 (2020), pp. 6051–6059. [DOI] [PubMed] [Google Scholar]
- 71.O’Brien JGK, Jemas A, Asare-Okai PN, Am Ende CW, Fox JM: Probing the Mechanism of Photoaffinity Labeling by Dialkyldiazirines through Bioorthogonal Capture of Diazoalkanes Org Lett, 22 (2020), pp. 9415–9420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.West AV, Muncipinto G, Wu HY, Huang AC, Labenski MT, Jones LH, Woo CM: Labeling Preferences of Diazirines with Protein Biomolecules J Am Chem Soc, 143 (2021), pp. 6691–6700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bach K, Beerkens BLH, Zanon PRA, Hacker SM: Light-Activatable, 2,5-Disubstituted Tetrazoles for the Proteome-wide Profiling of Aspartates and Glutamates in Living Bacteria ACS Cent Sci, 6 (2020), pp. 546–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Parker CG, Galmozzi A, Wang Y, Correia BE, Sasaki K, Joslyn CM, Kim AS, Cavallaro CL, Lawrence RM, Johnson SR, et al. : Ligand and Target Discovery by Fragment-Based Screening in Human Cells Cell, 168 (2017), pp. 527–541 e529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Cho H, Shen Q, Zhang LH, Okumura M, Kawakami A, Ambrose J, Sigoillot F, Miller HR, Gleim S, Cobos-Correa A, et al. : CYP27A1-dependent anti-melanoma activity of limonoid natural products targets mitochondrial metabolism Cell Chem Biol, 28 (2021), pp. 1407–1419 e1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Eaton JK, Furst L, Ruberto RA, Moosmayer D, Hilpmann A, Ryan MJ, Zimmermann K, Cai LL, Niehues M, Badock V, et al. : Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles Nat Chem Biol, 16 (2020), pp. 497–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Eaton JK, Ruberto RA, Kramm A, Viswanathan VS, Schreiber SL: Diacylfuroxans Are Masked Nitrile Oxides That Inhibit GPX4 Covalently J Am Chem Soc, 141 (2019), pp. 20407–20415. [DOI] [PubMed] [Google Scholar]
- 78.Paxman R, Plate L, Blackwood EA, Glembotski C, Powers ET, Wiseman RL, Kelly JW: Pharmacologic ATF6 activating compounds are metabolically activated to selectively modify endoplasmic reticulum proteins Elife, 7 (2018), pp. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wang Y, Dix MM, Bianco G, Remsberg JR, Lee HY, Kalocsay M, Gygi SP, Forli S, Vite G, Lawrence RM, et al. : Expedited mapping of the ligandable proteome using fully functionalized enantiomeric probe pairs Nat Chem, 11 (2019), pp. 1113–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Li Q: Application of Fragment-Based Drug Discovery to Versatile Targets Front Mol Biosci, 7 (2020), pp. 180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.*.Abbasov ME, Kavanagh ME, Ichu TA, Lazear MR, Tao Y, Crowley VM, Am Ende CW, Hacker SM, Ho J, Dix MM, et al. : A proteome-wide atlas of lysine-reactive chemistry Nat Chem, 13 (2021), pp. 1081–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper profiles proteome reactivity of diverse aminophilic chemotypes and utilizes selected fragments to disrupt various protein activities
- 82.Resnick E, Bradley A, Gan J, Douangamath A, Krojer T, Sethi R, Geurink PP, Aimon A, Amitai G, Bellini D, et al. : Rapid Covalent-Probe Discovery by Electrophile-Fragment Screening J Am Chem Soc, 141 (2019), pp. 8951–8968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Mountford SJ, Anderson BM, Xu B, Tay ESV, Szabo M, Hoang ML, Diao J, Aurelio L, Campden RI, Lindstrom E, et al. : Application of a Sulfoxonium Ylide Electrophile to Generate Cathepsin X-Selective Activity-Based Probes ACS Chem Biol, 15 (2020), pp. 718–727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wang Z, Zhang X, Zhang H, Tang Y, Pan H, Wang H, Ji T, Guo Y, Gao Q, Song T, et al. : Discovery of a Fluorogenic Probe for In Situ Pyruvate Kinase M2 Isoform (PKM2) Labeling through Chemoselective SNAr with a Binding Site Lysine Residue Anal Chem, 93 (2021), pp. 9669–9676. [DOI] [PubMed] [Google Scholar]
- 85.Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, Ferrin TE: UCSF Chimera--a visualization system for exploratory research and analysis J Comput Chem, 25 (2004), pp. 1605–1612. [DOI] [PubMed] [Google Scholar]
