Reversible Covalent Inhibition─Desired Covalent Adduct Formation by Mass ActionClick to copy article linkArticle link copied!
- Disha PatelDisha PatelDepartment of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, CanadaMore by Disha Patel
- Zil E HumaZil E HumaDepartment of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, CanadaMore by Zil E Huma
- Dustin Duncan*Dustin Duncan*Email: [email protected]Department of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, CanadaMore by Dustin Duncan
Abstract
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
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Special Issue
Published as part of ACS Chemical Biology virtual special issue “Exploring Covalent Modulators in Drug Discovery and Chemical Biology”.
Introduction
Design of Noncovalent and Covalent Inhibitors
Incorporating the law of mass action into drug design
Modeling in covalent inhibitor design
Chemical characteristics of reversible covalent warheads
Biochemical analysis of reversible covalent warheads
Development of highly selective JAK3 inhibitors
Development of Bruton Tyrosine Kinase inhibitors for immune diseases
Development of inhibitors for proteases
General considerations for developing reversible covalent inhibitors
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. |
Conclusion and future directions for reversible covalent inhibitors
Acknowledgments
D.D. acknowledges start-up funds provided by Brock University. We thank F. Hammerer and N. Häggman for their discussions and insights.
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
This article references 111 other publications.
- 1Fleming, 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), 780– 790Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3MvpslCisA%253D%253D&md5=3c01ee5f9da7a862f0bd23359d039371On the antibacterial action of cultures of a penicillium, with special reference to their use in the isolation of B. influenzae. 1929Fleming ABulletin of the World Health Organization (2001), 79 (8), 780-90 ISSN:0042-9686.There is no expanded citation for this reference.
- 2Duncan, D.; Auclair, K. Itaconate: An Antimicrobial Metabolite of Macrophages. Can. J. Chem. 2022, 100 (2), 104– 113, DOI: 10.1139/cjc-2021-0117Google ScholarThere is no corresponding record for this reference.
- 3Ray, 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), 1470– 1473, DOI: 10.1021/acschembio.8b00225Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXpsl2nsLc%253D&md5=d2dc8f255fd7e5f7537c57a0c5a3343eThe nitro group as a masked electrophile in covalent enzyme inhibitionRay, Sneha; Kreitler, Dale F.; Gulick, Andrew M.; Murkin, Andrew S.ACS Chemical Biology (2018), 13 (6), 1470-1473CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)We report the unprecedented reaction between a nitroalkane and an active-site cysteine residue to yield a thiohydroximate adduct. Structural and kinetic evidence suggests the nitro group is activated by conversion to its nitronic acid tautomer within the active site. The nitro group, therefore, shows promise as a masked electrophile in the design of covalent inhibitors targeting binding pockets with appropriately placed cysteine and general acid residues.
- 4Yuan, H.; Barnes, K. R.; Weissleder, R.; Cantley, L.; Josephson, L. Covalent Reactions of Wortmannin under Physiological Conditions. Chem. Biol. 2007, 14 (3), 321– 328, DOI: 10.1016/j.chembiol.2007.02.007Google ScholarThere is no corresponding record for this reference.
- 5De Vita, E. 10 Years into the Resurgence of Covalent Drugs. Future Med. Chem. 2021, 13 (2), 193– 210, DOI: 10.4155/fmc-2020-0236Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3szjt1ertQ%253D%253D&md5=71ac37667e2f40fa3f6de36c07fea9ec10 years into the resurgence of covalent drugsDe Vita ElenaFuture medicinal chemistry (2021), 13 (2), 193-210 ISSN:.In the first decade of targeted covalent inhibition, scientists have successfully reversed the previous trend that had impeded the use of covalent inhibition in drug development. Successes in the clinic, mainly in the field of kinase inhibitors, are existing proof that safe covalent inhibitors can be designed and employed to develop effective treatments. The case of KRASG12C covalent inhibitors entering clinical trials in 2019 has been among the hottest topics discussed in drug discovery, raising expectations for the future of the field. In this perspective, an overview of the milestones hit with targeted covalent inhibitors, as well as the promise and the needs of current research, are presented. While recent results have confirmed the potential that was foreseen, many questions remain unexplored in this branch of precision medicine.
- 6Baillie, T. A. Drug-Protein Adducts: Past, Present, and Future. Med. Chem. Res. 2020, 29 (7), 1093– 1104, DOI: 10.1007/s00044-020-02567-8Google ScholarThere is no corresponding record for this reference.
- 7Potashman, M. H.; Duggan, M. E. Covalent Modifiers: An Orthogonal Approach to Drug Design. J. Med. Chem. 2009, 52 (5), 1231– 1246, DOI: 10.1021/jm8008597Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhs1Sltbw%253D&md5=f8380d5a1cdd8638ad2a1e0405d25267Covalent Modifiers: An Orthogonal Approach to Drug DesignPotashman, Michele H.; Duggan, Mark E.Journal of Medicinal Chemistry (2009), 52 (5), 1231-1246CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. In this article we review a variety of examples in which therapeutic targets are covalently bound by small-mol. drugs or by compds. in advanced clin. development. The covalent interactions can be either reversible or irreversible, depending on the reaction partners.
- 8Boike, L.; Henning, N. J.; Nomura, D. K. Advances in Covalent Drug Discovery. Nat. Rev. Drug Discovery 2022, 21 (12), 881– 898, DOI: 10.1038/s41573-022-00542-zGoogle Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xit1Clt7nE&md5=b41441b3a7bc5f2e87bedf243fa43faaAdvances in covalent drug discoveryBoike, Lydia; Henning, Nathaniel J.; Nomura, Daniel K.Nature Reviews Drug Discovery (2022), 21 (12), 881-898CODEN: NRDDAG; ISSN:1474-1776. (Nature Portfolio)A review. Covalent drugs have been used to treat diseases for more than a century, but tools that facilitate the rational design of covalent drugs have emerged more recently. The purposeful addn. of reactive functional groups to existing ligands can enable potent and selective inhibition of target proteins, as demonstrated by the covalent epidermal growth factor receptor (EGFR) and Bruton's tyrosine kinase (BTK) inhibitors used to treat various cancers. Moreover, the identification of covalent ligands through 'electrophile-first' approaches has also led to the discovery of covalent drugs, such as covalent inhibitors for KRAS(G12C) and SARS-CoV-2 main protease. In particular, the discovery of KRAS(G12C) inhibitors validates the use of covalent screening technologies, which have become more powerful and widespread over the past decade. Chemoproteomics platforms have emerged to complement covalent ligand screening and assist in ligand discovery, selectivity profiling and target identification. This Review showcases covalent drug discovery milestones with emphasis on the lessons learned from these programs and how an evolving toolbox of covalent drug discovery techniques facilitates success in this field.
- 9Kenakin, T. The Mass Action Equation in Pharmacology. Br. J. Clin. Pharmacol. 2016, 81 (1), 41– 51, DOI: 10.1111/bcp.12810Google ScholarThere is no corresponding record for this reference.
- 10Pottel, J.; Levit, A.; Korczynska, M.; Fischer, M.; Shoichet, B. K. The Recognition of Unrelated Ligands by Identical Proteins. ACS Chem. Biol. 2018, 13 (9), 2522– 2533, DOI: 10.1021/acschembio.8b00443Google ScholarThere is no corresponding record for this reference.
- 11Knockenhauer, K. E.; Copeland, R. A. The Importance of Binding Kinetics and Drug-Target Residence Time in Pharmacology. Br. J. Pharmacol. 2023, 1– 14, DOI: 10.1111/bph.16104Google ScholarThere is no corresponding record for this reference.
- 12Zhang, G.; Zhang, J.; Gao, Y.; Li, Y.; Li, Y. Strategies for Targeting Undruggable Targets. Expert Opin. Drug Discovery 2022, 17 (1), 55– 69, DOI: 10.1080/17460441.2021.1969359Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2crgvFelug%253D%253D&md5=20ebb3e7bd0c80af87802eea43a05dfaStrategies for targeting undruggable targetsZhang Gong; Zhang Juan; Gao Yuting; Li Yangfeng; Li Yizhou; Li YizhouExpert opinion on drug discovery (2022), 17 (1), 55-69 ISSN:.INTRODUCTION: Undruggable targets refer to clinically meaningful therapeutic targets that are 'difficult to drug' or 'yet to be drugged' via traditional approaches. Featuring characteristics of lacking defined ligand-binding pockets, non-catalytic protein-protein interaction functional modes and less-investigated 3D structures, these undruggable targets have been targeted with novel therapeutic entities developed with the progress of unconventional drug discovery approaches, such as targeted degradation molecules and display technologies. AREA COVERED: This review first presents the concept of 'undruggable' exemplified by RAS and other targets. Next, detailed strategies are illustrated in two aspects: innovation of therapeutic entities and development of unconventional drug discovery technologies. Finally, case studies covering typical undruggable targets (Bcl-2, p53, and RAS) are depicted to further demonstrate the feasibility of the strategies and entities above. EXPERT OPINION: Targeting the undruggable expands the scope of therapeutically reachable targets. Consequently, it represents the drug discovery frontier. Biomedical studies are capable of dissecting disease mechanisms, thus broadening the list of undruggable targets. Encouraged by the recent approval of the KRAS inhibitor Sotorasib, we believe that merging multiple discovery approaches and exploiting various novel therapeutic entities would pave the way for dealing with more 'undruggable' targets in the future.
- 13Akçay, 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), 931– 936, DOI: 10.1038/nchembio.2174Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVKqtb3P&md5=bda5720579a9b5e6027632d2cffba2e1Inhibition of Mcl-1 through covalent modification of a noncatalytic lysine side chainAkcay, Gizem; Belmonte, Matthew A.; Aquila, Brian; Chuaqui, Claudio; Hird, Alexander W.; Lamb, Michelle L.; Rawlins, Philip B.; Su, Nancy; Tentarelli, Sharon; Grimster, Neil P.; Su, QibinNature Chemical Biology (2016), 12 (11), 931-936CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Targeted covalent inhibition of disease-assocd. proteins has become a powerful methodol. in the field of drug discovery, leading to the approval of new therapeutics. Nevertheless, current approaches are often limited owing to their reliance on a cysteine residue to generate the covalent linkage. Here the authors used aryl boronic acid carbonyl warheads to covalently target a noncatalytic lysine side chain, and generated to the knowledge the first reversible covalent inhibitors for Mcl-1, a protein-protein interaction (PPI) target that has proven difficult to inhibit via traditional medicinal chem. strategies. These covalent binders exhibited improved potency in comparison to noncovalent congeners, as demonstrated in biochem. and cell-based assays. The authors identified Lys234 as the residue involved in covalent modification, via point mutation. The covalent binders discovered in this study will serve as useful starting points for the development of Mcl-1 therapeutics and probes to interrogate Mcl-1-dependent biol. phenomena.
- 14Huang, 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/s41392-021-00780-4Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2cfivFanug%253D%253D&md5=05a5959689e63910ecf752595d4407ffKRAS mutation: from undruggable to druggable in cancerHuang Lamei; Guo Zhixing; Wang Fang; Fu LiwuSignal transduction and targeted therapy (2021), 6 (1), 386 ISSN:.Cancer is the leading cause of death worldwide, and its treatment and outcomes have been dramatically revolutionised by targeted therapies. As the most frequently mutated oncogene, Kirsten rat sarcoma viral oncogene homologue (KRAS) has attracted substantial attention. The understanding of KRAS is constantly being updated by numerous studies on KRAS in the initiation and progression of cancer diseases. However, KRAS has been deemed a challenging therapeutic target, even "undruggable", after drug-targeting efforts over the past four decades. Recently, there have been surprising advances in directly targeted drugs for KRAS, especially in KRAS (G12C) inhibitors, such as AMG510 (sotorasib) and MRTX849 (adagrasib), which have obtained encouraging results in clinical trials. Excitingly, AMG510 was the first drug-targeting KRAS (G12C) to be approved for clinical use this year. This review summarises the most recent understanding of fundamental aspects of KRAS, the relationship between the KRAS mutations and tumour immune evasion, and new progress in targeting KRAS, particularly KRAS (G12C). Moreover, the possible mechanisms of resistance to KRAS (G12C) inhibitors and possible combination therapies are summarised, with a view to providing the best regimen for individualised treatment with KRAS (G12C) inhibitors and achieving truly precise treatment.
- 15Mons, 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/cpz1.419Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitF2ksrrF&md5=1ffa766bb1fc413a27c47d458a3f54e7A Comprehensive Guide for Assessing Covalent Inhibition in Enzymatic Assays Illustrated with Kinetic SimulationsMons, Elma; Roet, Sander; Kim, Robbert Q.; Mulder, Monique P. C.Current Protocols (2022), 2 (6), e419CODEN: CPURDB; ISSN:2691-1299. (John Wiley & Sons, Inc.)Covalent inhibition has become more accepted in the past two decades, as illustrated by the clin. approval of several irreversible inhibitors designed to covalently modify their target. Elucidation of the structure-activity relationship and potency of such inhibitors requires a detailed kinetic evaluation. Here, we elucidate the relationship between the exptl. read-out and the underlying inhibitor binding kinetics. Interactive kinetic simulation scripts are employed to highlight the effects of in vitro enzyme activity assay conditions and inhibitor binding mode, thereby showcasing which assumptions and corrections are crucial. Four stepwise protocols to assess the biochem. potency of (ir)reversible covalent enzyme inhibitors targeting a nucleophilic active site residue are included, with accompanying data anal. tailored to the covalent binding mode. Together, this will serve as a guide to make an educated decision regarding the most suitable method to assess covalent inhibition potency.
- 16Chatterjee, 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), 17945– 17952, DOI: 10.1021/jacs.7b08938Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslyrs7bI&md5=e6a38ff570134e956eb43bd4a874ca55Can Relative Binding Free Energy Predict Selectivity of Reversible Covalent Inhibitors?Chatterjee, Payal; Botello-Smith, Wesley M.; Zhang, Han; Qian, Li; Alsamarah, Abdelaziz; Kent, David; Lacroix, Jerome J.; Baudry, Michel; Luo, YunJournal of the American Chemical Society (2017), 139 (49), 17945-17952CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Reversible covalent inhibitors have many clin. advantages over noncovalent or irreversible covalent drugs. However, apart from selecting a warhead, substantial efforts in design and synthesis are needed to optimize noncovalent interactions to improve target-selective binding. Computational prediction of binding affinity for reversible covalent inhibitors presents a unique challenge since the binding process consists of multiple steps, which are not necessarily independent of each other. In this study, the authors lay out the relation between relative binding free energy and the overall reversible covalent binding affinity using a two-state binding model. To prove the concept, the authors employed free energy perturbation (FEP) coupled with λ-exchange mol. dynamics method to calc. the binding free energy of a series of α-ketoamide analogs relative to a common warhead scaffold, in both noncovalent and covalent binding states, and for two highly homologous proteases, calpain-1 and calpain-2. The authors conclude that covalent binding state alone, in general, can be used to predict reversible covalent binding selectivity. However, exceptions may exist. Therefore, the authors also discuss the conditions under which the noncovalent binding step is no longer negligible and propose to combine the relative FEP calcns. with a single QM/MM calcn. of warhead to predict the binding affinity and binding kinetics. The FEP calcns. also revealed that covalent and noncovalent binding states of an inhibitor do not necessarily exhibit the same selectivity. Thus, investigating both binding states, as well as the kinetics will provide extremely useful information for optimizing reversible covalent inhibitors.
- 17De Cesco, S.; Kurian, J.; Dufresne, C.; Mittermaier, A. K.; Moitessier, N. Covalent Inhibitors Design and Discovery. Eur. J. Med. Chem. 2017, 138, 96– 114, DOI: 10.1016/j.ejmech.2017.06.019Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVKhsLrI&md5=551c9a9c4bd69d4ad4d9156c315d5a94Covalent inhibitors design and discoveryDe Cesco, Stephane; Kurian, Jerry; Dufresne, Caroline; Mittermaier, Anthony K.; Moitessier, NicolasEuropean Journal of Medicinal Chemistry (2017), 138 (), 96-114CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)In the history of therapeutics, covalent drugs occupy a very distinct category. While representing a significant fraction of the drugs on the market, very few have been deliberately designed to interact covalently with their biol. target. In this review, the prevalence of covalent drugs will first be briefly covered, followed by an introduction to their mechanisms of action and more detailed discussions of their discovery and the development of safe and efficient covalent enzyme inhibitors. All stages of a drug discovery program will be covered, from target considerations to lead optimization, strategies to tune reactivity and computational methods. The goal of this article is to provide an overview of the field and to outline good practices that are needed for the proper assessment and development of covalent inhibitors as well as a good understanding of the potential and limitations of current computational methods for the design of covalent drugs.
- 18Plescia, 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), 7874– 7884, DOI: 10.1021/acs.jmedchem.9b00642Google ScholarThere is no corresponding record for this reference.
- 19Masuda, 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), 399– 409, DOI: 10.1248/cpb.c17-00854Google ScholarThere is no corresponding record for this reference.
- 20Awoonor-Williams, E.; Walsh, A. G.; Rowley, C. N. Modeling Covalent-Modifier Drugs. Biochim. Biophys. Acta Proteins Proteomics 2017, 1865 (11), 1664– 1675, DOI: 10.1016/j.bbapap.2017.05.009Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXotVOrtbo%253D&md5=ac579900da99ea005d0d562b53a1bca2Modeling covalent-modifier drugsAwoonor-Williams, Ernest; Walsh, Andrew G.; Rowley, Christopher N.Biochimica et Biophysica Acta, Proteins and Proteomics (2017), 1865 (11_Part_B), 1664-1675CODEN: BBAPBW; ISSN:1570-9639. (Elsevier B.V.)A review. In this review, we present a summary of how computer modeling has been used in the development of covalent-modifier drugs. Covalent-modifier drugs bind by forming a chem. bond with their target. This covalent binding can improve the selectivity of the drug for a target with complementary reactivity and result in increased binding affinities due to the strength of the covalent bond formed. In some cases, this results in irreversible inhibition of the target, but some targeted covalent inhibitor (TCI) drugs bind covalently but reversibly. Computer modeling is widely used in drug discovery, but different computational methods must be used to model covalent modifiers because of the chem. bonds formed. Structural and bioinformatic anal. has identified sites of modification that could yield selectivity for a chosen target. Docking methods, which are used to rank binding poses of large sets of inhibitors, have been augmented to support the formation of protein-ligand bonds and are now capable of predicting the binding pose of covalent modifiers accurately. The pKa's of amino acids can be calcd. in order to assess their reactivity towards electrophiles. QM/MM methods have been used to model the reaction mechanisms of covalent modification. The continued development of these tools will allow computation to aid in the development of new covalent-modifier drugs. This article is part of a Special Issue entitled: Biophysics in Canada, edited by Lewis Kay, John Baenziger, Albert Berghuis and Peter Tieleman.
- 21Scarpino, A.; Ferenczy, G. G.; Keserű, G. M. Binding Mode Prediction and Virtual Screening Applications by Covalent Docking. Methods Mol. Biol. Clifton NJ. 2021, 2266, 73– 88, DOI: 10.1007/978-1-0716-1209-5_4Google ScholarThere is no corresponding record for this reference.
- 22Faridoon; 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), 1039– 1062, DOI: 10.1007/s00044-023-03065-3Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXovFKlurc%253D&md5=2f6664586b401207cbe840a4d50cee32An update on the discovery and development of reversible covalent inhibitorsFaridoon; Ng, Raymond; Zhang, Guiping; Li, Jie JackMedicinal Chemistry Research (2023), 32 (6), 1039-1062CODEN: MCREEB; ISSN:1054-2523. (Springer)A review. Small mol. drugs that covalently bind irreversibly to their target proteins have several advantages over conventional reversible inhibitors. They include increased duration of action, less-frequent drug dosing, reduced pharmacokinetic sensitivity, and the potential to target intractable shallow binding sites. Despite these advantages, the key challenges of irreversible covalent drugs are their potential for off-target toxicities and immunogenicity risks. Incorporating reversibility into covalent drugs would lead to less off-target toxicity by forming reversible adducts with off-target proteins and thus reducing the risk of idiosyncratic toxicities caused by the permanent modification of proteins, which leads to higher levels of potential haptens. Herein, we systematically review electrophilic warheads employed during the development of reversible covalent drugs. We hope the structural insights of electrophilic warheads would provide helpful information to medicinal chemists and aid in designing covalent drugs with better on-target selectivity and improved safety.
- 23Martin, J. S.; MacKenzie, C. J.; Fletcher, D.; Gilbert, I. H. Characterising Covalent Warhead Reactivity. Bioorg. Med. Chem. 2019, 27 (10), 2066– 2074, DOI: 10.1016/j.bmc.2019.04.002Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXntVGgsbo%253D&md5=487334d8d0a8b03d0a25d6d87c1b4329Characterising covalent warhead reactivityMartin, James S.; MacKenzie, Claire J.; Fletcher, Daniel; Gilbert, Ian H.Bioorganic & Medicinal Chemistry (2019), 27 (10), 2066-2074CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Many drugs currently used are covalent inhibitors and irreversibly inhibit their targets. Most of these were discovered through serendipity. Covalent inhibitions can have many advantages from a pharmacokinetic perspective. However, until recently most organizations have shied away from covalent compd. design due to fears of non-specific inhibition of off-target proteins leading to toxicity risks. However, there has been a renewed interest in covalent modifiers as potential drugs, as it possible to get highly selective compds. It is therefore important to know how reactive a warhead is and to be able to select the least reactive warhead possible to avoid toxicity. A robust NMR based assay was developed and used to measure the reactivity of a variety of covalent warheads against serine and cysteine - the two most common targets for covalent drugs. A selection of these warheads also had their reactivity measured against threonine, tyrosine, lysine, histidine and arginine to better understand our ability to target non-traditional residues. The reactivity was also measured at various pHs to assess what effect the environment in the active site would have on these reactions. The reactivity of a covalent modifier was found to be very dependent on the amino acid residue.
- 24Péczka, N.; Orgován, Z.; Ábrányi-Balogh, P.; Keserű, G. M. Electrophilic Warheads in Covalent Drug Discovery: An Overview. Expert Opin. Drug Discovery 2022, 17 (4), 413– 422, DOI: 10.1080/17460441.2022.2034783Google ScholarThere is no corresponding record for this reference.
- 25Jackson, 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), 839– 885, DOI: 10.1021/acs.jmedchem.6b00788Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitV2rtbnM&md5=36d3be5bf688c4e4a8cf4bcf42073009Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition ReactionsJackson, Paul A.; Widen, John C.; Harki, Daniel A.; Brummond, Kay M.Journal of Medicinal Chemistry (2017), 60 (3), 839-885CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Although Michael acceptors display a potent and broad spectrum of bioactivity, they have largely been ignored in drug discovery because of their presumed indiscriminate reactivity. As such, a dearth of information exists relevant to the thiol reactivity of natural products and their analogs possessing this moiety. In the midst of recently approved acrylamide-contg. drugs, it is clear that a good understanding of the hetero-Michael addn. reaction and the relative reactivities of biol. thiols with Michael acceptors under physiol. conditions is needed for the design and use of these compds. as biol. tools and potential therapeutics. This Perspective provides information that will contribute to this understanding, such as kinetics of thiol addn. reactions, bioactivities, as well as steric and electronic factors that influence the electrophilicity and reversibility of Michael acceptors. This Perspective is focused on α,β-unsatd. carbonyls given their preponderance in bioactive natural products.
- 26Watt, 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), 8898– 8906, DOI: 10.1039/D2OB01369JGoogle ScholarThere is no corresponding record for this reference.
- 27Watt, 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), 2204– 2212, DOI: 10.1039/D2OB02223KGoogle ScholarThere is no corresponding record for this reference.
- 28Lohman, 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), 14473– 14475, DOI: 10.1021/ja406381bGoogle Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVGjtLzN&md5=f7d0c69687c776310dda4facf967db64Catalysis by desolvation: The catalytic prowess of SAM-dependent halide-alkylating enzymesLohman, Danielle C.; Edwards, David R.; Wolfenden, RichardJournal of the American Chemical Society (2013), 135 (39), 14473-14475CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)In the biol. fixation of halide ions, several enzymes have been found to catalyze alkyl transfer from S-adenosylmethionine (SAM) to halide ions. It proved possible to measure the rates of reaction of the trimethylsulfonium ion with I-, Br-, Cl-, F-, HO-, and H2O in water at elevated temps. Comparison of the resulting 2nd-order rate consts., extrapolated to 25°, with values of kcat/Km reported for fluorinase and chlorinase indicated that these enzymes enhanced the rates of alkyl halide formation by factors of 2 × 1015- and 1 × 1017-fold, resp. These rate enhancements, achieved without the assistance of cofactors, metal ions, or general acid-base catalysis, were the largest that have been reported for an enzyme that acts on 2 substrates.
- 29Parvez, S.; Long, M. J. C.; Poganik, J. R.; Aye, Y. Redox Signaling by Reactive Electrophiles and Oxidants. Chem. Rev. 2018, 118 (18), 8798– 8888, DOI: 10.1021/acs.chemrev.7b00698Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFyjurnI&md5=2cc16400a3dcab3bac28c25132595c7aRedox Signaling by Reactive Electrophiles and OxidantsParvez, Saba; Long, Marcus J. C.; Poganik, Jesse R.; Aye, YimonChemical Reviews (Washington, DC, United States) (2018), 118 (18), 8798-8888CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. The concept of cell signaling in the context of non-enzyme-assisted protein modifications by reactive electrophilic and oxidative species, broadly known as redox signaling, is a uniquely complex topic that has been approached from numerous different and multidisciplinary angles. The authors' review reflects on 5 aspects crit. for understanding how Nature harnesses these non-canonical post-translational modifications to coordinate distinct cellular activities: (1) specific players and their generation; (2) physicochem. properties; (3) mechanisms of action; (4) methods of interrogation; and (5) functional roles in health and disease. Emphasis is primarily placed on the latest progress in the field, but several aspects of classical work likely forgotten/lost are also recollected. For researchers with interests in getting into the field, this review is anticipated to function as a primer. For the expert, the aim is to stimulate thought and discussion about fundamentals of redox signaling mechanisms, and nuances of specificity/selectivity and timing in this sophisticated yet fascinating arena at the crossroads of chem. and biol.
- 30Krishnan, 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), 12624– 12630, DOI: 10.1021/ja505194wGoogle Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVWrtbvK&md5=de052bac07c8920cb807c05f71f4f95dDesign of Reversible, Cysteine-Targeted Michael Acceptors Guided by Kinetic and Computational AnalysisKrishnan, Shyam; Miller, Rand M.; Tian, Boxue; Mullins, R. Dyche; Jacobson, Matthew P.; Taunton, JackJournal of the American Chemical Society (2014), 136 (36), 12624-12630CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Electrophilic probes that covalently modify a cysteine thiol often show enhanced pharmacol. potency and selectivity. Although reversible Michael acceptors have been reported, the structural requirements for reversibility are poorly understood. Here, we report a novel class of acrylonitrile-based Michael acceptors, activated by aryl or heteroaryl electron-withdrawing groups. We demonstrate that thiol adducts of these acrylonitriles undergo β-elimination at rates that span more than 3 orders of magnitude. These rates correlate inversely with the computed proton affinity of the corresponding carbanions, enabling the intrinsic reversibility of the thiol-Michael reaction to be tuned in a predictable manner. We apply these principles to the design of new reversible covalent kinase inhibitors with improved properties. A cocrystal structure of one such inhibitor reveals specific noncovalent interactions between the 1,2,4-triazole activating group and the kinase. Our exptl. and computational study enables the design of new Michael acceptors, expanding the palette of reversible, cysteine-targeted electrophiles.
- 31Ma, 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), 6146– 6162, DOI: 10.1021/acs.jmedchem.9b00387Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFGqsLjI&md5=2de757026f2542bff8092060a72107a5Application of Dually Activated Michael Acceptor to the Rational Design of Reversible Covalent Inhibitor for Enterovirus 71 3C ProteaseMa, Yuying; Li, Linfeng; He, Shuai; Shang, Chengyou; Sun, Yang; Liu, Ning; Meek, Thomas D.; Wang, Yaxin; Shang, LuqingJournal of Medicinal Chemistry (2019), 62 (13), 6146-6162CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Targeted covalent inhibitors (TCIs) have attracted growing attention from the pharmaceutical industry in recent decades because they have potential advantages in terms of efficacy, selectivity, and safety. TCIs have recently evolved into a new version with reversibility that can be systematically modulated. This feature may diminish the risk of haptenization and help optimize the drug-target residence time as needed. The enteroviral 3C protease (3Cpro) is a valuable therapeutic target, but the development of 3Cpro inhibitors is far from satisfactory. Therefore, we aimed to apply a reversible TCI approach to the design of novel 3Cpro inhibitors. The introduction of various substituents onto the α-carbon of classical Michael acceptors yielded inhibitors bearing several classes of warheads. Using steady-state kinetics and biomol. mass spectrometry, we confirmed the mode of reversible covalent inhibition and elucidated the mechanism by which the potency and reversibility were affected by electronic and steric factors. This research produced several potent inhibitors with good selectivity and suitable reversibility; moreover, it validated the reversible TCI approach in the field of viral infection, suggesting broader applications in the design of reversible covalent inhibitors for other proteases.
- 32Bradshaw, 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), 525– 531, DOI: 10.1038/nchembio.1817Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFeju7%252FE&md5=85a73d9ecd62695d03166f7009c68dd8Prolonged and tunable residence time using reversible covalent kinase inhibitorsBradshaw, J. Michael; McFarland, Jesse M.; Paavilainen, Ville O.; Bisconte, Angelina; Tam, Danny; Phan, Vernon T.; Romanov, Sergei; Finkle, David; Shu, Jin; Patel, Vaishali; Ton, Tony; Li, Xiaoyan; Loughhead, David G.; Nunn, Philip A.; Karr, Dane E.; Gerritsen, Mary E.; Funk, Jens Oliver; Owens, Timothy D.; Verner, Erik; Brameld, Ken A.; Hill, Ronald J.; Goldstein, David M.; Taunton, JackNature Chemical Biology (2015), 11 (7), 525-531CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Drugs with prolonged on-target residence times often show superior efficacy, yet general strategies for optimizing drug-target residence time are lacking. Here the authors made progress toward this elusive goal by targeting a noncatalytic cysteine in Bruton's tyrosine kinase (BTK) with reversible covalent inhibitors. Using an inverted orientation of the cysteine-reactive cyanoacrylamide electrophile, the authors identified potent and selective BTK inhibitors that demonstrated biochem. residence times spanning from minutes to 7 d. An inverted cyanoacrylamide with prolonged residence time in vivo remained bound to BTK for more than 18 h after clearance from the circulation. The inverted cyanoacrylamide strategy was further used to discover fibroblast growth factor receptor (FGFR) kinase inhibitors with residence times of several days, demonstrating the generalizability of the approach. Targeting of noncatalytic cysteines with inverted cyanoacrylamides may serve as a broadly applicable platform that facilitates 'residence time by design', the ability to modulate and improve the duration of target engagement in vivo.
- 33Zhou, 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.2022.114571Google ScholarThere is no corresponding record for this reference.
- 34Ehmke, 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), 5764– 5768, DOI: 10.1039/c2ob00034bGoogle Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVSlsr3J&md5=45858b629d24a54b72cd7e3b7ba15e5bTuning and predicting biological affinity: aryl nitriles as cysteine protease inhibitorsEhmke, Veronika; Quinsaat, Jose Enrico Q.; Rivera-Fuentes, Pablo; Heindl, Cornelia; Freymond, Celine; Rottmann, Matthias; Brun, Reto; Schirmeister, Tanja; Diederich, FrancoisOrganic & Biomolecular Chemistry (2012), 10 (30), 5764-5768CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)A series of aryl nitrile-based ligands were prepd. to investigate the effect of their electrophilicity on the affinity against the cysteine proteases rhodesain and human cathepsin L. D. functional theory calcns. provided relative reactivities of the nitriles, enabling prediction of their biol. affinity and cytotoxicity and a clear structure-activity relationship.
- 35Langrish, 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), 1454– 1468, DOI: 10.4049/jimmunol.2001130Google ScholarThere is no corresponding record for this reference.
- 36Jung, 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), 12322– 12358, DOI: 10.1021/acs.jmedchem.1c01002Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslCitbvM&md5=322d56630ffe7e55bfd42ba0f8d62e97Fluorovinylsulfones and -Sulfonates as Potent Covalent Reversible Inhibitors of the Trypanosomal Cysteine Protease Rhodesain: Structure-Activity Relationship, Inhibition Mechanism, Metabolism, and In Vivo StudiesJung, Sascha; Fuchs, Natalie; Johe, Patrick; Wagner, Annika; Diehl, Erika; Yuliani, Tri; Zimmer, Collin; Barthels, Fabian; Zimmermann, Robert A.; Klein, Philipp; Waigel, Waldemar; Meyr, Jessica; Opatz, Till; Tenzer, Stefan; Distler, Ute; Raeder, Hans-Joachim; Kersten, Christian; Engels, Bernd; Hellmich, Ute A.; Klein, Jochen; Schirmeister, TanjaJournal of Medicinal Chemistry (2021), 64 (16), 12322-12358CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Rhodesain is a major cysteine protease of Trypanosoma brucei rhodesiense, a pathogen causing Human African Trypanosomiasis, and a validated drug target. Recently, we reported the development of α-halovinylsulfones as a new class of covalent reversible cysteine protease inhibitors. Here, α-fluorovinylsulfones/-sulfonates were optimized for rhodesain based on mol. modeling approaches. I (X = F), the most potent and selective inhibitor in the series, shows a single-digit nanomolar affinity and high selectivity toward mammalian cathepsins B and L. Enzymic diln. assays and MS expts. indicate that I (X = F) is a slow-tight binder (Ki = 3 nM). Furthermore, the nonfluorinated I (X = H) shows favorable metab. and biodistribution by accumulation in mice brain tissue after i.p. and oral administration. The highest antitrypanosomal activity was obsd. for inhibitors with an N-terminal 2,3-dihydrobenzo[b][1,4]dioxine group and a 4-Me-Phe residue in P2 with nanomolar EC50 values (0.14/0.80μM). The different mechanisms of reversible and irreversible inhibitors were explained using QM/MM calcns. and MD simulations.
- 37Feral, 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, 1775– 1800, DOI: 10.1007/s00726-023-03286-1Google ScholarThere is no corresponding record for this reference.
- 38Fairhurst, 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), 12542– 12573, DOI: 10.1021/acs.jmedchem.0c01019Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVeru77E&md5=94bfa1740e9cac34d0ceac67bc9181a7Discovery of Roblitinib (FGF401) as a Reversible-Covalent Inhibitor of the Kinase Activity of Fibroblast Growth Factor Receptor 4Fairhurst, Robin A.; Knoepfel, Thomas; Buschmann, Nicole; Leblanc, Catherine; Mah, Robert; Todorov, Milen; Nimsgern, Pierre; Ripoche, Sebastien; Niklaus, Michel; Warin, Nicolas; Luu, Van Huy; Madoerin, Mario; Wirth, Jasmin; Graus-Porta, Diana; Weiss, Andreas; Kiffe, Michael; Wartmann, Markus; Kinyamu-Akunda, Jacqueline; Sterker, Dario; Stamm, Christelle; Adler, Flavia; Buhles, Alexandra; Schadt, Heiko; Couttet, Philippe; Blank, Jutta; Galuba, Inga; Trappe, Jorg; Voshol, Johannes; Ostermann, Nils; Zou, Chao; Berghausen, Jorg; Del Rio Espinola, Alberto; Jahnke, Wolfgang; Furet, PascalJournal of Medicinal Chemistry (2020), 63 (21), 12542-12573CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)FGF19 signaling through the FGFR4/β-klotho receptor complex has been shown to be a key driver of growth and survival in a subset of hepatocellular carcinomas, making selective FGFR4 inhibition an attractive treatment opportunity. A kinome-wide sequence alignment highlighted a poorly conserved cysteine residue within the FGFR4 ATP-binding site at position 552, two positions beyond the gate-keeper residue. Several strategies for targeting this cysteine to identify FGFR4 selective inhibitor starting points are summarized which made use of both rational and unbiased screening approaches. The optimization of a 2-formylquinoline amide hit series is described in which the aldehyde makes a hemithioacetal reversible-covalent interaction with cysteine 552. Key challenges addressed during the optimization are improving the FGFR4 potency, metabolic stability, and soly. leading ultimately to the highly selective first-in-class clin. candidate roblitinib.
- 39Shindo, 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), 250– 258, DOI: 10.1038/s41589-018-0204-3Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXlvFSgur4%253D&md5=f6c8886d540ea26a8abecd950feba913Selective and reversible modification of kinase cysteines with chlorofluoroacetamidesShindo, Naoya; Fuchida, Hirokazu; Sato, Mami; Watari, Kosuke; Shibata, Tomohiro; Kuwata, Keiko; Miura, Chizuru; Okamoto, Kei; Hatsuyama, Yuji; Tokunaga, Keisuke; Sakamoto, Seiichi; Morimoto, Satoshi; Abe, Yoshito; Shiroishi, Mitsunori; Caaveiro, Jose M. M.; Ueda, Tadashi; Tamura, Tomonori; Matsunaga, Naoya; Nakao, Takaharu; Koyanagi, Satoru; Ohdo, Shigehiro; Yamaguchi, Yasuchika; Hamachi, Itaru; Ono, Mayumi; Ojida, AkioNature Chemical Biology (2019), 15 (3), 250-258CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Irreversible inhibition of disease-assocd. proteins with small mols. is a powerful approach for achieving increased and sustained pharmacol. potency. Here, we introduce α-chlorofluoroacetamide (CFA) as a novel warhead of targeted covalent inhibitor (TCI). Despite weak intrinsic reactivity, CFA-appended quinazoline showed high reactivity toward Cys797 of epidermal growth factor receptor (EGFR). In cells, CFA-quinazoline showed higher target specificity for EGFR than the corresponding Michael acceptors in a wide concn. range (0.1-10 μM). The cysteine adduct of the CFA deriv. was susceptible to hydrolysis and reversibly yielded intact thiol but was stable in solvent-sequestered ATP-binding pocket of EGFR. This environment-dependent hydrolysis can potentially reduce off-target protein modification by CFA-based drugs. Oral administration of CFA quinazoline, (2R)-1-(2-chloro-2-fluoroacetyl)-N-[4-[(3-chloro-4-fluorophenyl)amino]-7-[3-(4-morpholinyl)-propoxy]quinazolin-6-yl]pyrrolidine-2-carboxamide [2226257-92-5] (NS-062, compd. 51) significantly suppressed tumor growth in a mouse xenograft model. Further, CFA-appended pyrazolopyrimidine irreversibly inhibited Bruton's tyrosine kinase with higher target specificity. These results demonstrate the utility of CFA as a new class warheads for TCI.
- 40Tallon, 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), 16069– 16080, DOI: 10.1021/jacs.3c04444Google ScholarThere is no corresponding record for this reference.
- 41Ingiliz, 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), 1194– 1199, DOI: 10.1111/j.1478-3231.2012.02796.xGoogle ScholarThere is no corresponding record for this reference.
- 42Teicher, B. A.; Tomaszewski, J. E. Proteasome Inhibitors. Biochem. Pharmacol. 2015, 96 (1), 1– 9, DOI: 10.1016/j.bcp.2015.04.008Google ScholarThere is no corresponding record for this reference.
- 43Metcalf, 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), 321– 326, DOI: 10.1021/acsmedchemlett.6b00491Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFWqur0%253D&md5=63766931fdf731c68fd284cf86cc77d0Discovery of GBT440, an Orally Bioavailable R-State Stabilizer of Sickle Cell HemoglobinMetcalf, Brian; Chuang, Chihyuan; Dufu, Kobina; Patel, Mira P.; Silva-Garcia, Abel; Johnson, Carl; Lu, Qing; Partridge, James R.; Patskovska, Larysa; Patskovsky, Yury; Almo, Steven C.; Jacobson, Matthew P.; Hua, Lan; Xu, Qing; Gwaltney, Stephen L.; Yee, Calvin; Harris, Jason; Morgan, Bradley P.; James, Joyce; Xu, Donghong; Hutchaleelaha, Athiwat; Paulvannan, Kumar; Oksenberg, Donna; Li, ZheACS Medicinal Chemistry Letters (2017), 8 (3), 321-326CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)The authors report the discovery of a new potent allosteric effector of sickle cell Hb, GBT440 I, that increases the affinity of Hb for oxygen and consequently inhibits its polymn. when subjected to hypoxic conditions. Unlike earlier allosteric activators that bind covalently to Hb in a 2:1 stoichiometry, I binds with a 1:1 stoichiometry. Compd. I is orally bioavailable and partitions highly and favorably into the red blood cell with a RBC/plasma ratio of ∼150. This partitioning onto the target protein is anticipated to allow therapeutic concns. to be achieved in the red blood cell at low plasma concns. I is in Phase 2 clin. trials for the treatment of sickle cell disease (NCT02285088).
- 44Reja, 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), 1152– 1157, DOI: 10.1021/jacs.1c12702Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xht1egs7s%253D&md5=962dedfe735ef240b3430dfd74cdf5cbLysine-Targeting Reversible Covalent Inhibitors with Long Residence TimeReja, Rahi M.; Wang, Wenjian; Lyu, Yuhan; Haeffner, Fredrik; Gao, JianminJournal of the American Chemical Society (2022), 144 (3), 1152-1157CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We report a new reversible lysine conjugation that features a novel diazaborine product and much slowed dissocn. kinetics in comparison to the previously known iminoboronate chem. Incorporating the diazaborine-forming warhead RMR1 to a peptide ligand gives potent and long-acting reversible covalent inhibitors of the staphylococcal sortase. The efficacy of sortase inhibition is demonstrated via biochem. and cell-based assays. A comparative study of RMR1 and an iminoboronate-forming warhead highlights the significance and potential of modulating bond dissocn. kinetics in achieving long-acting reversible covalent inhibitors.
- 45Serafimova, 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), 471– 476, DOI: 10.1038/nchembio.925Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XkvVKitrw%253D&md5=6455a9a029714beb317286f71ac1326bReversible targeting of noncatalytic cysteines with chemically tuned electrophilesSerafimova, Iana M.; Pufall, Miles A.; Krishnan, Shyam; Duda, Katarzyna; Cohen, Michael S.; Maglathlin, Rebecca L.; McFarland, Jesse M.; Miller, Rand M.; Froedin, Morten; Taunton, JackNature Chemical Biology (2012), 8 (5), 471-476CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Targeting noncatalytic cysteine residues with irreversible acrylamide-based inhibitors is a powerful approach for enhancing pharmacol. potency and selectivity. Nevertheless, concerns about off-target modification motivate the development of reversible cysteine-targeting strategies. Here we show that electron-deficient olefins, including acrylamides, can be tuned to react with cysteine thiols in a rapidly reversible manner. Installation of a nitrile group increased the olefins' intrinsic reactivity, but, paradoxically, eliminated the formation of irreversible adducts. Incorporation of these electrophiles into a noncovalent kinase-recognition scaffold produced slowly dissocg., covalent inhibitors of the p90 ribosomal protein S6 kinase RSK2. A cocrystal structure revealed specific noncovalent interactions that stabilize the complex by positioning the electrophilic carbon near the targeted cysteine. Disruption of these interactions by protein unfolding or proteolysis promoted instantaneous cleavage of the covalent bond. Our results establish a chem.-based framework for engineering sustained covalent inhibition without accumulating permanently modified proteins and peptides.
- 46Forster, 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), 1335– 1340, DOI: 10.1016/j.chembiol.2016.10.008Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVeqsLrJ&md5=348c5b5fbfa91fd02fa21d984620ea16Selective JAK3 Inhibitors with a Covalent Reversible Binding Mode Targeting a New Induced Fit Binding PocketForster, Michael; Chaikuad, Apirat; Bauer, Silke M.; Holstein, Julia; Robers, Matthew B.; Corona, Cesear R.; Gehringer, Matthias; Pfaffenrot, Ellen; Ghoreschi, Kamran; Knapp, Stefan; Laufer, Stefan A.Cell Chemical Biology (2016), 23 (11), 1335-1340CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)Janus kinases (JAKs) are a family of cytoplasmatic tyrosine kinases that are attractive targets for the development of anti-inflammatory drugs given their roles in cytokine signaling. One question regarding JAKs and their inhibitors that remains under intensive debate is whether JAK inhibitors should be isoform selective. Since JAK3 functions are restricted to immune cells, an isoform-selective inhibitor for JAK3 could be esp. valuable to achieve clin. more useful and precise effects. However, the high degree of structural conservation makes isoform-selective targeting a challenging task. Here, we present picomolar inhibitors with unprecedented kinome-wide selectivity for JAK3. Selectivity was achieved by concurrent covalent reversible targeting of a JAK3-specific cysteine residue and a ligand-induced binding pocket. We confirmed that in vitro activity and selectivity translate well into the cellular environment and suggest that our inhibitors are powerful tools to elucidate JAK3-specific functions.
- 47Dietze, 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), 1097– 1103, DOI: 10.1021/tx970090uGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK1c%252FgvFSgtQ%253D%253D&md5=9f71f1d99f6cdbc647091eacb12e6feeInactivation of glyceraldehyde-3-phosphate dehydrogenase by a reactive metabolite of acetaminophen and mass spectral characterization of an arylated active site peptideDietze E C; Schafer A; Omichinski J G; Nelson S DChemical research in toxicology (1997), 10 (10), 1097-103 ISSN:0893-228X.Acetaminophen (4'-hydroxyacetanilide, APAP) is a widely used analgesic and antipyretic drug that can cause hepatic necrosis under some circumstances via cytochrome P450-mediated oxidation to a reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI). Although the mechanism of hepatocellular injury caused by APAP is not fully understood, it is known that NAPQI forms covalent adducts with several hepatocellular proteins. Reported here is the identification of one of these proteins as glyceraldehyde-3-phosphate dehydrogenase [GAPDH, D-glyceraldehyde-3-phosphate: NAD+ oxidoreductase (phosphorylating), EC 1.2.1.12]. Two hours after the administration of hepatotoxic doses of [14C]APAP to mice, at a time prior to overt cell damage, hepatocellular GAPDH activity was significantly decreased concurrent with the formation of a 14C-labeled GAPDH adduct. A nonhepatotoxic regioisomer of APAP, 3'-hydroxyacetanilide (AMAP), was found to decrease GAPDH activity to a lesser extent than APAP, and radiolabel from [14C]AMAP bound to a lesser extent to GAPDH at a time when its overall binding to hepatocellular proteins was almost equivalent to that of APAP. In order to determine the nature of the covalent adduct between GAPDH and APAP, its major reactive and toxic metabolite, NAPQI, was incubated with purified porcine muscle GAPDH. Microsequencing analysis and fast atom bombardment mass spectrometry (FAB-MS) with collision-induced dissociation (CID) were used to characterize one of the adducts as APAP bound to the cysteinyl sulfhydryl group of Cys-149 in the active site peptide of GAPDH.
- 48Rombach, 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), 178– 184, DOI: 10.1021/tx970076hGoogle Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXhslarsLc%253D&md5=1367c196a23344bf587b9d346bc5e7d1Identification of a Rat Liver Microsomal Esterase as a Target Protein for Bromobenzene MetabolitesRombach, Elizabeth M.; Hanzlik, Robert P.Chemical Research in Toxicology (1998), 11 (3), 178-184CODEN: CRTOEC; ISSN:0893-228X. (American Chemical Society)To identify proteins targeted by bromobenzene metabolites, we incubated [14C]bromobenzene in vitro with liver microsomes from phenobarbital-induced rats under conditions which typically led to covalent binding of 2-4 nmol equiv of bromobenzene/mg of protein. Microsomal proteins were solubilized with detergent, sepd. by chromatog. and electrophoresis, and analyzed for 14C by phosphorimaging of stained blots. Much of the radioactivity was assocd. with several bands of proteins of ∼50-60 kDa, plus another prominent band around 70 kDa, but labeling d. appeared to vary considerably overall. A major radiolabeled protein was purified by preparative electrophoresis and submitted to automated Edman microsequencing. Its N-terminal sequence was found to correspond to that of a known rat liver microsomal carboxylesterase (E.C. 3.1.1.1) previously identified as a target for reactive metabolites of halothane. The extent to which covalent modification of this protein by reactive metabolites contributes to the prodn. of hepatotoxic effects remains to be detd.
- 49Tailor, A.; Waddington, J. C.; Meng, X.; Park, B. K. Mass Spectrometric and Functional Aspects of Drug-Protein Conjugation. Chem. Res. Toxicol. 2016, 29 (12), 1912– 1935, DOI: 10.1021/acs.chemrestox.6b00147Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFymu7fI&md5=488bc6cacaf831918db1c28c2f70aa8cMass Spectrometric and Functional Aspects of Drug-Protein ConjugationTailor, Arun; Waddington, James C.; Meng, Xiaoli; Park, B. KevinChemical Research in Toxicology (2016), 29 (12), 1912-1935CODEN: CRTOEC; ISSN:0893-228X. (American Chemical Society)A review. The covalent binding of drugs (metabolites) to proteins to form drug-protein adducts can have an adverse effect on the body. These adducts are thought to be responsible for idiosyncratic drug reactions including severe drug hypersensitivity reactions. Major advances in proteomics technol. have allowed for the identification and quantification of target proteins for certain drugs. Human serum albumin (HSA) and Hb have been identified as accessible targets, and potential biomarkers for drug-protein adducts formation, for numerous drugs (metabolites) including β-lactam antibiotics, reactive drug metabolites such as quinone imines (acetaminophen) and acyl glucuronides (diclofenac), and covalent inhibitors (neratinib). For example, MS/MS anal. of plasma samples from patients taking flucloxacillin revealed that flucloxacillin and its 5-hydroxymethyl metabolite formed covalent adducts with lysine residues on albumin via opening of the β-lactam ring. Other proteins such as P 450 and keratin are also potential targets for covalent binding. However, for most drugs, the properties of these target proteins including their location, their quantity, the timing of conjugate generation, and their biol. function are not well understood. In this review, currently available proteomic technologies including MS/MS anal. to identify antigens, precise location of modifications, and the immunol. consequence of hapten-protein complex are illustrated. Moving forward, identification of the nature of the antigenic determinants that trigger immune responses to drug protein adducts will increase the authors' ability to predict idiosyncratic toxicity for a given compd.
- 50Jeffery, D. A.; Bogyo, M. Chemical Proteomics and Its Application to Drug Discovery. Curr. Opin. Biotechnol. 2003, 14 (1), 87– 95, DOI: 10.1016/S0958-1669(02)00010-1Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXps1WmtQ%253D%253D&md5=9d77283dcea8e41a994dbcd4d14b382bChemical proteomics and its application to drug discoveryJeffery, Douglas A.; Bogyo, MatthewCurrent Opinion in Biotechnology (2003), 14 (1), 87-95CODEN: CUOBE3; ISSN:0958-1669. (Elsevier Science Ltd.)A review with refs. The completion of the human genome sequencing project has provided a flood of new information that is likely to change the way scientists approach the study of complex biol. systems. A major challenge lies in translating this information into new and better ways to treat human disease. The multidisciplinary science of chem. proteomics can be used to distill this flood of new information. This approach makes use of synthetic small mols. that can be used to covalently modify a set of related enzymes and subsequently allow their purifn. and/or identification as valid drug targets. Furthermore, such methods enable rapid biochem. anal. and small-mol. screening of targets thereby accelerating the often difficult process of target validation and drug discovery.
- 51Weerapana, 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), 790– 795, DOI: 10.1038/nature09472Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVGhsr%252FI&md5=277e436e7a8f4dfff0b548bce52ac40dQuantitative reactivity profiling predicts functional cysteines in proteomesWeerapana, Eranthie; Wang, Chu; Simon, Gabriel M.; Richter, Florian; Khare, Sagar; Dillon, Myles B. D.; Bachovchin, Daniel A.; Mowen, Kerri; Baker, David; Cravatt, Benjamin F.Nature (London, United Kingdom) (2010), 468 (7325), 790-795CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Cysteine is the most intrinsically nucleophilic amino acid in proteins, where its reactivity is tuned to perform diverse biochem. functions. The absence of a consensus sequence that defines functional cysteines in proteins has hindered their discovery and characterization. Here we describe a proteomics method to profile quant. the intrinsic reactivity of cysteine residues en masse directly in native biol. systems. Hyper-reactivity was a rare feature among cysteines and it was found to specify a wide range of activities, including nucleophilic and reductive catalysis and sites of oxidative modification. Hyper-reactive cysteines were identified in several proteins of uncharacterized function, including a residue conserved across eukaryotic phylogeny that we show is required for yeast viability and is involved in iron-sulfur protein biogenesis. We also demonstrate that quant. reactivity profiling can form the basis for screening and functional assignment of cysteines in computationally designed proteins, where it discriminated catalytically active from inactive cysteine hydrolase designs.
- 52Backus, 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), 570– 574, DOI: 10.1038/nature18002Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtVSksbnN&md5=6fab087c042de735f8a5931e9f5b946cProteome-wide covalent ligand discovery in native biological systemsBackus, Keriann M.; Correia, Bruno E.; Lum, Kenneth M.; Forli, Stefano; Horning, Benjamin D.; Gonzalez-Paez, Gonzalo E.; Chatterjee, Sandip; Lanning, Bryan R.; Teijaro, John R.; Olson, Arthur J.; Wolan, Dennis W.; Cravatt, Benjamin F.Nature (London, United Kingdom) (2016), 534 (7608), 570-574CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Small mols. are powerful tools for investigating protein function and can serve as leads for new therapeutics. Most human proteins, however, lack small-mol. ligands, and entire protein classes are considered 'undruggable'. Fragment-based ligand discovery can identify small-mol. probes for proteins that have proven difficult to target using high-throughput screening of complex compd. libraries. Although reversibly binding ligands are commonly pursued, covalent fragments provide an alternative route to small-mol. probes, including those that can access regions of proteins that are difficult to target through binding affinity alone. Here we report a quant. anal. of cysteine-reactive small-mol. fragments screened against thousands of proteins in human proteomes and cells. Covalent ligands were identified for >700 cysteines found in both druggable proteins and proteins deficient in chem. probes, including transcription factors, adaptor/scaffolding proteins, and uncharacterized proteins. Among the atypical ligand-protein interactions discovered were compds. that react preferentially with pro- (inactive) caspases. We used these ligands to distinguish extrinsic apoptosis pathways in human cell lines vs. primary human T cells, showing that the former is largely mediated by caspase-8 while the latter depends on both caspase-8 and -10. Fragment-based covalent ligand discovery provides a greatly expanded portrait of the ligandable proteome and furnishes compds. that can illuminate protein functions in native biol. systems.
- 53Abbasov, 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), 1081– 1092, DOI: 10.1038/s41557-021-00765-4Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvF2ju7rJ&md5=90e566b7a65ebfca5108a1c44f06f575A proteome-wide atlas of lysine-reactive chemistryAbbasov, Mikail E.; Kavanagh, Madeline E.; Ichu, Taka-Aki; Lazear, Michael R.; Tao, Yongfeng; Crowley, Vincent M.; am Ende, Christopher W.; Hacker, Stephan M.; Ho, Jordan; Dix, Melissa M.; Suciu, Radu; Hayward, Matthew M.; Kiessling, Laura L.; Cravatt, Benjamin F.Nature Chemistry (2021), 13 (11), 1081-1092CODEN: NCAHBB; ISSN:1755-4330. (Nature Portfolio)Recent advances in chem. proteomics have begun to characterize the reactivity and ligandability of lysines on a global scale. Yet, only a limited diversity of aminophilic electrophiles have been evaluated for interactions with the lysine proteome. Here, we report an in-depth profiling of >30 uncharted aminophilic chemotypes that greatly expands the content of ligandable lysines in human proteins. Aminophilic electrophiles showed disparate proteomic reactivities that range from selective interactions with a handful of lysines to, for a set of dicarboxaldehyde fragments, remarkably broad engagement of the covalent small-mol.-lysine interactions captured by the entire library. We used these latter 'scout' electrophiles to efficiently map ligandable lysines in primary human immune cells under stimulatory conditions. Finally, we show that aminophilic compds. perturb diverse biochem. functions through site-selective modification of lysines in proteins, including protein-RNA interactions implicated in innate immune responses. These findings support the broad potential of covalent chem. for targeting functional lysines in the human proteome.
- 54Hayes, J. D.; Flanagan, J. U.; Jowsey, I. R. Glutathione Transferases. Annu. Rev. Pharmacol. Toxicol. 2005, 45, 51– 88, DOI: 10.1146/annurev.pharmtox.45.120403.095857Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXisVWjtrk%253D&md5=c74f17b1381f046770a7f6faf5c51291Glutathione transferasesHayes, John D.; Flanagan, Jack U.; Jowsey, Ian R.Annual Review of Pharmacology and Toxicology (2005), 45 (), 51-88, 1 plateCODEN: ARPTDI; ISSN:0362-1642. (Annual Reviews Inc.)A review. The authors describe the 3 mammalian glutathione S-transferase (GST) families, namely cytosolic, mitochondrial, and microsomal GSTs, the latter now designated MAPEG (Membrane-Assocd. Proteins in Eicosanoid and Glutathione metab.). In addn. to detoxifying electrophilic xenobiotics, such as chem. carcinogens, environmental pollutants, and antitumor agents, these GSTs inactivate endogenous α,β-unsatd. aldehydes, quinones, epoxides, and hydroperoxides formed as secondary metabolites during oxidative stress. These enzymes are also intimately involved in the biosynthesis of leukotrienes, prostaglandins, testosterone, and progesterone, as well as the degrdn. of tyrosine. Among their substrates, GSTs conjugate the signaling mols., 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) and 4-hydroxynonenal, with glutathione, and consequently they antagonize expression of genes trans-activated by peroxisome proliferator-activated receptor γ (PPARγ) and nuclear factor-erythroid 2 p45-related factor 2 (Nrf2). Through metab. of 15d-PGJ2, GST may enhance gene expression driven by nuclear factor-κB (NF-κB). Cytosolic human GST exhibit genetic polymorphisms and this variation can increase susceptibility to carcinogenesis and inflammatory disease. Polymorphisms in human MAPEG are assocd. with alterations in lung function and increased risk of myocardial infarction and stroke. Targeted disruption of murine genes has demonstrated that cytosolic GST isoenzymes are broadly cytoprotective, whereas MAPEG proteins have pro-inflammatory activities. Furthermore, knockout of mouse GSTA4 and GSTZ1 leads to overexpression of transferases in the Alpha, Mu, and Pi classes, an observation suggesting they are part of an adaptive mechanism that responds to endogenous chem. cues such as 4-hydroxynonenal and tyrosine degrdn. products. Consistent with this hypothesis, the promoters of cytosolic GST and MAPEG genes contain antioxidant response elements through which they are transcriptionally activated during exposure to Michael reaction acceptors and oxidative stress.
- 55Jiang, 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.abd9847Google ScholarThere is no corresponding record for this reference.
- 56Jeffries, 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/ijms23094733Google ScholarThere is no corresponding record for this reference.
- 57Quanrud, 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), 1661– 1676, DOI: 10.1021/acschembio.3c00338Google ScholarThere is no corresponding record for this reference.
- 58Julio, 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 .Google ScholarThere is no corresponding record for this reference.
- 59Adair, 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.202000267Google ScholarThere is no corresponding record for this reference.
- 60Weltzien, H. U.; Padovan, E. Molecular Features of Penicillin Allergy. J. Invest. Dermatol. 1998, 110 (3), 203– 206, DOI: 10.1046/j.1523-1747.1998.00122.xGoogle ScholarThere is no corresponding record for this reference.
- 61Pirmohamed, 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), 236– 244, DOI: 10.1016/j.jaci.2015.06.022Google ScholarThere is no corresponding record for this reference.
- 62Agashe, R. P.; Lippman, S. M.; Kurzrock, R. JAK: Not Just Another Kinase. Mol. Cancer Ther. 2022, 21 (12), 1757– 1764, DOI: 10.1158/1535-7163.MCT-22-0323Google ScholarThere is no corresponding record for this reference.
- 63Goedken, 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), 4573– 4589, DOI: 10.1074/jbc.M114.595181Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXjsF2lsLY%253D&md5=4d99cf5b13fd05df5c4abc197c1fe284Tricyclic Covalent Inhibitors Selectively Target Jak3 through an Active Site ThiolGoedken, Eric R.; Argiriadi, Maria A.; Banach, David L.; Fiamengo, Bryan A.; Foley, Sage E.; Frank, Kristine E.; George, Jonathan S.; Harris, Christopher M.; Hobson, Adrian D.; Ihle, David C.; Marcotte, Douglas; Merta, Philip J.; Michalak, Mark E.; Murdock, Sara E.; Tomlinson, Medha J.; Voss, Jeffrey W.Journal of Biological Chemistry (2015), 290 (8), 4573-4589CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The action of Janus kinases (JAKs) is required for multiple cytokine signaling pathways, and as such, JAK inhibitors hold promise for treatment of autoimmune disorders, including rheumatoid arthritis, inflammatory bowel disease, and psoriasis. However, due to high similarity in the active sites of the four members (Jak1, Jak2, Jak3, and Tyk2), developing selective inhibitors within this family is challenging. We have designed and characterized substituted, tricyclic Jak3 inhibitors that selectively avoid inhibition of the other JAKs. This is accomplished through a covalent interaction between an inhibitor contg. a terminal electrophile and an active site cysteine (Cys-909). We found that these ATP competitive compds. are irreversible inhibitors of Jak3 enzyme activity in vitro. They possess high selectivity against other kinases and can potently (IC50 < 100 nm) inhibit Jak3 activity in cell-based assays. These results suggest irreversible inhibitors of this class may be useful selective agents, both as tools to probe Jak3 biol. and potentially as therapies for autoimmune diseases.
- 64Chen, 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), 225– 242, DOI: 10.1080/13543776.2022.2023129Google ScholarThere is no corresponding record for this reference.
- 65Shindo, 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.2021.116386Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVSrtrjL&md5=d746f5fd8ac5778e1b23e8ef678ceec6Recent progress in covalent warheads for in vivo targeting of endogenous proteinsShindo, Naoya; Ojida, AkioBioorganic & Medicinal Chemistry (2021), 47 (), 116386CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Covalent drugs exert potent and durable activity by chem. modification of the endogenous target protein in vivo. To maximize the pharmacol. efficacy while alleviating the risk of toxicity due to nonspecific off-target reactions, current covalent drug discovery focuses on the development of targeted covalent inhibitors (TCIs), wherein a reactive group (warhead) is strategically incorporated onto a reversible ligand of the target protein to facilitate specific covalent engagement. Various aspects of warheads, such as intrinsic reactivity, chemoselectivity, mode of reaction, and reversibility of the covalent engagement, would affect the target selectivity of TCIs. Although TCIs clin. approved to date largely rely on Michael acceptor-type electrophiles for cysteine targeting, a wide array of novel warheads have been devised and tested in TCI development in recent years. In this short review, we provide an overview of recent progress in chem. for selective covalent targeting of proteins and their applications in TCI designs.
- 66Forster, 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), 5350– 5366, DOI: 10.1021/acs.jmedchem.8b00571Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVGjsrbN&md5=8acdec137df746f854bbee75a10d5f70Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4-d]pyrrolo[2,3-b]pyridine ScaffoldForster, Michael; Chaikuad, Apirat; Dimitrov, Teodor; Doering, Eva; Holstein, Julia; Berger, Benedict-Tilman; Gehringer, Matthias; Ghoreschi, Kamran; Mueller, Susanne; Knapp, Stefan; Laufer, Stefan A.Journal of Medicinal Chemistry (2018), 61 (12), 5350-5366CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Janus kinases are major drivers of immune signaling and have been the focus of anti-inflammatory drug discovery for more than a decade. Because of the invariable colocalization of JAK1 and JAK3 at cytokine receptors, the question if selective JAK3 inhibition is sufficient to effectively block downstream signaling has been highly controversial. Recently, we discovered the covalent-reversible JAK3 inhibitor FM-381 (23) featuring high isoform and kinome selectivity. Crystallog. revealed that this inhibitor induces an unprecedented binding pocket by interactions of a nitrile substituent with arginine residues in JAK3. Herein, we describe detailed structure-activity relationships necessary for induction of the arginine pocket and the impact of this structural change on potency, isoform selectivity, and efficacy in cellular models. Furthermore, we evaluated the stability of this novel inhibitor class in in vitro metabolic assays and were able to demonstrate an adequate stability of key compd. 23 for in vivo use.
- 67Laux, 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), 573– 602, DOI: 10.1021/acsptsci.2c00054Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xhsl2nur7M&md5=31a71f1a28ac2893f116b25e3972fc36Pharmacokinetic Optimization of Small Molecule Janus Kinase 3 Inhibitors to Target Immune CellsLaux, Julian; Forster, Michael; Riexinger, Laura; Schwamborn, Anna; Guezguez, Jamil; Pokoj, Christina; Kudolo, Mark; Berger, Lena M.; Knapp, Stefan; Schollmeyer, Dieter; Guse, Jan; Burnet, Michael; Laufer, Stefan A.ACS Pharmacology & Translational Science (2022), 5 (8), 573-602CODEN: APTSFN; ISSN:2575-9108. (American Chemical Society)Modulation of Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling is a promising method of treating autoimmune diseases, and the profound potency of clin. compds. makes this mode of action particularly attractive. Other questions that remain unanswered also include: What is the ideal selectivity between JAK1 and JAK3. Which cells are most relevant to JAK blockade. And what is the ideal tissue distribution pattern for addressing specific autoimmune conditions. We hypothesized that JAK3 selectivity is most relevant to low-dose clin. effects and interleukin-10 (IL-10) stimulation in particular, that immune cells are the most important compartment, and that distribution to inflamed tissue is the most important pharmacokinetic characteristic for in vivo disease modification. To test these hypotheses, we prepd. modified derivs. of JAK3 specific inhibitors that target C909 near the ATP binding site based on FM-381, first reported in 2016; a compd. class that was hitherto limited in uptake and exposure in vivo. These limits appear to be due to metabolic instability of side groups binding in the selectivity pocket. We identified derivs. with improved stability and tissue exposure. Conjugation to macrolide scaffolds with medium chain linkers was sufficient to stabilize the compds. and improve transport to organs while maintaining JAK3 affinity. These conjugates are inflammation targeted JAK3 inhibitors with long tissue half-lives and high exposure to activated immune cells.
- 68Burger, J. A. Bruton Tyrosine Kinase Inhibitors: Present and Future. Cancer J. 2019, 25 (6), 386– 393, DOI: 10.1097/PPO.0000000000000412Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlOms7zM&md5=574551611f4a8f843cf1ce2cd0db1697Bruton Tyrosine Kinase Inhibitors: Present and FutureBurger, Jan A.Cancer Journal (Philadelphia, PA, United States) (2019), 25 (6), 386-393CODEN: CAJOCB; ISSN:1528-9117. (Lippincott Williams & Wilkins)A review. Bruton tyrosine kinase (BTK) is a nonreceptor tyrosine kinase that plays a central role in the signal transduction of the B-cell antigen receptor and other cell surface receptors, both in normal and malignant B lymphocytes. B-cell antigen receptor signaling is activated in secondary lymphatic organs and drives the proliferation of malignant B cells, including chronic lymphocytic leukemia (CLL) cells. During the last 10 years, BTK inhibitors (BTKis) are increasingly replacing chemotherapy-based regimen, esp. in patients with CLL and mantle cell lymphoma (MCL). Bruton tyrosine kinase inhibitors are particularly active in patients with CLL and MCL, but also received approval for Waldenstroem macroglobulinemia, small lymphocytic lymphoma, marginal zone lymphoma, and chronic graft-vs.-host disease. Current clin. practice is continuous long-term administration of BTKi, which can be complicated by adverse effects or the development of drug resistance. Alternatives to long-term use of BTKi are being developed, such as combination therapies, permitting for limited duration therapy. Second-generation BTKis are under development, which differ from ibrutinib, the first-in-class BTKi, in their specificity for BTK, and therefore may differentiate themselves from ibrutinib in terms of adverse effects or efficacy.
- 69Khan, W. N. Regulation of B Lymphocyte Development and Activation by Bruton’s Tyrosine Kinase. Immunol. Res. 2001, 23 (2–3), 147– 156, DOI: 10.1385/IR:23:2-3:147Google ScholarThere is no corresponding record for this reference.
- 70Guldenpfennig, C.; Teixeiro, E.; Daniels, M. NF-kB’s Contribution to B Cell Fate Decisions. Front. Immunol. 2023, 14, 1214095, DOI: 10.3389/fimmu.2023.1214095Google ScholarThere is no corresponding record for this reference.
- 71Staudt, L. M. Oncogenic Activation of NF-kappaB. Cold Spring Harb. Perspect. Biol. 2010, 2 (6), a000109, DOI: 10.1101/cshperspect.a000109Google ScholarThere is no corresponding record for this reference.
- 72Small 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 .Google ScholarThere is no corresponding record for this reference.
- 73Wu, 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/s13045-016-0250-9Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXis1eisrw%253D&md5=a6a8f9b9236abe7ef1054a41e1c71e5aAcalabrutinib (ACP-196): a selective second-generation BTK inhibitorWu, Jingjing; Zhang, Mingzhi; Liu, DelongJournal of Hematology & Oncology (2016), 9 (), 21/1-21/4CODEN: JHOOAO; ISSN:1756-8722. (BioMed Central Ltd.)More and more targeted agents become available for B cell malignancies with increasing precision and potency. The first-in-class Bruton's tyrosine kinase (BTK) inhibitor, ibrutinib, has been in clin. use for the treatment of chronic lymphocytic leukemia, mantle cell lymphoma, and Waldenstrom's macroglobulinemia. More selective BTK inhibitors (ACP-196, ONO/GS-4059, BGB-3111, CC-292) are being explored. Acalabrutinib (ACP-196) is a novel irreversible second-generation BTK inhibitor that was shown to be more potent and selective than ibrutinib. This review summarized the preclin. research and clin. data of acalabrutinib.
- 74Guo, 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), 7923– 7940, DOI: 10.1021/acs.jmedchem.9b00687Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsFSku7bP&md5=5cdb551d296e60eb1e82b03b7a0384eeDiscovery of Zanubrutinib (BGB-3111), a Novel, Potent, and Selective Covalent Inhibitor of Bruton's Tyrosine KinaseGuo, Yunhang; Liu, Ye; Hu, Nan; Yu, Desheng; Zhou, Changyou; Shi, Gongyin; Zhang, Bo; Wei, Min; Liu, Junhua; Luo, Lusong; Tang, Zhiyu; Song, Huipeng; Guo, Yin; Liu, Xuesong; Su, Dan; Zhang, Shuo; Song, Xiaomin; Zhou, Xing; Hong, Yuan; Chen, Shuaishuai; Cheng, Zhenzhen; Young, Steve; Wei, Qiang; Wang, Haisheng; Wang, Qiuwen; Lv, Lei; Wang, Fan; Xu, Haipeng; Sun, Hanzi; Xing, Haimei; Li, Na; Zhang, Wei; Wang, Zhongbo; Liu, Guodong; Sun, Zhijian; Zhou, Dongping; Li, Wei; Liu, Libin; Wang, Lai; Wang, ZhiweiJournal of Medicinal Chemistry (2019), 62 (17), 7923-7940CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Aberrant activation of Bruton's tyrosine kinase (BTK) plays an important role in pathogenesis of B-cell lymphomas, suggesting that inhibition of BTK is useful in the treatment of hematol. malignancies. The discovery of a more selective on-target covalent BTK inhibitor is of high value. Herein, we disclose the discovery and preclin. characterization of a potent, selective, and irreversible BTK inhibitor as our clin. candidate by using in vitro potency, selectivity, pharmacokinetics (PK), and in vivo pharmacodynamic for prioritizing compds. Compd. BGB-3111 (31a, Zanubrutinib) demonstrates (i) potent activity against BTK and excellent selectivity over other TEC, EGFR and Src family kinases, (ii) desirable ADME, excellent in vivo pharmacodynamic in mice and efficacy in OCI-LY10 xenograft models.
- 75Liclican, 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.2020.129531Google ScholarThere is no corresponding record for this reference.
- 76Tam, 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-xGoogle ScholarThere is no corresponding record for this reference.
- 77Estupiñán, 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.2021.630942Google Scholar77https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3sfjtlCrtg%253D%253D&md5=c559e266e46358b280c7835a9574d965Comparative Analysis of BTK Inhibitors and Mechanisms Underlying Adverse EffectsEstupinan H Yesid; Berglof Anna; Zain Rula; Smith C I Edvard; Estupinan H Yesid; Zain RulaFrontiers in cell and developmental biology (2021), 9 (), 630942 ISSN:2296-634X.The cytoplasmic protein-tyrosine kinase BTK plays an essential role for differentiation and survival of B-lineage cells and, hence, represents a suitable drug target. The number of BTK inhibitors (BTKis) in the clinic has increased considerably and currently amounts to at least 22. First-in-class was ibrutinib, an irreversible binder forming a covalent bond to a cysteine in the catalytic region of the kinase, for which we have identified 228 active trials listed at ClinicalTrials.gov. Next-generation inhibitors, acalabrutinib and zanubrutinib, are approved both in the United States and in Europe, and zanubrutinib also in China, while tirabrutinib is currently only registered in Japan. In most cases, these compounds have been used for the treatment of B-lymphocyte tumors. However, an increasing number of trials instead addresses autoimmunity and inflammation in multiple sclerosis, rheumatoid arthritis, pemphigus and systemic lupus erythematosus with the use of either irreversibly binding inhibitors, e.g., evobrutinib and tolebrutinib, or reversibly binding inhibitors, like fenebrutinib. Adverse effects (AEs) have predominantly implicated inhibition of other kinases with a BTKi-binding cysteine in their catalytic domain. Analysis of the reported AEs suggests that ibrutinib-associated atrial fibrillation is caused by binding to ERBB2/HER2 and ERBB4/HER4. However, the binding pattern of BTKis to various additional kinases does not correlate with the common assumption that skin manifestations and diarrhoeas are off-target effects related to EGF receptor inhibition. Moreover, dermatological toxicities, diarrhoea, bleedings and invasive fungal infections often develop early after BTKi treatment initiation and subsequently subside. Conversely, cardiovascular AEs, like hypertension and various forms of heart disease, often persist.
- 78Mato, 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), 874– 879, DOI: 10.3324/haematol.2017.182907Google ScholarThere is no corresponding record for this reference.
- 79Sharman, 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_1.4060.4060Google ScholarThere is no corresponding record for this reference.
- 80Winqvist, 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), e208– e210, DOI: 10.3324/haematol.2018.198820Google ScholarThere is no corresponding record for this reference.
- 81ClinicalTrials.gov. Rilzabrutinib. https://clinicaltrials.gov/search?intr=Rilzabrutinib (accessed 2023–11–28).Google ScholarThere is no corresponding record for this reference.
- 82Oda, K. New Families of Carboxyl Peptidases: Serine-Carboxyl Peptidases and Glutamic Peptidases. J. Biochem. (Tokyo) 2012, 151 (1), 13– 25, DOI: 10.1093/jb/mvr129Google ScholarThere is no corresponding record for this reference.
- 83Ćwilichowska, 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.2022.101144Google ScholarThere is no corresponding record for this reference.
- 84Manasanch, E. E.; Orlowski, R. Z. Proteasome Inhibitors in Cancer Therapy. Nat. Rev. Clin. Oncol. 2017, 14 (7), 417– 433, DOI: 10.1038/nrclinonc.2016.206Google Scholar84https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFGlsbg%253D&md5=7246069f357a4cc9449f822c57767811Proteasome inhibitors in cancer therapyManasanch, Elisabet E.; Orlowski, Robert Z.Nature Reviews Clinical Oncology (2017), 14 (7), 417-433CODEN: NRCOAA; ISSN:1759-4774. (Nature Publishing Group)The ubiquitin proteasome pathway was discovered in the 1980s to be a central component of the cellular protein-degrdn. machinery with essential functions in homeostasis, which include preventing the accumulation of misfolded or deleterious proteins. Cancer cells produce proteins that promote both cell survival and proliferation, and/or inhibit mechanisms of cell death. This notion set the stage for preclin. testing of proteasome inhibitors as a means to shift this fine equil. towards cell death. Since the late 1990s, clin. trials have been conducted for a variety of malignancies, leading to regulatory approvals of proteasome inhibitors to treat multiple myeloma and mantle-cell lymphoma. First-generation and second-generation proteasome inhibitors can elicit deep initial responses in patients with myeloma, for whom these drugs have dramatically improved outcomes, but relapses are frequent and acquired resistance to treatment eventually emerges. In addn., promising preclin. data obtained with proteasome inhibitors in models of solid tumors have not been confirmed in the clinic, indicating the importance of primary resistance. Investigation of the mechanisms of resistance is, therefore, essential to further maximize the utility of this class of drugs in the era of personalized medicine. Herein, we discuss the advances and challenges resulting from the introduction of proteasome inhibitors into the clinic.
- 85Flint, 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), 401– 411, DOI: 10.1128/AAC.01081-08Google ScholarThere is no corresponding record for this reference.
- 86Augeri, 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), 5025– 5037, DOI: 10.1021/jm050261pGoogle Scholar86https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXlsVeltb0%253D&md5=878355880133bb0ddd6bd14ecbf1d070Discovery and Preclinical Profile of Saxagliptin (BMS-477118): A Highly Potent, Long-Acting, Orally Active Dipeptidyl Peptidase IV Inhibitor for the Treatment of Type 2 DiabetesAugeri, David J.; Robl, Jeffrey A.; Betebenner, David A.; Magnin, David R.; Khanna, Ashish; Robertson, James G.; Wang, Aiying; Simpkins, Ligaya M.; Taunk, Prakash; Huang, Qi; Han, Song-Ping; Abboa-Offei, Benoni; Cap, Michael; Xin, Li; Tao, Li; Tozzo, Effie; Welzel, Gustav E.; Egan, Donald M.; Marcinkeviciene, Jovita; Chang, Shu Y.; Biller, Scott A.; Kirby, Mark S.; Parker, Rex A.; Hamann, Lawrence G.Journal of Medicinal Chemistry (2005), 48 (15), 5025-5037CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Efforts to further elucidate structure-activity relationships (SAR) within the authors previously disclosed series of β-quaternary amino acid linked L-cis-4,5-methanoprolinenitrile dipeptidyl peptidase IV (DPP-IV) inhibitors led to the investigation of vinyl substitution at the β-position of α-cycloalkyl-substituted glycines. Despite poor systemic exposure, vinyl-substituted compds. showed extended duration of action in acute rat ex vivo plasma DPP-IV inhibition models. Oxygenated putative metabolites were prepd. and were shown to exhibit the potency and extended duration of action of their precursors in efficacy models measuring glucose clearance in Zuckerfa/fa rats. Extension of this approach to adamantylglycine-derived inhibitors led to the discovery of highly potent inhibitors, including hydroxyadamantyl compd. BMS-477118 (saxagliptin), a highly efficacious, stable, and long-acting DPP-IV inhibitor, which is currently undergoing clin. trials for treatment of type 2 diabetes.
- 87Lin, 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), 1813– 1822, DOI: 10.1128/AAC.50.5.1813-1822.2006Google ScholarThere is no corresponding record for this reference.
- 88Focosi, 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.2022.106708Google ScholarThere is no corresponding record for this reference.
- 89Wen, W.; Qi, Z.; Wang, J. The Function and Mechanism of Enterovirus 71 (EV71) 3C Protease. Curr. Microbiol. 2020, 77 (9), 1968– 1975, DOI: 10.1007/s00284-020-02082-4Google ScholarThere is no corresponding record for this reference.
- 90Zeng, 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), 1762– 1766, DOI: 10.1016/j.bmcl.2016.02.039Google ScholarThere is no corresponding record for this reference.
- 91Zhai, 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, 559– 573, DOI: 10.1016/j.ejmech.2016.08.064Google Scholar91https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsV2jurnN&md5=41cabd5d56ab0164e48cbd7806dbf2c7Structure-activity relationship study of peptidomimetic aldehydes as enterovirus 71 3C protease inhibitorsZhai, Yangyang; Ma, Yuying; Ma, Fei; Nie, Quandeng; Ren, Xuejiao; Wang, Yaxin; Shang, Luqing; Yin, ZhengEuropean Journal of Medicinal Chemistry (2016), 124 (), 559-573CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A series of peptidomimetic aldehydes were designed, synthesized, and evaluated for their biochem. activity against 3C protease (3Cpro) and anti-enterovirus 71 (EV71) activity in vitro. Mol. docking revealed that 5s (IC50 = 0.22 ± 0.07 μM, EC50 = 0.18 ± 0.05 μM) could bind well to the active site of EV71 3Cpro, which was consistent with the biol. data compared to ref. 5a (IC50 = 0.54 ± 0.02 μM, EC50 = 0.26 ± 0.07 μM). Structure and relationship study led to the discovery of aldehyde 5x (IC50 = 0.10 ± 0.02 μM, EC50 = 0.11 ± 0.07 μM), which exhibited the most potent 3Cpro inhibitory and antiviral activity.
- 92Luo, Y. L. Mechanism-Based and Computational-Driven Covalent Drug Design. J. Chem. Inf. Model. 2021, 61 (11), 5307– 5311, DOI: 10.1021/acs.jcim.1c01278Google ScholarThere is no corresponding record for this reference.
- 93Mihalovits, 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), 6579– 6594, DOI: 10.1021/acs.jcim.0c00834Google Scholar93https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFSqu73N&md5=61ff455099a14db29ffd081cd3fd613eAffinity and Selectivity Assessment of Covalent Inhibitors by Free Energy CalculationsMihalovits, Levente M.; Ferenczy, Gyorgy G.; Keseru, Gyorgy M.Journal of Chemical Information and Modeling (2020), 60 (12), 6579-6594CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Covalent inhibitors have been gaining increased attention in drug discovery due to their beneficial properties such as long residence time, high biochem. efficiency, and specificity. Optimization of covalent inhibitors is a complex task that involves parallel monitoring of the noncovalent recognition elements and the covalent reactivity of the mols. to avoid potential idiosyncratic side effects. This challenge calls for special design protocols, including a variety of computational chem. methods. Covalent inhibition proceeds through multiple steps, and calcg. free energy changes of the subsequent binding events along the overall binding process would help us to better control the design of drug candidates. Inspired by the recent success of free energy calcns. on reversible binders, we developed a complex protocol to compute free energies related to the noncovalent and covalent binding steps with thermodn. integration and hybrid quantum mech./mol. mech. (QM/MM) potential of mean force (PMF) calcns., resp. In optimization settings, we examd. two therapeutically relevant proteins complexed with congeneric sets of irreversible cysteine targeting covalent inhibitors. In the selectivity paradigm, we studied the irreversible binding of covalent inhibitors to phylogenetically close targets by a mutational approach. The results of the calcns. are in good agreement with the exptl. free energy values derived from the inhibition and kinetic consts. (Ki and kinact) of the enzyme-inhibitor binding. The proposed method might be a powerful tool to predict the potency, selectivity, and binding mechanism of irreversible covalent inhibitors.
- 94Mihalovits, 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.26768Google ScholarThere is no corresponding record for this reference.
- 95Lonsdale, 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), 3124– 3137, DOI: 10.1021/acs.jcim.7b00553Google Scholar95https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsl2gsLjF&md5=722e83699c58dee1009b87c8c31bdeccExpanding the Armory: Predicting and Tuning Covalent Warhead ReactivityLonsdale, Richard; Burgess, Jonathan; Colclough, Nicola; Davies, Nichola L.; Lenz, Eva M.; Orton, Alexandra L.; Ward, Richard A.Journal of Chemical Information and Modeling (2017), 57 (12), 3124-3137CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Targeted covalent inhibition is an established approach for increasing the potency and selectivity of potential drug candidates, as well as identifying potent and selective tool compds. for target validation studies. It is evident that identification of reversible recognition elements is essential for selective covalent inhibition, but this must also be achieved with the appropriate level of inherent reactivity of the reactive functionality (or "warhead"). Structural changes that increase or decrease warhead reactivity, guided by methods to predict the effect of those changes, have the potential to tune warhead reactivity and negate issues related to potency and/or toxicity. The half-life to adduct formation with glutathione (GSH t1/2) is a useful assay for measuring the reactivity of cysteine-targeting covalent warheads but is limited to synthesized mols. In this manuscript the authors assess the ability of several exptl. and computational approaches to predict GSH t1/2 for a range of cysteine targeting warheads, including a novel method based on pKa. Furthermore, matched mol. pairs anal. has been performed against the internal compd. collection, revealing structure-activity relationships between a selection of different covalent warheads. These observations and methods of prediction will be valuable in the design of new covalent inhibitors with desired levels of reactivity.
- 96Oballa, 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), 998– 1002, DOI: 10.1016/j.bmcl.2006.11.044Google Scholar96https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht12qtr4%253D&md5=b8fff00bdd6a746753f8f244d25b0cc8A generally applicable method for assessing the electrophilicity and reactivity of diverse nitrile-containing compoundsOballa, Renata M.; Truchon, Jean-Francois; Bayly, Christopher I.; Chauret, Nathalie; Day, Stephen; Crane, Sheldon; Berthelette, CarlBioorganic & Medicinal Chemistry Letters (2007), 17 (4), 998-1002CODEN: BMCLE8; ISSN:0960-894X. (Elsevier Ltd.)Nitrile-based inhibitors of cathepsin K have been known for some time and mechanism-of-action studies have demonstrated that cysteinyl proteases interact with nitriles in a reversible fashion. Three main classes of nitrile-contg. inhibitors have been published in the cathepsin K field: (i) cyanamides, (ii) arom. nitriles, and (iii) aminoacetonitriles. A computational approach was used to calc. the theor. reactivities of diverse nitriles and this was found to correlate with their extent of reactivity with free cysteine. Moreover, there is a tentative link between high reactivity with cysteine and the potential to lead to irreversible covalent binding to proteins.
- 97Copeland, 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), 730– 739, DOI: 10.1038/nrd2082Google Scholar97https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XptVCltro%253D&md5=60ede2301584b10ac4e8fa18e1e6d107Drug-target residence time and its implications for lead optimizationCopeland, Robert A.; Pompliano, David L.; Meek, Thomas D.Nature Reviews Drug Discovery (2006), 5 (9), 730-739CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Much of drug discovery today is predicated on the concept of selective targeting of particular bioactive macromols. by low-mol.-mass drugs. The binding of drugs to their macromol. targets is therefore seen as paramount for pharmacol. activity. In vitro assessment of drug-target interactions is classically quantified in terms of binding parameters such as IC50 or Kd. This article presents an alternative perspective on drug optimization in terms of drug-target binary complex residence time, as quantified by the dissociative half-life of the drug-target binary complex. We describe the potential advantages of long residence time in terms of duration of pharmacol. effect and target selectivity.
- 98Bernetti, M.; Masetti, M.; Rocchia, W.; Cavalli, A. Kinetics of Drug Binding and Residence Time. Annu. Rev. Phys. Chem. 2019, 70, 143– 171, DOI: 10.1146/annurev-physchem-042018-052340Google Scholar98https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjsFSgsro%253D&md5=6016da9b20e539f43e627432018ae51bKinetics of Drug Binding and Residence TimeBernetti, Mattia; Masetti, Matteo; Rocchia, Walter; Cavalli, AndreaAnnual Review of Physical Chemistry (2019), 70 (), 143-171CODEN: ARPLAP; ISSN:0066-426X. (Annual Reviews)The kinetics of drug binding and unbinding is assuming an increasingly crucial role in the long, costly process of bringing a new medicine to patients. For example, the time a drug spends in contact with its biol. target is known as residence time (the inverse of the kinetic const. of the drug-target unbinding, 1/koff). Recent reports suggest that residence time could predict drug efficacy in vivo, perhaps even more effectively than conventional thermodn. parameters (free energy, enthalpy, entropy). There are many exptl. and computational methods for predicting drug-target residence time at an early stage of drug discovery programs. Here, we review and discuss the methodol. approaches to estg. drug binding kinetics and residence time. We first introduce the theor. background of drug binding kinetics from a physicochem. standpoint. We then analyze the recent literature in the field, starting from the exptl. methodologies and applications thereof and moving to theor. and computational approaches to the kinetics of drug binding and unbinding. We acknowledge the central role of mol. dynamics and related methods, which comprise a great no. of the computational methods and applications reviewed here. However, we also consider kinetic Monte Carlo. We conclude with the outlook that drug (un)binding kinetics may soon become a go/no go step in the discovery and development of new medicines.
- 99Ren, 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), 493– 510, DOI: 10.1007/s10928-022-09822-yGoogle ScholarThere is no corresponding record for this reference.
- 100Frühauf, A.; Wolff, B.; Schweipert, M.; Meyer-Almes, F.-J. Synthesis and Characterization of Reversible Covalent HDAC4 Inhibitors. Methods Mol. Biol. Clifton NJ. 2023, 2589, 207– 221, DOI: 10.1007/978-1-0716-2788-4_14Google ScholarThere is no corresponding record for this reference.
- 101Forman, H. J.; Zhang, H.; Rinna, A. Glutathione: Overview of Its Protective Roles, Measurement, and Biosynthesis. Mol. Aspects Med. 2009, 30 (1–2), 1– 12, DOI: 10.1016/j.mam.2008.08.006Google Scholar101https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXltFelurY%253D&md5=c1b1aa2b03389bf1434af0762df78fe1Glutathione: Overview of its protective roles, measurement, and biosynthesisForman, Henry Jay; Zhang, Hongqiao; Rinna, AlessandraMolecular Aspects of Medicine (2009), 30 (1-2), 1-12CODEN: MAMED5; ISSN:0098-2997. (Elsevier B.V.)This review is the introduction to a special issue concerning, glutathione (GSH), the most abundant low mol. wt. thiol compd. synthesized in cells. GSH plays crit. roles in protecting cells from oxidative damage and the toxicity of xenobiotic electrophiles, and maintaining redox homeostasis. Here, the functions and GSH and the sources of oxidants and electrophiles, the elimination of oxidants by redn. and electrophiles by conjugation with GSH are briefly described. Methods of assessing GSH status in the cells are also described. GSH synthesis and its regulation are addressed along with therapeutic approaches for manipulating GSH content that have been proposed. The purpose here is to provide a brief overview of some of the important aspects of glutathione metab. as part of this special issue that will provide a more comprehensive review of the state of knowledge regarding this essential mol.
- 102Wu, G.; Fang, Y.-Z.; Yang, S.; Lupton, J. R.; Turner, N. D. Glutathione Metabolism and Its Implications for Health. J. Nutr. 2004, 134 (3), 489– 492, DOI: 10.1093/jn/134.3.489Google Scholar102https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXitlCrurc%253D&md5=98f0af4f642d488fbfa0b4f43632512cGlutathione metabolism and its implications for healthWu, Guoyao; Fang, Yun-Zhong; Yang, Sheng; Lupton, Joanne R.; Turner, Nancy D.Journal of Nutrition (2004), 134 (3), 489-492CODEN: JONUAI; ISSN:0022-3166. (American Society for Nutritional Sciences)A review. Glutathione (γ-glutamyl-cysteinyl-glycine; GSH) is the most abundant low-mol.-wt. thiol, and GSH/glutathione disulfide is the major redox couple in animal cells. The synthesis of GSH from glutamate, cysteine, and glycine is catalyzed sequentially by two cytosolic enzymes, γ-glutamylcysteine synthetase and GSH synthetase. Compelling evidence shows that GSH synthesis is regulated primarily by γ-glutamylcysteine synthetase activity, cysteine availability, and GSH feedback inhibition. Animal and human studies demonstrate that adequate protein nutrition is crucial for the maintenance of GSH homeostasis. In addn., enteral or parenteral cystine, methionine, N-acetyl-cysteine, and L-2-oxothiazolidine-4-carboxylate are effective precursors of cysteine for tissue GSH synthesis. Glutathione plays important roles in antioxidant defense, nutrient metab., and regulation of cellular events (including gene expression, DNA and protein synthesis, cell proliferation and apoptosis, signal transduction, cytokine prodn. and immune response, and protein glutathionylation). Glutathione deficiency contributes to oxidative stress, which plays a key role in aging and the pathogenesis of many diseases (including kwashiorkor, seizure, Alzheimer's disease, Parkinson's disease, liver disease, cystic fibrosis, sickle cell anemia, HIV, AIDS, cancer, heart attack, stroke, and diabetes). New knowledge of the nutritional regulation of GSH metab. is crit. for the development of effective strategies to improve health and to treat these diseases.
- 103Bansal, A.; Simon, M. C. Glutathione Metabolism in Cancer Progression and Treatment Resistance. J. Cell Biol. 2018, 217 (7), 2291– 2298, DOI: 10.1083/jcb.201804161Google Scholar103https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFGntb3N&md5=2d071bd5b51fa8628dd56a2ee683931dGlutathione metabolism in cancer progression and treatment resistanceBansal, Ankita; Simon, M. CelesteJournal of Cell Biology (2018), 217 (7), 2291-2298CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)Glutathione (GSH) is the most abundant antioxidant found in living organisms and has multiple functions, most of which maintain cellular redox homeostasis. GSH preserves sufficient levels of cysteine and detoxifies xenobiotics while also conferring therapeutic resistance to cancer cells. However, GSH metab. plays both beneficial and pathogenic roles in a variety of malignancies. It is crucial to the removal and detoxification of carcinogens, and alterations in this pathway can have a profound effect on cell survival. Excess GSH promotes tumor progression, where elevated levels correlate with increased metastasis. In this review, we discuss recent studies that focus on deciphering the role of GSH in tumor initiation and progression as well as mechanisms underlying how GSH imparts treatment resistance to growing cancers. Targeting GSH synthesis/utilization therefore represents a potential means of rendering tumor cells more susceptible to different treatment options such as chemotherapy and radiotherapy.
- 104Bajic, 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: 10.1155/2019/5028181Google ScholarThere is no corresponding record for this reference.
- 105Fu, 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), 2891– 2919, DOI: 10.1038/s41596-020-0352-2Google Scholar105https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVWgt7bJ&md5=aebfe7aa84c68df1202c7f984b2672beA quantitative thiol reactivity profiling platform to analyze redox and electrophile reactive cysteine proteomesFu, Ling; Li, Zongmin; Liu, Keke; Tian, Caiping; He, Jixiang; He, Jingyang; He, Fuchu; Xu, Ping; Yang, JingNature Protocols (2020), 15 (9), 2891-2919CODEN: NPARDW; ISSN:1750-2799. (Nature Research)Cysteine is unique among all protein-coding amino acids, owing to its intrinsically high nucleophilicity. The cysteinyl thiol group can be covalently modified by a broad range of redox mechanisms or by various electrophiles derived from exogenous or endogenous sources. Measuring the response of protein cysteines to redox perturbation or electrophiles is crit. for understanding the underlying mechanisms involved. Activity-based protein profiling based on thiol-reactive probes has been the method of choice for such analyses. We therefore adapted this approach and developed a new chemoproteomic platform, termed 'QTRP' (quant. thiol reactivity profiling), that relies on the ability of a com. available thiol-reactive probe IPM (2-iodo-N-(prop-2-yn-1-yl)acetamide) to covalently label, enrich and quantify the reactive cysteinome in cells and tissues. Here, we provide a detailed and updated workflow of QTRP that includes procedures for (i) labeling of the reactive cysteinome from cell or tissue samples (e.g., control vs. treatment) with IPM, (ii) processing the protein samples into tryptic peptides and tagging the probe-modified peptides with isotopically labeled azido-biotin reagents contg. a photo-cleavable linker via click chem. reaction, (iii) capturing biotin-conjugated peptides with streptavidin beads, (iv) identifying and quantifying the photo-released peptides by mass spectrometry (MS)-based shotgun proteomics and (v) interpreting MS data by a streamlined informatic pipeline using a proteomics software, pFind 3, and an automatic post-processing algorithm. We also exemplified here how to use QTRP for mining H2O2-sensitive cysteines and for detg. the intrinsic reactivity of cysteines in a complex proteome. We anticipate that this protocol should find broad applications in redox biol., chem. biol. and the pharmaceutical industry. The protocol for sample prepn. takes 3 d, whereas MS measurements and data analyses require 75 min and <30 min, resp., per sample.
- 106Kuljanin, 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), 630– 641, DOI: 10.1038/s41587-020-00778-3Google Scholar106https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXkslSrug%253D%253D&md5=d256bc7ff87c75e4929fe4a6649242b4Reimagining high-throughput profiling of reactive cysteines for cell-based screening of large electrophile librariesKuljanin, Miljan; Mitchell, Dylan C.; Schweppe, Devin K.; Gikandi, Ajami S.; Nusinow, David P.; Bulloch, Nathan J.; Vinogradova, Ekaterina V.; Wilson, David L.; Kool, Eric T.; Mancias, Joseph D.; Cravatt, Benjamin F.; Gygi, Steven P.Nature Biotechnology (2021), 39 (5), 630-641CODEN: NABIF9; ISSN:1087-0156. (Nature Portfolio)Current methods used for measuring amino acid side-chain reactivity lack the throughput needed to screen large chem. libraries for interactions across the proteome. Here we redesigned the work flow for activity-based protein profiling of reactive cysteine residues by using a smaller desthiobiotin-based probe, sample multiplexing, reduced protein starting amts. and software to boost data acquisition in real time on the mass spectrometer. Our method, streamlined cysteine activity-based protein profiling (SLC-ABPP), achieved a 42-fold improvement in sample throughput, corresponding to profiling library members at a depth of >8,000 reactive cysteine sites at 18 min per compd. We applied it to identify proteome-wide targets of covalent inhibitors to mutant Kirsten rat sarcoma (KRAS)G12C and Bruton's tyrosine kinase (BTK). In addn., we created a resource of cysteine reactivity to 285 electrophiles in three human cell lines, which includes >20,000 cysteines from >6,000 proteins per line. The goal of proteome-wide profiling of cysteine reactivity across thousand-member libraries under several cellular contexts is now within reach.
- 107Yamamoto, 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), 1169– 1203, DOI: 10.1152/physrev.00023.2017Google Scholar107https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXkvFektrc%253D&md5=906947c8ad97c48f60e46bc44103664aThe KEAP1-NRF2 system: a thiol-based sensor-effector apparatus for maintaining redox homeostasisYamamoto, Masayuki; Kensler, Thomas W.; Motohashi, HozumiPhysiological Reviews (2018), 98 (3), 1169-1203CODEN: PHREA7; ISSN:1522-1210. (American Physiological Society)A review. The Kelch-like ECH-assocd. protein 1-NF-E2-related factor 2 (KEAP1-NRF2) system forms the major node of cellular and organismal defense against oxidative and electrophilic stresses of both exogenous and endogenous origins. KEAP1 acts as a cysteine thiol-rich sensor of redox insults, whereas NRF2 is a transcription factor that robustly transduces chem. signals to regulate a battery of cytoprotective genes. KEAP1 represses NRF2 activity under quiescent conditions, whereas NRF2 is liberated from KEAP1-mediated repression on exposure to stresses. The rapid inducibility of a response based on a derepression mechanism is an important feature of the KEAP1-NRF2 system. Recent studies have unveiled the complexities of the functional contributions of the KEAP1-NRF2 system and defined its broader involvement in biol. processes, including cell proliferation and differentiation, as well as cytoprotection. In this review, we describe historical milestones in the initial characterization of the KEAP1-NRF2 system and provide a comprehensive overview of the mol. mechanisms governing the functions of KEAP1 and NRF2, as well as their roles in physiol. and pathol. We also refer to the clin. significance of the KEAP1-NRF2 system as an important prophylactic and therapeutic target for various diseases, particularly aging-related disorders. We believe that controlled harnessing of the KEAP1-NRF2 system is a key to healthy aging and well-being in humans.
- 108Unoki, 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/ijms21020545Google ScholarThere is no corresponding record for this reference.
- 109Backus, K. M.; Cao, J.; Maddox, S. M. Opportunities and Challenges for the Development of Covalent Chemical Immunomodulators. Bioorg. Med. Chem. 2019, 27 (15), 3421– 3439, DOI: 10.1016/j.bmc.2019.05.050Google Scholar109https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFOku73L&md5=fbec9cd36ed1c0f3e9dbacf4d613730bOpportunities and challenges for the development of covalent chemical immunomodulatorsBackus, Keriann M.; Cao, Jian; Maddox, Sean M.Bioorganic & Medicinal Chemistry (2019), 27 (15), 3421-3439CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A review. Compds. that react irreversibly with cysteines have reemerged as potent and selective tools for altering protein function, serving as chem. probes and even clin. approved drugs. The exquisite sensitivity of human immune cell signaling pathways to oxidative stress indicates the likely, yet still underexploited, general utility of covalent probes for selective chem. immunomodulation. Here, we provide an overview of immunomodulatory cysteines, including identification of electrophilic compds. available to label these residues. We focus our discussion on three protein classes essential for cell signaling, which span the 'druggability' spectrum from amenable to chem. probes (kinases), somewhat druggable (proteases), to inaccessible (phosphatases). Using existing inhibitors as a guide, we identify general strategies to guide the development of covalent probes for selected undruggable classes of proteins and propose the application of such compds. to alter immune cell functions.
- 110Lincoln, 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), 166– 174, DOI: 10.1021/acssensors.1c01940Google ScholarThere is no corresponding record for this reference.
- 111Zheng, S.; Liu, G. Polymeric Emissive Materials Based on Dynamic Covalent Bonds. Mol. Basel Switz. 2022, 27 (19), 6635, DOI: 10.3390/molecules27196635Google ScholarThere is no corresponding record for this reference.
Cited By
This article has not yet been cited by other publications.
Article Views
Altmetric
Citations
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.
Recommended Articles
References
This article references 111 other publications.
- 1Fleming, 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), 780– 7901https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3MvpslCisA%253D%253D&md5=3c01ee5f9da7a862f0bd23359d039371On the antibacterial action of cultures of a penicillium, with special reference to their use in the isolation of B. influenzae. 1929Fleming ABulletin of the World Health Organization (2001), 79 (8), 780-90 ISSN:0042-9686.There is no expanded citation for this reference.
- 2Duncan, D.; Auclair, K. Itaconate: An Antimicrobial Metabolite of Macrophages. Can. J. Chem. 2022, 100 (2), 104– 113, DOI: 10.1139/cjc-2021-0117There is no corresponding record for this reference.
- 3Ray, 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), 1470– 1473, DOI: 10.1021/acschembio.8b002253https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXpsl2nsLc%253D&md5=d2dc8f255fd7e5f7537c57a0c5a3343eThe nitro group as a masked electrophile in covalent enzyme inhibitionRay, Sneha; Kreitler, Dale F.; Gulick, Andrew M.; Murkin, Andrew S.ACS Chemical Biology (2018), 13 (6), 1470-1473CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)We report the unprecedented reaction between a nitroalkane and an active-site cysteine residue to yield a thiohydroximate adduct. Structural and kinetic evidence suggests the nitro group is activated by conversion to its nitronic acid tautomer within the active site. The nitro group, therefore, shows promise as a masked electrophile in the design of covalent inhibitors targeting binding pockets with appropriately placed cysteine and general acid residues.
- 4Yuan, H.; Barnes, K. R.; Weissleder, R.; Cantley, L.; Josephson, L. Covalent Reactions of Wortmannin under Physiological Conditions. Chem. Biol. 2007, 14 (3), 321– 328, DOI: 10.1016/j.chembiol.2007.02.007There is no corresponding record for this reference.
- 5De Vita, E. 10 Years into the Resurgence of Covalent Drugs. Future Med. Chem. 2021, 13 (2), 193– 210, DOI: 10.4155/fmc-2020-02365https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3szjt1ertQ%253D%253D&md5=71ac37667e2f40fa3f6de36c07fea9ec10 years into the resurgence of covalent drugsDe Vita ElenaFuture medicinal chemistry (2021), 13 (2), 193-210 ISSN:.In the first decade of targeted covalent inhibition, scientists have successfully reversed the previous trend that had impeded the use of covalent inhibition in drug development. Successes in the clinic, mainly in the field of kinase inhibitors, are existing proof that safe covalent inhibitors can be designed and employed to develop effective treatments. The case of KRASG12C covalent inhibitors entering clinical trials in 2019 has been among the hottest topics discussed in drug discovery, raising expectations for the future of the field. In this perspective, an overview of the milestones hit with targeted covalent inhibitors, as well as the promise and the needs of current research, are presented. While recent results have confirmed the potential that was foreseen, many questions remain unexplored in this branch of precision medicine.
- 6Baillie, T. A. Drug-Protein Adducts: Past, Present, and Future. Med. Chem. Res. 2020, 29 (7), 1093– 1104, DOI: 10.1007/s00044-020-02567-8There is no corresponding record for this reference.
- 7Potashman, M. H.; Duggan, M. E. Covalent Modifiers: An Orthogonal Approach to Drug Design. J. Med. Chem. 2009, 52 (5), 1231– 1246, DOI: 10.1021/jm80085977https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhs1Sltbw%253D&md5=f8380d5a1cdd8638ad2a1e0405d25267Covalent Modifiers: An Orthogonal Approach to Drug DesignPotashman, Michele H.; Duggan, Mark E.Journal of Medicinal Chemistry (2009), 52 (5), 1231-1246CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. In this article we review a variety of examples in which therapeutic targets are covalently bound by small-mol. drugs or by compds. in advanced clin. development. The covalent interactions can be either reversible or irreversible, depending on the reaction partners.
- 8Boike, L.; Henning, N. J.; Nomura, D. K. Advances in Covalent Drug Discovery. Nat. Rev. Drug Discovery 2022, 21 (12), 881– 898, DOI: 10.1038/s41573-022-00542-z8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xit1Clt7nE&md5=b41441b3a7bc5f2e87bedf243fa43faaAdvances in covalent drug discoveryBoike, Lydia; Henning, Nathaniel J.; Nomura, Daniel K.Nature Reviews Drug Discovery (2022), 21 (12), 881-898CODEN: NRDDAG; ISSN:1474-1776. (Nature Portfolio)A review. Covalent drugs have been used to treat diseases for more than a century, but tools that facilitate the rational design of covalent drugs have emerged more recently. The purposeful addn. of reactive functional groups to existing ligands can enable potent and selective inhibition of target proteins, as demonstrated by the covalent epidermal growth factor receptor (EGFR) and Bruton's tyrosine kinase (BTK) inhibitors used to treat various cancers. Moreover, the identification of covalent ligands through 'electrophile-first' approaches has also led to the discovery of covalent drugs, such as covalent inhibitors for KRAS(G12C) and SARS-CoV-2 main protease. In particular, the discovery of KRAS(G12C) inhibitors validates the use of covalent screening technologies, which have become more powerful and widespread over the past decade. Chemoproteomics platforms have emerged to complement covalent ligand screening and assist in ligand discovery, selectivity profiling and target identification. This Review showcases covalent drug discovery milestones with emphasis on the lessons learned from these programs and how an evolving toolbox of covalent drug discovery techniques facilitates success in this field.
- 9Kenakin, T. The Mass Action Equation in Pharmacology. Br. J. Clin. Pharmacol. 2016, 81 (1), 41– 51, DOI: 10.1111/bcp.12810There is no corresponding record for this reference.
- 10Pottel, J.; Levit, A.; Korczynska, M.; Fischer, M.; Shoichet, B. K. The Recognition of Unrelated Ligands by Identical Proteins. ACS Chem. Biol. 2018, 13 (9), 2522– 2533, DOI: 10.1021/acschembio.8b00443There is no corresponding record for this reference.
- 11Knockenhauer, K. E.; Copeland, R. A. The Importance of Binding Kinetics and Drug-Target Residence Time in Pharmacology. Br. J. Pharmacol. 2023, 1– 14, DOI: 10.1111/bph.16104There is no corresponding record for this reference.
- 12Zhang, G.; Zhang, J.; Gao, Y.; Li, Y.; Li, Y. Strategies for Targeting Undruggable Targets. Expert Opin. Drug Discovery 2022, 17 (1), 55– 69, DOI: 10.1080/17460441.2021.196935912https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2crgvFelug%253D%253D&md5=20ebb3e7bd0c80af87802eea43a05dfaStrategies for targeting undruggable targetsZhang Gong; Zhang Juan; Gao Yuting; Li Yangfeng; Li Yizhou; Li YizhouExpert opinion on drug discovery (2022), 17 (1), 55-69 ISSN:.INTRODUCTION: Undruggable targets refer to clinically meaningful therapeutic targets that are 'difficult to drug' or 'yet to be drugged' via traditional approaches. Featuring characteristics of lacking defined ligand-binding pockets, non-catalytic protein-protein interaction functional modes and less-investigated 3D structures, these undruggable targets have been targeted with novel therapeutic entities developed with the progress of unconventional drug discovery approaches, such as targeted degradation molecules and display technologies. AREA COVERED: This review first presents the concept of 'undruggable' exemplified by RAS and other targets. Next, detailed strategies are illustrated in two aspects: innovation of therapeutic entities and development of unconventional drug discovery technologies. Finally, case studies covering typical undruggable targets (Bcl-2, p53, and RAS) are depicted to further demonstrate the feasibility of the strategies and entities above. EXPERT OPINION: Targeting the undruggable expands the scope of therapeutically reachable targets. Consequently, it represents the drug discovery frontier. Biomedical studies are capable of dissecting disease mechanisms, thus broadening the list of undruggable targets. Encouraged by the recent approval of the KRAS inhibitor Sotorasib, we believe that merging multiple discovery approaches and exploiting various novel therapeutic entities would pave the way for dealing with more 'undruggable' targets in the future.
- 13Akçay, 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), 931– 936, DOI: 10.1038/nchembio.217413https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVKqtb3P&md5=bda5720579a9b5e6027632d2cffba2e1Inhibition of Mcl-1 through covalent modification of a noncatalytic lysine side chainAkcay, Gizem; Belmonte, Matthew A.; Aquila, Brian; Chuaqui, Claudio; Hird, Alexander W.; Lamb, Michelle L.; Rawlins, Philip B.; Su, Nancy; Tentarelli, Sharon; Grimster, Neil P.; Su, QibinNature Chemical Biology (2016), 12 (11), 931-936CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Targeted covalent inhibition of disease-assocd. proteins has become a powerful methodol. in the field of drug discovery, leading to the approval of new therapeutics. Nevertheless, current approaches are often limited owing to their reliance on a cysteine residue to generate the covalent linkage. Here the authors used aryl boronic acid carbonyl warheads to covalently target a noncatalytic lysine side chain, and generated to the knowledge the first reversible covalent inhibitors for Mcl-1, a protein-protein interaction (PPI) target that has proven difficult to inhibit via traditional medicinal chem. strategies. These covalent binders exhibited improved potency in comparison to noncovalent congeners, as demonstrated in biochem. and cell-based assays. The authors identified Lys234 as the residue involved in covalent modification, via point mutation. The covalent binders discovered in this study will serve as useful starting points for the development of Mcl-1 therapeutics and probes to interrogate Mcl-1-dependent biol. phenomena.
- 14Huang, 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/s41392-021-00780-414https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2cfivFanug%253D%253D&md5=05a5959689e63910ecf752595d4407ffKRAS mutation: from undruggable to druggable in cancerHuang Lamei; Guo Zhixing; Wang Fang; Fu LiwuSignal transduction and targeted therapy (2021), 6 (1), 386 ISSN:.Cancer is the leading cause of death worldwide, and its treatment and outcomes have been dramatically revolutionised by targeted therapies. As the most frequently mutated oncogene, Kirsten rat sarcoma viral oncogene homologue (KRAS) has attracted substantial attention. The understanding of KRAS is constantly being updated by numerous studies on KRAS in the initiation and progression of cancer diseases. However, KRAS has been deemed a challenging therapeutic target, even "undruggable", after drug-targeting efforts over the past four decades. Recently, there have been surprising advances in directly targeted drugs for KRAS, especially in KRAS (G12C) inhibitors, such as AMG510 (sotorasib) and MRTX849 (adagrasib), which have obtained encouraging results in clinical trials. Excitingly, AMG510 was the first drug-targeting KRAS (G12C) to be approved for clinical use this year. This review summarises the most recent understanding of fundamental aspects of KRAS, the relationship between the KRAS mutations and tumour immune evasion, and new progress in targeting KRAS, particularly KRAS (G12C). Moreover, the possible mechanisms of resistance to KRAS (G12C) inhibitors and possible combination therapies are summarised, with a view to providing the best regimen for individualised treatment with KRAS (G12C) inhibitors and achieving truly precise treatment.
- 15Mons, 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/cpz1.41915https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitF2ksrrF&md5=1ffa766bb1fc413a27c47d458a3f54e7A Comprehensive Guide for Assessing Covalent Inhibition in Enzymatic Assays Illustrated with Kinetic SimulationsMons, Elma; Roet, Sander; Kim, Robbert Q.; Mulder, Monique P. C.Current Protocols (2022), 2 (6), e419CODEN: CPURDB; ISSN:2691-1299. (John Wiley & Sons, Inc.)Covalent inhibition has become more accepted in the past two decades, as illustrated by the clin. approval of several irreversible inhibitors designed to covalently modify their target. Elucidation of the structure-activity relationship and potency of such inhibitors requires a detailed kinetic evaluation. Here, we elucidate the relationship between the exptl. read-out and the underlying inhibitor binding kinetics. Interactive kinetic simulation scripts are employed to highlight the effects of in vitro enzyme activity assay conditions and inhibitor binding mode, thereby showcasing which assumptions and corrections are crucial. Four stepwise protocols to assess the biochem. potency of (ir)reversible covalent enzyme inhibitors targeting a nucleophilic active site residue are included, with accompanying data anal. tailored to the covalent binding mode. Together, this will serve as a guide to make an educated decision regarding the most suitable method to assess covalent inhibition potency.
- 16Chatterjee, 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), 17945– 17952, DOI: 10.1021/jacs.7b0893816https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslyrs7bI&md5=e6a38ff570134e956eb43bd4a874ca55Can Relative Binding Free Energy Predict Selectivity of Reversible Covalent Inhibitors?Chatterjee, Payal; Botello-Smith, Wesley M.; Zhang, Han; Qian, Li; Alsamarah, Abdelaziz; Kent, David; Lacroix, Jerome J.; Baudry, Michel; Luo, YunJournal of the American Chemical Society (2017), 139 (49), 17945-17952CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Reversible covalent inhibitors have many clin. advantages over noncovalent or irreversible covalent drugs. However, apart from selecting a warhead, substantial efforts in design and synthesis are needed to optimize noncovalent interactions to improve target-selective binding. Computational prediction of binding affinity for reversible covalent inhibitors presents a unique challenge since the binding process consists of multiple steps, which are not necessarily independent of each other. In this study, the authors lay out the relation between relative binding free energy and the overall reversible covalent binding affinity using a two-state binding model. To prove the concept, the authors employed free energy perturbation (FEP) coupled with λ-exchange mol. dynamics method to calc. the binding free energy of a series of α-ketoamide analogs relative to a common warhead scaffold, in both noncovalent and covalent binding states, and for two highly homologous proteases, calpain-1 and calpain-2. The authors conclude that covalent binding state alone, in general, can be used to predict reversible covalent binding selectivity. However, exceptions may exist. Therefore, the authors also discuss the conditions under which the noncovalent binding step is no longer negligible and propose to combine the relative FEP calcns. with a single QM/MM calcn. of warhead to predict the binding affinity and binding kinetics. The FEP calcns. also revealed that covalent and noncovalent binding states of an inhibitor do not necessarily exhibit the same selectivity. Thus, investigating both binding states, as well as the kinetics will provide extremely useful information for optimizing reversible covalent inhibitors.
- 17De Cesco, S.; Kurian, J.; Dufresne, C.; Mittermaier, A. K.; Moitessier, N. Covalent Inhibitors Design and Discovery. Eur. J. Med. Chem. 2017, 138, 96– 114, DOI: 10.1016/j.ejmech.2017.06.01917https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVKhsLrI&md5=551c9a9c4bd69d4ad4d9156c315d5a94Covalent inhibitors design and discoveryDe Cesco, Stephane; Kurian, Jerry; Dufresne, Caroline; Mittermaier, Anthony K.; Moitessier, NicolasEuropean Journal of Medicinal Chemistry (2017), 138 (), 96-114CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)In the history of therapeutics, covalent drugs occupy a very distinct category. While representing a significant fraction of the drugs on the market, very few have been deliberately designed to interact covalently with their biol. target. In this review, the prevalence of covalent drugs will first be briefly covered, followed by an introduction to their mechanisms of action and more detailed discussions of their discovery and the development of safe and efficient covalent enzyme inhibitors. All stages of a drug discovery program will be covered, from target considerations to lead optimization, strategies to tune reactivity and computational methods. The goal of this article is to provide an overview of the field and to outline good practices that are needed for the proper assessment and development of covalent inhibitors as well as a good understanding of the potential and limitations of current computational methods for the design of covalent drugs.
- 18Plescia, 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), 7874– 7884, DOI: 10.1021/acs.jmedchem.9b00642There is no corresponding record for this reference.
- 19Masuda, 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), 399– 409, DOI: 10.1248/cpb.c17-00854There is no corresponding record for this reference.
- 20Awoonor-Williams, E.; Walsh, A. G.; Rowley, C. N. Modeling Covalent-Modifier Drugs. Biochim. Biophys. Acta Proteins Proteomics 2017, 1865 (11), 1664– 1675, DOI: 10.1016/j.bbapap.2017.05.00920https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXotVOrtbo%253D&md5=ac579900da99ea005d0d562b53a1bca2Modeling covalent-modifier drugsAwoonor-Williams, Ernest; Walsh, Andrew G.; Rowley, Christopher N.Biochimica et Biophysica Acta, Proteins and Proteomics (2017), 1865 (11_Part_B), 1664-1675CODEN: BBAPBW; ISSN:1570-9639. (Elsevier B.V.)A review. In this review, we present a summary of how computer modeling has been used in the development of covalent-modifier drugs. Covalent-modifier drugs bind by forming a chem. bond with their target. This covalent binding can improve the selectivity of the drug for a target with complementary reactivity and result in increased binding affinities due to the strength of the covalent bond formed. In some cases, this results in irreversible inhibition of the target, but some targeted covalent inhibitor (TCI) drugs bind covalently but reversibly. Computer modeling is widely used in drug discovery, but different computational methods must be used to model covalent modifiers because of the chem. bonds formed. Structural and bioinformatic anal. has identified sites of modification that could yield selectivity for a chosen target. Docking methods, which are used to rank binding poses of large sets of inhibitors, have been augmented to support the formation of protein-ligand bonds and are now capable of predicting the binding pose of covalent modifiers accurately. The pKa's of amino acids can be calcd. in order to assess their reactivity towards electrophiles. QM/MM methods have been used to model the reaction mechanisms of covalent modification. The continued development of these tools will allow computation to aid in the development of new covalent-modifier drugs. This article is part of a Special Issue entitled: Biophysics in Canada, edited by Lewis Kay, John Baenziger, Albert Berghuis and Peter Tieleman.
- 21Scarpino, A.; Ferenczy, G. G.; Keserű, G. M. Binding Mode Prediction and Virtual Screening Applications by Covalent Docking. Methods Mol. Biol. Clifton NJ. 2021, 2266, 73– 88, DOI: 10.1007/978-1-0716-1209-5_4There is no corresponding record for this reference.
- 22Faridoon; 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), 1039– 1062, DOI: 10.1007/s00044-023-03065-322https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXovFKlurc%253D&md5=2f6664586b401207cbe840a4d50cee32An update on the discovery and development of reversible covalent inhibitorsFaridoon; Ng, Raymond; Zhang, Guiping; Li, Jie JackMedicinal Chemistry Research (2023), 32 (6), 1039-1062CODEN: MCREEB; ISSN:1054-2523. (Springer)A review. Small mol. drugs that covalently bind irreversibly to their target proteins have several advantages over conventional reversible inhibitors. They include increased duration of action, less-frequent drug dosing, reduced pharmacokinetic sensitivity, and the potential to target intractable shallow binding sites. Despite these advantages, the key challenges of irreversible covalent drugs are their potential for off-target toxicities and immunogenicity risks. Incorporating reversibility into covalent drugs would lead to less off-target toxicity by forming reversible adducts with off-target proteins and thus reducing the risk of idiosyncratic toxicities caused by the permanent modification of proteins, which leads to higher levels of potential haptens. Herein, we systematically review electrophilic warheads employed during the development of reversible covalent drugs. We hope the structural insights of electrophilic warheads would provide helpful information to medicinal chemists and aid in designing covalent drugs with better on-target selectivity and improved safety.
- 23Martin, J. S.; MacKenzie, C. J.; Fletcher, D.; Gilbert, I. H. Characterising Covalent Warhead Reactivity. Bioorg. Med. Chem. 2019, 27 (10), 2066– 2074, DOI: 10.1016/j.bmc.2019.04.00223https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXntVGgsbo%253D&md5=487334d8d0a8b03d0a25d6d87c1b4329Characterising covalent warhead reactivityMartin, James S.; MacKenzie, Claire J.; Fletcher, Daniel; Gilbert, Ian H.Bioorganic & Medicinal Chemistry (2019), 27 (10), 2066-2074CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Many drugs currently used are covalent inhibitors and irreversibly inhibit their targets. Most of these were discovered through serendipity. Covalent inhibitions can have many advantages from a pharmacokinetic perspective. However, until recently most organizations have shied away from covalent compd. design due to fears of non-specific inhibition of off-target proteins leading to toxicity risks. However, there has been a renewed interest in covalent modifiers as potential drugs, as it possible to get highly selective compds. It is therefore important to know how reactive a warhead is and to be able to select the least reactive warhead possible to avoid toxicity. A robust NMR based assay was developed and used to measure the reactivity of a variety of covalent warheads against serine and cysteine - the two most common targets for covalent drugs. A selection of these warheads also had their reactivity measured against threonine, tyrosine, lysine, histidine and arginine to better understand our ability to target non-traditional residues. The reactivity was also measured at various pHs to assess what effect the environment in the active site would have on these reactions. The reactivity of a covalent modifier was found to be very dependent on the amino acid residue.
- 24Péczka, N.; Orgován, Z.; Ábrányi-Balogh, P.; Keserű, G. M. Electrophilic Warheads in Covalent Drug Discovery: An Overview. Expert Opin. Drug Discovery 2022, 17 (4), 413– 422, DOI: 10.1080/17460441.2022.2034783There is no corresponding record for this reference.
- 25Jackson, 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), 839– 885, DOI: 10.1021/acs.jmedchem.6b0078825https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitV2rtbnM&md5=36d3be5bf688c4e4a8cf4bcf42073009Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition ReactionsJackson, Paul A.; Widen, John C.; Harki, Daniel A.; Brummond, Kay M.Journal of Medicinal Chemistry (2017), 60 (3), 839-885CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Although Michael acceptors display a potent and broad spectrum of bioactivity, they have largely been ignored in drug discovery because of their presumed indiscriminate reactivity. As such, a dearth of information exists relevant to the thiol reactivity of natural products and their analogs possessing this moiety. In the midst of recently approved acrylamide-contg. drugs, it is clear that a good understanding of the hetero-Michael addn. reaction and the relative reactivities of biol. thiols with Michael acceptors under physiol. conditions is needed for the design and use of these compds. as biol. tools and potential therapeutics. This Perspective provides information that will contribute to this understanding, such as kinetics of thiol addn. reactions, bioactivities, as well as steric and electronic factors that influence the electrophilicity and reversibility of Michael acceptors. This Perspective is focused on α,β-unsatd. carbonyls given their preponderance in bioactive natural products.
- 26Watt, 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), 8898– 8906, DOI: 10.1039/D2OB01369JThere is no corresponding record for this reference.
- 27Watt, 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), 2204– 2212, DOI: 10.1039/D2OB02223KThere is no corresponding record for this reference.
- 28Lohman, 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), 14473– 14475, DOI: 10.1021/ja406381b28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVGjtLzN&md5=f7d0c69687c776310dda4facf967db64Catalysis by desolvation: The catalytic prowess of SAM-dependent halide-alkylating enzymesLohman, Danielle C.; Edwards, David R.; Wolfenden, RichardJournal of the American Chemical Society (2013), 135 (39), 14473-14475CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)In the biol. fixation of halide ions, several enzymes have been found to catalyze alkyl transfer from S-adenosylmethionine (SAM) to halide ions. It proved possible to measure the rates of reaction of the trimethylsulfonium ion with I-, Br-, Cl-, F-, HO-, and H2O in water at elevated temps. Comparison of the resulting 2nd-order rate consts., extrapolated to 25°, with values of kcat/Km reported for fluorinase and chlorinase indicated that these enzymes enhanced the rates of alkyl halide formation by factors of 2 × 1015- and 1 × 1017-fold, resp. These rate enhancements, achieved without the assistance of cofactors, metal ions, or general acid-base catalysis, were the largest that have been reported for an enzyme that acts on 2 substrates.
- 29Parvez, S.; Long, M. J. C.; Poganik, J. R.; Aye, Y. Redox Signaling by Reactive Electrophiles and Oxidants. Chem. Rev. 2018, 118 (18), 8798– 8888, DOI: 10.1021/acs.chemrev.7b0069829https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFyjurnI&md5=2cc16400a3dcab3bac28c25132595c7aRedox Signaling by Reactive Electrophiles and OxidantsParvez, Saba; Long, Marcus J. C.; Poganik, Jesse R.; Aye, YimonChemical Reviews (Washington, DC, United States) (2018), 118 (18), 8798-8888CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. The concept of cell signaling in the context of non-enzyme-assisted protein modifications by reactive electrophilic and oxidative species, broadly known as redox signaling, is a uniquely complex topic that has been approached from numerous different and multidisciplinary angles. The authors' review reflects on 5 aspects crit. for understanding how Nature harnesses these non-canonical post-translational modifications to coordinate distinct cellular activities: (1) specific players and their generation; (2) physicochem. properties; (3) mechanisms of action; (4) methods of interrogation; and (5) functional roles in health and disease. Emphasis is primarily placed on the latest progress in the field, but several aspects of classical work likely forgotten/lost are also recollected. For researchers with interests in getting into the field, this review is anticipated to function as a primer. For the expert, the aim is to stimulate thought and discussion about fundamentals of redox signaling mechanisms, and nuances of specificity/selectivity and timing in this sophisticated yet fascinating arena at the crossroads of chem. and biol.
- 30Krishnan, 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), 12624– 12630, DOI: 10.1021/ja505194w30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVWrtbvK&md5=de052bac07c8920cb807c05f71f4f95dDesign of Reversible, Cysteine-Targeted Michael Acceptors Guided by Kinetic and Computational AnalysisKrishnan, Shyam; Miller, Rand M.; Tian, Boxue; Mullins, R. Dyche; Jacobson, Matthew P.; Taunton, JackJournal of the American Chemical Society (2014), 136 (36), 12624-12630CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Electrophilic probes that covalently modify a cysteine thiol often show enhanced pharmacol. potency and selectivity. Although reversible Michael acceptors have been reported, the structural requirements for reversibility are poorly understood. Here, we report a novel class of acrylonitrile-based Michael acceptors, activated by aryl or heteroaryl electron-withdrawing groups. We demonstrate that thiol adducts of these acrylonitriles undergo β-elimination at rates that span more than 3 orders of magnitude. These rates correlate inversely with the computed proton affinity of the corresponding carbanions, enabling the intrinsic reversibility of the thiol-Michael reaction to be tuned in a predictable manner. We apply these principles to the design of new reversible covalent kinase inhibitors with improved properties. A cocrystal structure of one such inhibitor reveals specific noncovalent interactions between the 1,2,4-triazole activating group and the kinase. Our exptl. and computational study enables the design of new Michael acceptors, expanding the palette of reversible, cysteine-targeted electrophiles.
- 31Ma, 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), 6146– 6162, DOI: 10.1021/acs.jmedchem.9b0038731https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFGqsLjI&md5=2de757026f2542bff8092060a72107a5Application of Dually Activated Michael Acceptor to the Rational Design of Reversible Covalent Inhibitor for Enterovirus 71 3C ProteaseMa, Yuying; Li, Linfeng; He, Shuai; Shang, Chengyou; Sun, Yang; Liu, Ning; Meek, Thomas D.; Wang, Yaxin; Shang, LuqingJournal of Medicinal Chemistry (2019), 62 (13), 6146-6162CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Targeted covalent inhibitors (TCIs) have attracted growing attention from the pharmaceutical industry in recent decades because they have potential advantages in terms of efficacy, selectivity, and safety. TCIs have recently evolved into a new version with reversibility that can be systematically modulated. This feature may diminish the risk of haptenization and help optimize the drug-target residence time as needed. The enteroviral 3C protease (3Cpro) is a valuable therapeutic target, but the development of 3Cpro inhibitors is far from satisfactory. Therefore, we aimed to apply a reversible TCI approach to the design of novel 3Cpro inhibitors. The introduction of various substituents onto the α-carbon of classical Michael acceptors yielded inhibitors bearing several classes of warheads. Using steady-state kinetics and biomol. mass spectrometry, we confirmed the mode of reversible covalent inhibition and elucidated the mechanism by which the potency and reversibility were affected by electronic and steric factors. This research produced several potent inhibitors with good selectivity and suitable reversibility; moreover, it validated the reversible TCI approach in the field of viral infection, suggesting broader applications in the design of reversible covalent inhibitors for other proteases.
- 32Bradshaw, 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), 525– 531, DOI: 10.1038/nchembio.181732https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFeju7%252FE&md5=85a73d9ecd62695d03166f7009c68dd8Prolonged and tunable residence time using reversible covalent kinase inhibitorsBradshaw, J. Michael; McFarland, Jesse M.; Paavilainen, Ville O.; Bisconte, Angelina; Tam, Danny; Phan, Vernon T.; Romanov, Sergei; Finkle, David; Shu, Jin; Patel, Vaishali; Ton, Tony; Li, Xiaoyan; Loughhead, David G.; Nunn, Philip A.; Karr, Dane E.; Gerritsen, Mary E.; Funk, Jens Oliver; Owens, Timothy D.; Verner, Erik; Brameld, Ken A.; Hill, Ronald J.; Goldstein, David M.; Taunton, JackNature Chemical Biology (2015), 11 (7), 525-531CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Drugs with prolonged on-target residence times often show superior efficacy, yet general strategies for optimizing drug-target residence time are lacking. Here the authors made progress toward this elusive goal by targeting a noncatalytic cysteine in Bruton's tyrosine kinase (BTK) with reversible covalent inhibitors. Using an inverted orientation of the cysteine-reactive cyanoacrylamide electrophile, the authors identified potent and selective BTK inhibitors that demonstrated biochem. residence times spanning from minutes to 7 d. An inverted cyanoacrylamide with prolonged residence time in vivo remained bound to BTK for more than 18 h after clearance from the circulation. The inverted cyanoacrylamide strategy was further used to discover fibroblast growth factor receptor (FGFR) kinase inhibitors with residence times of several days, demonstrating the generalizability of the approach. Targeting of noncatalytic cysteines with inverted cyanoacrylamides may serve as a broadly applicable platform that facilitates 'residence time by design', the ability to modulate and improve the duration of target engagement in vivo.
- 33Zhou, 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.2022.114571There is no corresponding record for this reference.
- 34Ehmke, 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), 5764– 5768, DOI: 10.1039/c2ob00034b34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVSlsr3J&md5=45858b629d24a54b72cd7e3b7ba15e5bTuning and predicting biological affinity: aryl nitriles as cysteine protease inhibitorsEhmke, Veronika; Quinsaat, Jose Enrico Q.; Rivera-Fuentes, Pablo; Heindl, Cornelia; Freymond, Celine; Rottmann, Matthias; Brun, Reto; Schirmeister, Tanja; Diederich, FrancoisOrganic & Biomolecular Chemistry (2012), 10 (30), 5764-5768CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)A series of aryl nitrile-based ligands were prepd. to investigate the effect of their electrophilicity on the affinity against the cysteine proteases rhodesain and human cathepsin L. D. functional theory calcns. provided relative reactivities of the nitriles, enabling prediction of their biol. affinity and cytotoxicity and a clear structure-activity relationship.
- 35Langrish, 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), 1454– 1468, DOI: 10.4049/jimmunol.2001130There is no corresponding record for this reference.
- 36Jung, 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), 12322– 12358, DOI: 10.1021/acs.jmedchem.1c0100236https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslCitbvM&md5=322d56630ffe7e55bfd42ba0f8d62e97Fluorovinylsulfones and -Sulfonates as Potent Covalent Reversible Inhibitors of the Trypanosomal Cysteine Protease Rhodesain: Structure-Activity Relationship, Inhibition Mechanism, Metabolism, and In Vivo StudiesJung, Sascha; Fuchs, Natalie; Johe, Patrick; Wagner, Annika; Diehl, Erika; Yuliani, Tri; Zimmer, Collin; Barthels, Fabian; Zimmermann, Robert A.; Klein, Philipp; Waigel, Waldemar; Meyr, Jessica; Opatz, Till; Tenzer, Stefan; Distler, Ute; Raeder, Hans-Joachim; Kersten, Christian; Engels, Bernd; Hellmich, Ute A.; Klein, Jochen; Schirmeister, TanjaJournal of Medicinal Chemistry (2021), 64 (16), 12322-12358CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Rhodesain is a major cysteine protease of Trypanosoma brucei rhodesiense, a pathogen causing Human African Trypanosomiasis, and a validated drug target. Recently, we reported the development of α-halovinylsulfones as a new class of covalent reversible cysteine protease inhibitors. Here, α-fluorovinylsulfones/-sulfonates were optimized for rhodesain based on mol. modeling approaches. I (X = F), the most potent and selective inhibitor in the series, shows a single-digit nanomolar affinity and high selectivity toward mammalian cathepsins B and L. Enzymic diln. assays and MS expts. indicate that I (X = F) is a slow-tight binder (Ki = 3 nM). Furthermore, the nonfluorinated I (X = H) shows favorable metab. and biodistribution by accumulation in mice brain tissue after i.p. and oral administration. The highest antitrypanosomal activity was obsd. for inhibitors with an N-terminal 2,3-dihydrobenzo[b][1,4]dioxine group and a 4-Me-Phe residue in P2 with nanomolar EC50 values (0.14/0.80μM). The different mechanisms of reversible and irreversible inhibitors were explained using QM/MM calcns. and MD simulations.
- 37Feral, 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, 1775– 1800, DOI: 10.1007/s00726-023-03286-1There is no corresponding record for this reference.
- 38Fairhurst, 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), 12542– 12573, DOI: 10.1021/acs.jmedchem.0c0101938https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVeru77E&md5=94bfa1740e9cac34d0ceac67bc9181a7Discovery of Roblitinib (FGF401) as a Reversible-Covalent Inhibitor of the Kinase Activity of Fibroblast Growth Factor Receptor 4Fairhurst, Robin A.; Knoepfel, Thomas; Buschmann, Nicole; Leblanc, Catherine; Mah, Robert; Todorov, Milen; Nimsgern, Pierre; Ripoche, Sebastien; Niklaus, Michel; Warin, Nicolas; Luu, Van Huy; Madoerin, Mario; Wirth, Jasmin; Graus-Porta, Diana; Weiss, Andreas; Kiffe, Michael; Wartmann, Markus; Kinyamu-Akunda, Jacqueline; Sterker, Dario; Stamm, Christelle; Adler, Flavia; Buhles, Alexandra; Schadt, Heiko; Couttet, Philippe; Blank, Jutta; Galuba, Inga; Trappe, Jorg; Voshol, Johannes; Ostermann, Nils; Zou, Chao; Berghausen, Jorg; Del Rio Espinola, Alberto; Jahnke, Wolfgang; Furet, PascalJournal of Medicinal Chemistry (2020), 63 (21), 12542-12573CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)FGF19 signaling through the FGFR4/β-klotho receptor complex has been shown to be a key driver of growth and survival in a subset of hepatocellular carcinomas, making selective FGFR4 inhibition an attractive treatment opportunity. A kinome-wide sequence alignment highlighted a poorly conserved cysteine residue within the FGFR4 ATP-binding site at position 552, two positions beyond the gate-keeper residue. Several strategies for targeting this cysteine to identify FGFR4 selective inhibitor starting points are summarized which made use of both rational and unbiased screening approaches. The optimization of a 2-formylquinoline amide hit series is described in which the aldehyde makes a hemithioacetal reversible-covalent interaction with cysteine 552. Key challenges addressed during the optimization are improving the FGFR4 potency, metabolic stability, and soly. leading ultimately to the highly selective first-in-class clin. candidate roblitinib.
- 39Shindo, 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), 250– 258, DOI: 10.1038/s41589-018-0204-339https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXlvFSgur4%253D&md5=f6c8886d540ea26a8abecd950feba913Selective and reversible modification of kinase cysteines with chlorofluoroacetamidesShindo, Naoya; Fuchida, Hirokazu; Sato, Mami; Watari, Kosuke; Shibata, Tomohiro; Kuwata, Keiko; Miura, Chizuru; Okamoto, Kei; Hatsuyama, Yuji; Tokunaga, Keisuke; Sakamoto, Seiichi; Morimoto, Satoshi; Abe, Yoshito; Shiroishi, Mitsunori; Caaveiro, Jose M. M.; Ueda, Tadashi; Tamura, Tomonori; Matsunaga, Naoya; Nakao, Takaharu; Koyanagi, Satoru; Ohdo, Shigehiro; Yamaguchi, Yasuchika; Hamachi, Itaru; Ono, Mayumi; Ojida, AkioNature Chemical Biology (2019), 15 (3), 250-258CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Irreversible inhibition of disease-assocd. proteins with small mols. is a powerful approach for achieving increased and sustained pharmacol. potency. Here, we introduce α-chlorofluoroacetamide (CFA) as a novel warhead of targeted covalent inhibitor (TCI). Despite weak intrinsic reactivity, CFA-appended quinazoline showed high reactivity toward Cys797 of epidermal growth factor receptor (EGFR). In cells, CFA-quinazoline showed higher target specificity for EGFR than the corresponding Michael acceptors in a wide concn. range (0.1-10 μM). The cysteine adduct of the CFA deriv. was susceptible to hydrolysis and reversibly yielded intact thiol but was stable in solvent-sequestered ATP-binding pocket of EGFR. This environment-dependent hydrolysis can potentially reduce off-target protein modification by CFA-based drugs. Oral administration of CFA quinazoline, (2R)-1-(2-chloro-2-fluoroacetyl)-N-[4-[(3-chloro-4-fluorophenyl)amino]-7-[3-(4-morpholinyl)-propoxy]quinazolin-6-yl]pyrrolidine-2-carboxamide [2226257-92-5] (NS-062, compd. 51) significantly suppressed tumor growth in a mouse xenograft model. Further, CFA-appended pyrazolopyrimidine irreversibly inhibited Bruton's tyrosine kinase with higher target specificity. These results demonstrate the utility of CFA as a new class warheads for TCI.
- 40Tallon, 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), 16069– 16080, DOI: 10.1021/jacs.3c04444There is no corresponding record for this reference.
- 41Ingiliz, 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), 1194– 1199, DOI: 10.1111/j.1478-3231.2012.02796.xThere is no corresponding record for this reference.
- 42Teicher, B. A.; Tomaszewski, J. E. Proteasome Inhibitors. Biochem. Pharmacol. 2015, 96 (1), 1– 9, DOI: 10.1016/j.bcp.2015.04.008There is no corresponding record for this reference.
- 43Metcalf, 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), 321– 326, DOI: 10.1021/acsmedchemlett.6b0049143https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFWqur0%253D&md5=63766931fdf731c68fd284cf86cc77d0Discovery of GBT440, an Orally Bioavailable R-State Stabilizer of Sickle Cell HemoglobinMetcalf, Brian; Chuang, Chihyuan; Dufu, Kobina; Patel, Mira P.; Silva-Garcia, Abel; Johnson, Carl; Lu, Qing; Partridge, James R.; Patskovska, Larysa; Patskovsky, Yury; Almo, Steven C.; Jacobson, Matthew P.; Hua, Lan; Xu, Qing; Gwaltney, Stephen L.; Yee, Calvin; Harris, Jason; Morgan, Bradley P.; James, Joyce; Xu, Donghong; Hutchaleelaha, Athiwat; Paulvannan, Kumar; Oksenberg, Donna; Li, ZheACS Medicinal Chemistry Letters (2017), 8 (3), 321-326CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)The authors report the discovery of a new potent allosteric effector of sickle cell Hb, GBT440 I, that increases the affinity of Hb for oxygen and consequently inhibits its polymn. when subjected to hypoxic conditions. Unlike earlier allosteric activators that bind covalently to Hb in a 2:1 stoichiometry, I binds with a 1:1 stoichiometry. Compd. I is orally bioavailable and partitions highly and favorably into the red blood cell with a RBC/plasma ratio of ∼150. This partitioning onto the target protein is anticipated to allow therapeutic concns. to be achieved in the red blood cell at low plasma concns. I is in Phase 2 clin. trials for the treatment of sickle cell disease (NCT02285088).
- 44Reja, 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), 1152– 1157, DOI: 10.1021/jacs.1c1270244https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xht1egs7s%253D&md5=962dedfe735ef240b3430dfd74cdf5cbLysine-Targeting Reversible Covalent Inhibitors with Long Residence TimeReja, Rahi M.; Wang, Wenjian; Lyu, Yuhan; Haeffner, Fredrik; Gao, JianminJournal of the American Chemical Society (2022), 144 (3), 1152-1157CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We report a new reversible lysine conjugation that features a novel diazaborine product and much slowed dissocn. kinetics in comparison to the previously known iminoboronate chem. Incorporating the diazaborine-forming warhead RMR1 to a peptide ligand gives potent and long-acting reversible covalent inhibitors of the staphylococcal sortase. The efficacy of sortase inhibition is demonstrated via biochem. and cell-based assays. A comparative study of RMR1 and an iminoboronate-forming warhead highlights the significance and potential of modulating bond dissocn. kinetics in achieving long-acting reversible covalent inhibitors.
- 45Serafimova, 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), 471– 476, DOI: 10.1038/nchembio.92545https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XkvVKitrw%253D&md5=6455a9a029714beb317286f71ac1326bReversible targeting of noncatalytic cysteines with chemically tuned electrophilesSerafimova, Iana M.; Pufall, Miles A.; Krishnan, Shyam; Duda, Katarzyna; Cohen, Michael S.; Maglathlin, Rebecca L.; McFarland, Jesse M.; Miller, Rand M.; Froedin, Morten; Taunton, JackNature Chemical Biology (2012), 8 (5), 471-476CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Targeting noncatalytic cysteine residues with irreversible acrylamide-based inhibitors is a powerful approach for enhancing pharmacol. potency and selectivity. Nevertheless, concerns about off-target modification motivate the development of reversible cysteine-targeting strategies. Here we show that electron-deficient olefins, including acrylamides, can be tuned to react with cysteine thiols in a rapidly reversible manner. Installation of a nitrile group increased the olefins' intrinsic reactivity, but, paradoxically, eliminated the formation of irreversible adducts. Incorporation of these electrophiles into a noncovalent kinase-recognition scaffold produced slowly dissocg., covalent inhibitors of the p90 ribosomal protein S6 kinase RSK2. A cocrystal structure revealed specific noncovalent interactions that stabilize the complex by positioning the electrophilic carbon near the targeted cysteine. Disruption of these interactions by protein unfolding or proteolysis promoted instantaneous cleavage of the covalent bond. Our results establish a chem.-based framework for engineering sustained covalent inhibition without accumulating permanently modified proteins and peptides.
- 46Forster, 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), 1335– 1340, DOI: 10.1016/j.chembiol.2016.10.00846https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVeqsLrJ&md5=348c5b5fbfa91fd02fa21d984620ea16Selective JAK3 Inhibitors with a Covalent Reversible Binding Mode Targeting a New Induced Fit Binding PocketForster, Michael; Chaikuad, Apirat; Bauer, Silke M.; Holstein, Julia; Robers, Matthew B.; Corona, Cesear R.; Gehringer, Matthias; Pfaffenrot, Ellen; Ghoreschi, Kamran; Knapp, Stefan; Laufer, Stefan A.Cell Chemical Biology (2016), 23 (11), 1335-1340CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)Janus kinases (JAKs) are a family of cytoplasmatic tyrosine kinases that are attractive targets for the development of anti-inflammatory drugs given their roles in cytokine signaling. One question regarding JAKs and their inhibitors that remains under intensive debate is whether JAK inhibitors should be isoform selective. Since JAK3 functions are restricted to immune cells, an isoform-selective inhibitor for JAK3 could be esp. valuable to achieve clin. more useful and precise effects. However, the high degree of structural conservation makes isoform-selective targeting a challenging task. Here, we present picomolar inhibitors with unprecedented kinome-wide selectivity for JAK3. Selectivity was achieved by concurrent covalent reversible targeting of a JAK3-specific cysteine residue and a ligand-induced binding pocket. We confirmed that in vitro activity and selectivity translate well into the cellular environment and suggest that our inhibitors are powerful tools to elucidate JAK3-specific functions.
- 47Dietze, 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), 1097– 1103, DOI: 10.1021/tx970090u47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK1c%252FgvFSgtQ%253D%253D&md5=9f71f1d99f6cdbc647091eacb12e6feeInactivation of glyceraldehyde-3-phosphate dehydrogenase by a reactive metabolite of acetaminophen and mass spectral characterization of an arylated active site peptideDietze E C; Schafer A; Omichinski J G; Nelson S DChemical research in toxicology (1997), 10 (10), 1097-103 ISSN:0893-228X.Acetaminophen (4'-hydroxyacetanilide, APAP) is a widely used analgesic and antipyretic drug that can cause hepatic necrosis under some circumstances via cytochrome P450-mediated oxidation to a reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI). Although the mechanism of hepatocellular injury caused by APAP is not fully understood, it is known that NAPQI forms covalent adducts with several hepatocellular proteins. Reported here is the identification of one of these proteins as glyceraldehyde-3-phosphate dehydrogenase [GAPDH, D-glyceraldehyde-3-phosphate: NAD+ oxidoreductase (phosphorylating), EC 1.2.1.12]. Two hours after the administration of hepatotoxic doses of [14C]APAP to mice, at a time prior to overt cell damage, hepatocellular GAPDH activity was significantly decreased concurrent with the formation of a 14C-labeled GAPDH adduct. A nonhepatotoxic regioisomer of APAP, 3'-hydroxyacetanilide (AMAP), was found to decrease GAPDH activity to a lesser extent than APAP, and radiolabel from [14C]AMAP bound to a lesser extent to GAPDH at a time when its overall binding to hepatocellular proteins was almost equivalent to that of APAP. In order to determine the nature of the covalent adduct between GAPDH and APAP, its major reactive and toxic metabolite, NAPQI, was incubated with purified porcine muscle GAPDH. Microsequencing analysis and fast atom bombardment mass spectrometry (FAB-MS) with collision-induced dissociation (CID) were used to characterize one of the adducts as APAP bound to the cysteinyl sulfhydryl group of Cys-149 in the active site peptide of GAPDH.
- 48Rombach, 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), 178– 184, DOI: 10.1021/tx970076h48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXhslarsLc%253D&md5=1367c196a23344bf587b9d346bc5e7d1Identification of a Rat Liver Microsomal Esterase as a Target Protein for Bromobenzene MetabolitesRombach, Elizabeth M.; Hanzlik, Robert P.Chemical Research in Toxicology (1998), 11 (3), 178-184CODEN: CRTOEC; ISSN:0893-228X. (American Chemical Society)To identify proteins targeted by bromobenzene metabolites, we incubated [14C]bromobenzene in vitro with liver microsomes from phenobarbital-induced rats under conditions which typically led to covalent binding of 2-4 nmol equiv of bromobenzene/mg of protein. Microsomal proteins were solubilized with detergent, sepd. by chromatog. and electrophoresis, and analyzed for 14C by phosphorimaging of stained blots. Much of the radioactivity was assocd. with several bands of proteins of ∼50-60 kDa, plus another prominent band around 70 kDa, but labeling d. appeared to vary considerably overall. A major radiolabeled protein was purified by preparative electrophoresis and submitted to automated Edman microsequencing. Its N-terminal sequence was found to correspond to that of a known rat liver microsomal carboxylesterase (E.C. 3.1.1.1) previously identified as a target for reactive metabolites of halothane. The extent to which covalent modification of this protein by reactive metabolites contributes to the prodn. of hepatotoxic effects remains to be detd.
- 49Tailor, A.; Waddington, J. C.; Meng, X.; Park, B. K. Mass Spectrometric and Functional Aspects of Drug-Protein Conjugation. Chem. Res. Toxicol. 2016, 29 (12), 1912– 1935, DOI: 10.1021/acs.chemrestox.6b0014749https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFymu7fI&md5=488bc6cacaf831918db1c28c2f70aa8cMass Spectrometric and Functional Aspects of Drug-Protein ConjugationTailor, Arun; Waddington, James C.; Meng, Xiaoli; Park, B. KevinChemical Research in Toxicology (2016), 29 (12), 1912-1935CODEN: CRTOEC; ISSN:0893-228X. (American Chemical Society)A review. The covalent binding of drugs (metabolites) to proteins to form drug-protein adducts can have an adverse effect on the body. These adducts are thought to be responsible for idiosyncratic drug reactions including severe drug hypersensitivity reactions. Major advances in proteomics technol. have allowed for the identification and quantification of target proteins for certain drugs. Human serum albumin (HSA) and Hb have been identified as accessible targets, and potential biomarkers for drug-protein adducts formation, for numerous drugs (metabolites) including β-lactam antibiotics, reactive drug metabolites such as quinone imines (acetaminophen) and acyl glucuronides (diclofenac), and covalent inhibitors (neratinib). For example, MS/MS anal. of plasma samples from patients taking flucloxacillin revealed that flucloxacillin and its 5-hydroxymethyl metabolite formed covalent adducts with lysine residues on albumin via opening of the β-lactam ring. Other proteins such as P 450 and keratin are also potential targets for covalent binding. However, for most drugs, the properties of these target proteins including their location, their quantity, the timing of conjugate generation, and their biol. function are not well understood. In this review, currently available proteomic technologies including MS/MS anal. to identify antigens, precise location of modifications, and the immunol. consequence of hapten-protein complex are illustrated. Moving forward, identification of the nature of the antigenic determinants that trigger immune responses to drug protein adducts will increase the authors' ability to predict idiosyncratic toxicity for a given compd.
- 50Jeffery, D. A.; Bogyo, M. Chemical Proteomics and Its Application to Drug Discovery. Curr. Opin. Biotechnol. 2003, 14 (1), 87– 95, DOI: 10.1016/S0958-1669(02)00010-150https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXps1WmtQ%253D%253D&md5=9d77283dcea8e41a994dbcd4d14b382bChemical proteomics and its application to drug discoveryJeffery, Douglas A.; Bogyo, MatthewCurrent Opinion in Biotechnology (2003), 14 (1), 87-95CODEN: CUOBE3; ISSN:0958-1669. (Elsevier Science Ltd.)A review with refs. The completion of the human genome sequencing project has provided a flood of new information that is likely to change the way scientists approach the study of complex biol. systems. A major challenge lies in translating this information into new and better ways to treat human disease. The multidisciplinary science of chem. proteomics can be used to distill this flood of new information. This approach makes use of synthetic small mols. that can be used to covalently modify a set of related enzymes and subsequently allow their purifn. and/or identification as valid drug targets. Furthermore, such methods enable rapid biochem. anal. and small-mol. screening of targets thereby accelerating the often difficult process of target validation and drug discovery.
- 51Weerapana, 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), 790– 795, DOI: 10.1038/nature0947251https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsVGhsr%252FI&md5=277e436e7a8f4dfff0b548bce52ac40dQuantitative reactivity profiling predicts functional cysteines in proteomesWeerapana, Eranthie; Wang, Chu; Simon, Gabriel M.; Richter, Florian; Khare, Sagar; Dillon, Myles B. D.; Bachovchin, Daniel A.; Mowen, Kerri; Baker, David; Cravatt, Benjamin F.Nature (London, United Kingdom) (2010), 468 (7325), 790-795CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Cysteine is the most intrinsically nucleophilic amino acid in proteins, where its reactivity is tuned to perform diverse biochem. functions. The absence of a consensus sequence that defines functional cysteines in proteins has hindered their discovery and characterization. Here we describe a proteomics method to profile quant. the intrinsic reactivity of cysteine residues en masse directly in native biol. systems. Hyper-reactivity was a rare feature among cysteines and it was found to specify a wide range of activities, including nucleophilic and reductive catalysis and sites of oxidative modification. Hyper-reactive cysteines were identified in several proteins of uncharacterized function, including a residue conserved across eukaryotic phylogeny that we show is required for yeast viability and is involved in iron-sulfur protein biogenesis. We also demonstrate that quant. reactivity profiling can form the basis for screening and functional assignment of cysteines in computationally designed proteins, where it discriminated catalytically active from inactive cysteine hydrolase designs.
- 52Backus, 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), 570– 574, DOI: 10.1038/nature1800252https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtVSksbnN&md5=6fab087c042de735f8a5931e9f5b946cProteome-wide covalent ligand discovery in native biological systemsBackus, Keriann M.; Correia, Bruno E.; Lum, Kenneth M.; Forli, Stefano; Horning, Benjamin D.; Gonzalez-Paez, Gonzalo E.; Chatterjee, Sandip; Lanning, Bryan R.; Teijaro, John R.; Olson, Arthur J.; Wolan, Dennis W.; Cravatt, Benjamin F.Nature (London, United Kingdom) (2016), 534 (7608), 570-574CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Small mols. are powerful tools for investigating protein function and can serve as leads for new therapeutics. Most human proteins, however, lack small-mol. ligands, and entire protein classes are considered 'undruggable'. Fragment-based ligand discovery can identify small-mol. probes for proteins that have proven difficult to target using high-throughput screening of complex compd. libraries. Although reversibly binding ligands are commonly pursued, covalent fragments provide an alternative route to small-mol. probes, including those that can access regions of proteins that are difficult to target through binding affinity alone. Here we report a quant. anal. of cysteine-reactive small-mol. fragments screened against thousands of proteins in human proteomes and cells. Covalent ligands were identified for >700 cysteines found in both druggable proteins and proteins deficient in chem. probes, including transcription factors, adaptor/scaffolding proteins, and uncharacterized proteins. Among the atypical ligand-protein interactions discovered were compds. that react preferentially with pro- (inactive) caspases. We used these ligands to distinguish extrinsic apoptosis pathways in human cell lines vs. primary human T cells, showing that the former is largely mediated by caspase-8 while the latter depends on both caspase-8 and -10. Fragment-based covalent ligand discovery provides a greatly expanded portrait of the ligandable proteome and furnishes compds. that can illuminate protein functions in native biol. systems.
- 53Abbasov, 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), 1081– 1092, DOI: 10.1038/s41557-021-00765-453https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvF2ju7rJ&md5=90e566b7a65ebfca5108a1c44f06f575A proteome-wide atlas of lysine-reactive chemistryAbbasov, Mikail E.; Kavanagh, Madeline E.; Ichu, Taka-Aki; Lazear, Michael R.; Tao, Yongfeng; Crowley, Vincent M.; am Ende, Christopher W.; Hacker, Stephan M.; Ho, Jordan; Dix, Melissa M.; Suciu, Radu; Hayward, Matthew M.; Kiessling, Laura L.; Cravatt, Benjamin F.Nature Chemistry (2021), 13 (11), 1081-1092CODEN: NCAHBB; ISSN:1755-4330. (Nature Portfolio)Recent advances in chem. proteomics have begun to characterize the reactivity and ligandability of lysines on a global scale. Yet, only a limited diversity of aminophilic electrophiles have been evaluated for interactions with the lysine proteome. Here, we report an in-depth profiling of >30 uncharted aminophilic chemotypes that greatly expands the content of ligandable lysines in human proteins. Aminophilic electrophiles showed disparate proteomic reactivities that range from selective interactions with a handful of lysines to, for a set of dicarboxaldehyde fragments, remarkably broad engagement of the covalent small-mol.-lysine interactions captured by the entire library. We used these latter 'scout' electrophiles to efficiently map ligandable lysines in primary human immune cells under stimulatory conditions. Finally, we show that aminophilic compds. perturb diverse biochem. functions through site-selective modification of lysines in proteins, including protein-RNA interactions implicated in innate immune responses. These findings support the broad potential of covalent chem. for targeting functional lysines in the human proteome.
- 54Hayes, J. D.; Flanagan, J. U.; Jowsey, I. R. Glutathione Transferases. Annu. Rev. Pharmacol. Toxicol. 2005, 45, 51– 88, DOI: 10.1146/annurev.pharmtox.45.120403.09585754https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXisVWjtrk%253D&md5=c74f17b1381f046770a7f6faf5c51291Glutathione transferasesHayes, John D.; Flanagan, Jack U.; Jowsey, Ian R.Annual Review of Pharmacology and Toxicology (2005), 45 (), 51-88, 1 plateCODEN: ARPTDI; ISSN:0362-1642. (Annual Reviews Inc.)A review. The authors describe the 3 mammalian glutathione S-transferase (GST) families, namely cytosolic, mitochondrial, and microsomal GSTs, the latter now designated MAPEG (Membrane-Assocd. Proteins in Eicosanoid and Glutathione metab.). In addn. to detoxifying electrophilic xenobiotics, such as chem. carcinogens, environmental pollutants, and antitumor agents, these GSTs inactivate endogenous α,β-unsatd. aldehydes, quinones, epoxides, and hydroperoxides formed as secondary metabolites during oxidative stress. These enzymes are also intimately involved in the biosynthesis of leukotrienes, prostaglandins, testosterone, and progesterone, as well as the degrdn. of tyrosine. Among their substrates, GSTs conjugate the signaling mols., 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) and 4-hydroxynonenal, with glutathione, and consequently they antagonize expression of genes trans-activated by peroxisome proliferator-activated receptor γ (PPARγ) and nuclear factor-erythroid 2 p45-related factor 2 (Nrf2). Through metab. of 15d-PGJ2, GST may enhance gene expression driven by nuclear factor-κB (NF-κB). Cytosolic human GST exhibit genetic polymorphisms and this variation can increase susceptibility to carcinogenesis and inflammatory disease. Polymorphisms in human MAPEG are assocd. with alterations in lung function and increased risk of myocardial infarction and stroke. Targeted disruption of murine genes has demonstrated that cytosolic GST isoenzymes are broadly cytoprotective, whereas MAPEG proteins have pro-inflammatory activities. Furthermore, knockout of mouse GSTA4 and GSTZ1 leads to overexpression of transferases in the Alpha, Mu, and Pi classes, an observation suggesting they are part of an adaptive mechanism that responds to endogenous chem. cues such as 4-hydroxynonenal and tyrosine degrdn. products. Consistent with this hypothesis, the promoters of cytosolic GST and MAPEG genes contain antioxidant response elements through which they are transcriptionally activated during exposure to Michael reaction acceptors and oxidative stress.
- 55Jiang, 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.abd9847There is no corresponding record for this reference.
- 56Jeffries, 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/ijms23094733There is no corresponding record for this reference.
- 57Quanrud, 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), 1661– 1676, DOI: 10.1021/acschembio.3c00338There is no corresponding record for this reference.
- 58Julio, 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 .There is no corresponding record for this reference.
- 59Adair, 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.202000267There is no corresponding record for this reference.
- 60Weltzien, H. U.; Padovan, E. Molecular Features of Penicillin Allergy. J. Invest. Dermatol. 1998, 110 (3), 203– 206, DOI: 10.1046/j.1523-1747.1998.00122.xThere is no corresponding record for this reference.
- 61Pirmohamed, 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), 236– 244, DOI: 10.1016/j.jaci.2015.06.022There is no corresponding record for this reference.
- 62Agashe, R. P.; Lippman, S. M.; Kurzrock, R. JAK: Not Just Another Kinase. Mol. Cancer Ther. 2022, 21 (12), 1757– 1764, DOI: 10.1158/1535-7163.MCT-22-0323There is no corresponding record for this reference.
- 63Goedken, 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), 4573– 4589, DOI: 10.1074/jbc.M114.59518163https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXjsF2lsLY%253D&md5=4d99cf5b13fd05df5c4abc197c1fe284Tricyclic Covalent Inhibitors Selectively Target Jak3 through an Active Site ThiolGoedken, Eric R.; Argiriadi, Maria A.; Banach, David L.; Fiamengo, Bryan A.; Foley, Sage E.; Frank, Kristine E.; George, Jonathan S.; Harris, Christopher M.; Hobson, Adrian D.; Ihle, David C.; Marcotte, Douglas; Merta, Philip J.; Michalak, Mark E.; Murdock, Sara E.; Tomlinson, Medha J.; Voss, Jeffrey W.Journal of Biological Chemistry (2015), 290 (8), 4573-4589CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The action of Janus kinases (JAKs) is required for multiple cytokine signaling pathways, and as such, JAK inhibitors hold promise for treatment of autoimmune disorders, including rheumatoid arthritis, inflammatory bowel disease, and psoriasis. However, due to high similarity in the active sites of the four members (Jak1, Jak2, Jak3, and Tyk2), developing selective inhibitors within this family is challenging. We have designed and characterized substituted, tricyclic Jak3 inhibitors that selectively avoid inhibition of the other JAKs. This is accomplished through a covalent interaction between an inhibitor contg. a terminal electrophile and an active site cysteine (Cys-909). We found that these ATP competitive compds. are irreversible inhibitors of Jak3 enzyme activity in vitro. They possess high selectivity against other kinases and can potently (IC50 < 100 nm) inhibit Jak3 activity in cell-based assays. These results suggest irreversible inhibitors of this class may be useful selective agents, both as tools to probe Jak3 biol. and potentially as therapies for autoimmune diseases.
- 64Chen, 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), 225– 242, DOI: 10.1080/13543776.2022.2023129There is no corresponding record for this reference.
- 65Shindo, 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.2021.11638665https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVSrtrjL&md5=d746f5fd8ac5778e1b23e8ef678ceec6Recent progress in covalent warheads for in vivo targeting of endogenous proteinsShindo, Naoya; Ojida, AkioBioorganic & Medicinal Chemistry (2021), 47 (), 116386CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Covalent drugs exert potent and durable activity by chem. modification of the endogenous target protein in vivo. To maximize the pharmacol. efficacy while alleviating the risk of toxicity due to nonspecific off-target reactions, current covalent drug discovery focuses on the development of targeted covalent inhibitors (TCIs), wherein a reactive group (warhead) is strategically incorporated onto a reversible ligand of the target protein to facilitate specific covalent engagement. Various aspects of warheads, such as intrinsic reactivity, chemoselectivity, mode of reaction, and reversibility of the covalent engagement, would affect the target selectivity of TCIs. Although TCIs clin. approved to date largely rely on Michael acceptor-type electrophiles for cysteine targeting, a wide array of novel warheads have been devised and tested in TCI development in recent years. In this short review, we provide an overview of recent progress in chem. for selective covalent targeting of proteins and their applications in TCI designs.
- 66Forster, 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), 5350– 5366, DOI: 10.1021/acs.jmedchem.8b0057166https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVGjsrbN&md5=8acdec137df746f854bbee75a10d5f70Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4-d]pyrrolo[2,3-b]pyridine ScaffoldForster, Michael; Chaikuad, Apirat; Dimitrov, Teodor; Doering, Eva; Holstein, Julia; Berger, Benedict-Tilman; Gehringer, Matthias; Ghoreschi, Kamran; Mueller, Susanne; Knapp, Stefan; Laufer, Stefan A.Journal of Medicinal Chemistry (2018), 61 (12), 5350-5366CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Janus kinases are major drivers of immune signaling and have been the focus of anti-inflammatory drug discovery for more than a decade. Because of the invariable colocalization of JAK1 and JAK3 at cytokine receptors, the question if selective JAK3 inhibition is sufficient to effectively block downstream signaling has been highly controversial. Recently, we discovered the covalent-reversible JAK3 inhibitor FM-381 (23) featuring high isoform and kinome selectivity. Crystallog. revealed that this inhibitor induces an unprecedented binding pocket by interactions of a nitrile substituent with arginine residues in JAK3. Herein, we describe detailed structure-activity relationships necessary for induction of the arginine pocket and the impact of this structural change on potency, isoform selectivity, and efficacy in cellular models. Furthermore, we evaluated the stability of this novel inhibitor class in in vitro metabolic assays and were able to demonstrate an adequate stability of key compd. 23 for in vivo use.
- 67Laux, 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), 573– 602, DOI: 10.1021/acsptsci.2c0005467https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xhsl2nur7M&md5=31a71f1a28ac2893f116b25e3972fc36Pharmacokinetic Optimization of Small Molecule Janus Kinase 3 Inhibitors to Target Immune CellsLaux, Julian; Forster, Michael; Riexinger, Laura; Schwamborn, Anna; Guezguez, Jamil; Pokoj, Christina; Kudolo, Mark; Berger, Lena M.; Knapp, Stefan; Schollmeyer, Dieter; Guse, Jan; Burnet, Michael; Laufer, Stefan A.ACS Pharmacology & Translational Science (2022), 5 (8), 573-602CODEN: APTSFN; ISSN:2575-9108. (American Chemical Society)Modulation of Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling is a promising method of treating autoimmune diseases, and the profound potency of clin. compds. makes this mode of action particularly attractive. Other questions that remain unanswered also include: What is the ideal selectivity between JAK1 and JAK3. Which cells are most relevant to JAK blockade. And what is the ideal tissue distribution pattern for addressing specific autoimmune conditions. We hypothesized that JAK3 selectivity is most relevant to low-dose clin. effects and interleukin-10 (IL-10) stimulation in particular, that immune cells are the most important compartment, and that distribution to inflamed tissue is the most important pharmacokinetic characteristic for in vivo disease modification. To test these hypotheses, we prepd. modified derivs. of JAK3 specific inhibitors that target C909 near the ATP binding site based on FM-381, first reported in 2016; a compd. class that was hitherto limited in uptake and exposure in vivo. These limits appear to be due to metabolic instability of side groups binding in the selectivity pocket. We identified derivs. with improved stability and tissue exposure. Conjugation to macrolide scaffolds with medium chain linkers was sufficient to stabilize the compds. and improve transport to organs while maintaining JAK3 affinity. These conjugates are inflammation targeted JAK3 inhibitors with long tissue half-lives and high exposure to activated immune cells.
- 68Burger, J. A. Bruton Tyrosine Kinase Inhibitors: Present and Future. Cancer J. 2019, 25 (6), 386– 393, DOI: 10.1097/PPO.000000000000041268https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlOms7zM&md5=574551611f4a8f843cf1ce2cd0db1697Bruton Tyrosine Kinase Inhibitors: Present and FutureBurger, Jan A.Cancer Journal (Philadelphia, PA, United States) (2019), 25 (6), 386-393CODEN: CAJOCB; ISSN:1528-9117. (Lippincott Williams & Wilkins)A review. Bruton tyrosine kinase (BTK) is a nonreceptor tyrosine kinase that plays a central role in the signal transduction of the B-cell antigen receptor and other cell surface receptors, both in normal and malignant B lymphocytes. B-cell antigen receptor signaling is activated in secondary lymphatic organs and drives the proliferation of malignant B cells, including chronic lymphocytic leukemia (CLL) cells. During the last 10 years, BTK inhibitors (BTKis) are increasingly replacing chemotherapy-based regimen, esp. in patients with CLL and mantle cell lymphoma (MCL). Bruton tyrosine kinase inhibitors are particularly active in patients with CLL and MCL, but also received approval for Waldenstroem macroglobulinemia, small lymphocytic lymphoma, marginal zone lymphoma, and chronic graft-vs.-host disease. Current clin. practice is continuous long-term administration of BTKi, which can be complicated by adverse effects or the development of drug resistance. Alternatives to long-term use of BTKi are being developed, such as combination therapies, permitting for limited duration therapy. Second-generation BTKis are under development, which differ from ibrutinib, the first-in-class BTKi, in their specificity for BTK, and therefore may differentiate themselves from ibrutinib in terms of adverse effects or efficacy.
- 69Khan, W. N. Regulation of B Lymphocyte Development and Activation by Bruton’s Tyrosine Kinase. Immunol. Res. 2001, 23 (2–3), 147– 156, DOI: 10.1385/IR:23:2-3:147There is no corresponding record for this reference.
- 70Guldenpfennig, C.; Teixeiro, E.; Daniels, M. NF-kB’s Contribution to B Cell Fate Decisions. Front. Immunol. 2023, 14, 1214095, DOI: 10.3389/fimmu.2023.1214095There is no corresponding record for this reference.
- 71Staudt, L. M. Oncogenic Activation of NF-kappaB. Cold Spring Harb. Perspect. Biol. 2010, 2 (6), a000109, DOI: 10.1101/cshperspect.a000109There is no corresponding record for this reference.
- 72Small 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 .There is no corresponding record for this reference.
- 73Wu, 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/s13045-016-0250-973https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXis1eisrw%253D&md5=a6a8f9b9236abe7ef1054a41e1c71e5aAcalabrutinib (ACP-196): a selective second-generation BTK inhibitorWu, Jingjing; Zhang, Mingzhi; Liu, DelongJournal of Hematology & Oncology (2016), 9 (), 21/1-21/4CODEN: JHOOAO; ISSN:1756-8722. (BioMed Central Ltd.)More and more targeted agents become available for B cell malignancies with increasing precision and potency. The first-in-class Bruton's tyrosine kinase (BTK) inhibitor, ibrutinib, has been in clin. use for the treatment of chronic lymphocytic leukemia, mantle cell lymphoma, and Waldenstrom's macroglobulinemia. More selective BTK inhibitors (ACP-196, ONO/GS-4059, BGB-3111, CC-292) are being explored. Acalabrutinib (ACP-196) is a novel irreversible second-generation BTK inhibitor that was shown to be more potent and selective than ibrutinib. This review summarized the preclin. research and clin. data of acalabrutinib.
- 74Guo, 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), 7923– 7940, DOI: 10.1021/acs.jmedchem.9b0068774https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsFSku7bP&md5=5cdb551d296e60eb1e82b03b7a0384eeDiscovery of Zanubrutinib (BGB-3111), a Novel, Potent, and Selective Covalent Inhibitor of Bruton's Tyrosine KinaseGuo, Yunhang; Liu, Ye; Hu, Nan; Yu, Desheng; Zhou, Changyou; Shi, Gongyin; Zhang, Bo; Wei, Min; Liu, Junhua; Luo, Lusong; Tang, Zhiyu; Song, Huipeng; Guo, Yin; Liu, Xuesong; Su, Dan; Zhang, Shuo; Song, Xiaomin; Zhou, Xing; Hong, Yuan; Chen, Shuaishuai; Cheng, Zhenzhen; Young, Steve; Wei, Qiang; Wang, Haisheng; Wang, Qiuwen; Lv, Lei; Wang, Fan; Xu, Haipeng; Sun, Hanzi; Xing, Haimei; Li, Na; Zhang, Wei; Wang, Zhongbo; Liu, Guodong; Sun, Zhijian; Zhou, Dongping; Li, Wei; Liu, Libin; Wang, Lai; Wang, ZhiweiJournal of Medicinal Chemistry (2019), 62 (17), 7923-7940CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Aberrant activation of Bruton's tyrosine kinase (BTK) plays an important role in pathogenesis of B-cell lymphomas, suggesting that inhibition of BTK is useful in the treatment of hematol. malignancies. The discovery of a more selective on-target covalent BTK inhibitor is of high value. Herein, we disclose the discovery and preclin. characterization of a potent, selective, and irreversible BTK inhibitor as our clin. candidate by using in vitro potency, selectivity, pharmacokinetics (PK), and in vivo pharmacodynamic for prioritizing compds. Compd. BGB-3111 (31a, Zanubrutinib) demonstrates (i) potent activity against BTK and excellent selectivity over other TEC, EGFR and Src family kinases, (ii) desirable ADME, excellent in vivo pharmacodynamic in mice and efficacy in OCI-LY10 xenograft models.
- 75Liclican, 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.2020.129531There is no corresponding record for this reference.
- 76Tam, 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-xThere is no corresponding record for this reference.
- 77Estupiñán, 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.2021.63094277https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3sfjtlCrtg%253D%253D&md5=c559e266e46358b280c7835a9574d965Comparative Analysis of BTK Inhibitors and Mechanisms Underlying Adverse EffectsEstupinan H Yesid; Berglof Anna; Zain Rula; Smith C I Edvard; Estupinan H Yesid; Zain RulaFrontiers in cell and developmental biology (2021), 9 (), 630942 ISSN:2296-634X.The cytoplasmic protein-tyrosine kinase BTK plays an essential role for differentiation and survival of B-lineage cells and, hence, represents a suitable drug target. The number of BTK inhibitors (BTKis) in the clinic has increased considerably and currently amounts to at least 22. First-in-class was ibrutinib, an irreversible binder forming a covalent bond to a cysteine in the catalytic region of the kinase, for which we have identified 228 active trials listed at ClinicalTrials.gov. Next-generation inhibitors, acalabrutinib and zanubrutinib, are approved both in the United States and in Europe, and zanubrutinib also in China, while tirabrutinib is currently only registered in Japan. In most cases, these compounds have been used for the treatment of B-lymphocyte tumors. However, an increasing number of trials instead addresses autoimmunity and inflammation in multiple sclerosis, rheumatoid arthritis, pemphigus and systemic lupus erythematosus with the use of either irreversibly binding inhibitors, e.g., evobrutinib and tolebrutinib, or reversibly binding inhibitors, like fenebrutinib. Adverse effects (AEs) have predominantly implicated inhibition of other kinases with a BTKi-binding cysteine in their catalytic domain. Analysis of the reported AEs suggests that ibrutinib-associated atrial fibrillation is caused by binding to ERBB2/HER2 and ERBB4/HER4. However, the binding pattern of BTKis to various additional kinases does not correlate with the common assumption that skin manifestations and diarrhoeas are off-target effects related to EGF receptor inhibition. Moreover, dermatological toxicities, diarrhoea, bleedings and invasive fungal infections often develop early after BTKi treatment initiation and subsequently subside. Conversely, cardiovascular AEs, like hypertension and various forms of heart disease, often persist.
- 78Mato, 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), 874– 879, DOI: 10.3324/haematol.2017.182907There is no corresponding record for this reference.
- 79Sharman, 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_1.4060.4060There is no corresponding record for this reference.
- 80Winqvist, 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), e208– e210, DOI: 10.3324/haematol.2018.198820There is no corresponding record for this reference.
- 81ClinicalTrials.gov. Rilzabrutinib. https://clinicaltrials.gov/search?intr=Rilzabrutinib (accessed 2023–11–28).There is no corresponding record for this reference.
- 82Oda, K. New Families of Carboxyl Peptidases: Serine-Carboxyl Peptidases and Glutamic Peptidases. J. Biochem. (Tokyo) 2012, 151 (1), 13– 25, DOI: 10.1093/jb/mvr129There is no corresponding record for this reference.
- 83Ćwilichowska, 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.2022.101144There is no corresponding record for this reference.
- 84Manasanch, E. E.; Orlowski, R. Z. Proteasome Inhibitors in Cancer Therapy. Nat. Rev. Clin. Oncol. 2017, 14 (7), 417– 433, DOI: 10.1038/nrclinonc.2016.20684https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsFGlsbg%253D&md5=7246069f357a4cc9449f822c57767811Proteasome inhibitors in cancer therapyManasanch, Elisabet E.; Orlowski, Robert Z.Nature Reviews Clinical Oncology (2017), 14 (7), 417-433CODEN: NRCOAA; ISSN:1759-4774. (Nature Publishing Group)The ubiquitin proteasome pathway was discovered in the 1980s to be a central component of the cellular protein-degrdn. machinery with essential functions in homeostasis, which include preventing the accumulation of misfolded or deleterious proteins. Cancer cells produce proteins that promote both cell survival and proliferation, and/or inhibit mechanisms of cell death. This notion set the stage for preclin. testing of proteasome inhibitors as a means to shift this fine equil. towards cell death. Since the late 1990s, clin. trials have been conducted for a variety of malignancies, leading to regulatory approvals of proteasome inhibitors to treat multiple myeloma and mantle-cell lymphoma. First-generation and second-generation proteasome inhibitors can elicit deep initial responses in patients with myeloma, for whom these drugs have dramatically improved outcomes, but relapses are frequent and acquired resistance to treatment eventually emerges. In addn., promising preclin. data obtained with proteasome inhibitors in models of solid tumors have not been confirmed in the clinic, indicating the importance of primary resistance. Investigation of the mechanisms of resistance is, therefore, essential to further maximize the utility of this class of drugs in the era of personalized medicine. Herein, we discuss the advances and challenges resulting from the introduction of proteasome inhibitors into the clinic.
- 85Flint, 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), 401– 411, DOI: 10.1128/AAC.01081-08There is no corresponding record for this reference.
- 86Augeri, 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), 5025– 5037, DOI: 10.1021/jm050261p86https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXlsVeltb0%253D&md5=878355880133bb0ddd6bd14ecbf1d070Discovery and Preclinical Profile of Saxagliptin (BMS-477118): A Highly Potent, Long-Acting, Orally Active Dipeptidyl Peptidase IV Inhibitor for the Treatment of Type 2 DiabetesAugeri, David J.; Robl, Jeffrey A.; Betebenner, David A.; Magnin, David R.; Khanna, Ashish; Robertson, James G.; Wang, Aiying; Simpkins, Ligaya M.; Taunk, Prakash; Huang, Qi; Han, Song-Ping; Abboa-Offei, Benoni; Cap, Michael; Xin, Li; Tao, Li; Tozzo, Effie; Welzel, Gustav E.; Egan, Donald M.; Marcinkeviciene, Jovita; Chang, Shu Y.; Biller, Scott A.; Kirby, Mark S.; Parker, Rex A.; Hamann, Lawrence G.Journal of Medicinal Chemistry (2005), 48 (15), 5025-5037CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Efforts to further elucidate structure-activity relationships (SAR) within the authors previously disclosed series of β-quaternary amino acid linked L-cis-4,5-methanoprolinenitrile dipeptidyl peptidase IV (DPP-IV) inhibitors led to the investigation of vinyl substitution at the β-position of α-cycloalkyl-substituted glycines. Despite poor systemic exposure, vinyl-substituted compds. showed extended duration of action in acute rat ex vivo plasma DPP-IV inhibition models. Oxygenated putative metabolites were prepd. and were shown to exhibit the potency and extended duration of action of their precursors in efficacy models measuring glucose clearance in Zuckerfa/fa rats. Extension of this approach to adamantylglycine-derived inhibitors led to the discovery of highly potent inhibitors, including hydroxyadamantyl compd. BMS-477118 (saxagliptin), a highly efficacious, stable, and long-acting DPP-IV inhibitor, which is currently undergoing clin. trials for treatment of type 2 diabetes.
- 87Lin, 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), 1813– 1822, DOI: 10.1128/AAC.50.5.1813-1822.2006There is no corresponding record for this reference.
- 88Focosi, 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.2022.106708There is no corresponding record for this reference.
- 89Wen, W.; Qi, Z.; Wang, J. The Function and Mechanism of Enterovirus 71 (EV71) 3C Protease. Curr. Microbiol. 2020, 77 (9), 1968– 1975, DOI: 10.1007/s00284-020-02082-4There is no corresponding record for this reference.
- 90Zeng, 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), 1762– 1766, DOI: 10.1016/j.bmcl.2016.02.039There is no corresponding record for this reference.
- 91Zhai, 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, 559– 573, DOI: 10.1016/j.ejmech.2016.08.06491https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsV2jurnN&md5=41cabd5d56ab0164e48cbd7806dbf2c7Structure-activity relationship study of peptidomimetic aldehydes as enterovirus 71 3C protease inhibitorsZhai, Yangyang; Ma, Yuying; Ma, Fei; Nie, Quandeng; Ren, Xuejiao; Wang, Yaxin; Shang, Luqing; Yin, ZhengEuropean Journal of Medicinal Chemistry (2016), 124 (), 559-573CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A series of peptidomimetic aldehydes were designed, synthesized, and evaluated for their biochem. activity against 3C protease (3Cpro) and anti-enterovirus 71 (EV71) activity in vitro. Mol. docking revealed that 5s (IC50 = 0.22 ± 0.07 μM, EC50 = 0.18 ± 0.05 μM) could bind well to the active site of EV71 3Cpro, which was consistent with the biol. data compared to ref. 5a (IC50 = 0.54 ± 0.02 μM, EC50 = 0.26 ± 0.07 μM). Structure and relationship study led to the discovery of aldehyde 5x (IC50 = 0.10 ± 0.02 μM, EC50 = 0.11 ± 0.07 μM), which exhibited the most potent 3Cpro inhibitory and antiviral activity.
- 92Luo, Y. L. Mechanism-Based and Computational-Driven Covalent Drug Design. J. Chem. Inf. Model. 2021, 61 (11), 5307– 5311, DOI: 10.1021/acs.jcim.1c01278There is no corresponding record for this reference.
- 93Mihalovits, 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), 6579– 6594, DOI: 10.1021/acs.jcim.0c0083493https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFSqu73N&md5=61ff455099a14db29ffd081cd3fd613eAffinity and Selectivity Assessment of Covalent Inhibitors by Free Energy CalculationsMihalovits, Levente M.; Ferenczy, Gyorgy G.; Keseru, Gyorgy M.Journal of Chemical Information and Modeling (2020), 60 (12), 6579-6594CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Covalent inhibitors have been gaining increased attention in drug discovery due to their beneficial properties such as long residence time, high biochem. efficiency, and specificity. Optimization of covalent inhibitors is a complex task that involves parallel monitoring of the noncovalent recognition elements and the covalent reactivity of the mols. to avoid potential idiosyncratic side effects. This challenge calls for special design protocols, including a variety of computational chem. methods. Covalent inhibition proceeds through multiple steps, and calcg. free energy changes of the subsequent binding events along the overall binding process would help us to better control the design of drug candidates. Inspired by the recent success of free energy calcns. on reversible binders, we developed a complex protocol to compute free energies related to the noncovalent and covalent binding steps with thermodn. integration and hybrid quantum mech./mol. mech. (QM/MM) potential of mean force (PMF) calcns., resp. In optimization settings, we examd. two therapeutically relevant proteins complexed with congeneric sets of irreversible cysteine targeting covalent inhibitors. In the selectivity paradigm, we studied the irreversible binding of covalent inhibitors to phylogenetically close targets by a mutational approach. The results of the calcns. are in good agreement with the exptl. free energy values derived from the inhibition and kinetic consts. (Ki and kinact) of the enzyme-inhibitor binding. The proposed method might be a powerful tool to predict the potency, selectivity, and binding mechanism of irreversible covalent inhibitors.
- 94Mihalovits, 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.26768There is no corresponding record for this reference.
- 95Lonsdale, 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), 3124– 3137, DOI: 10.1021/acs.jcim.7b0055395https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsl2gsLjF&md5=722e83699c58dee1009b87c8c31bdeccExpanding the Armory: Predicting and Tuning Covalent Warhead ReactivityLonsdale, Richard; Burgess, Jonathan; Colclough, Nicola; Davies, Nichola L.; Lenz, Eva M.; Orton, Alexandra L.; Ward, Richard A.Journal of Chemical Information and Modeling (2017), 57 (12), 3124-3137CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Targeted covalent inhibition is an established approach for increasing the potency and selectivity of potential drug candidates, as well as identifying potent and selective tool compds. for target validation studies. It is evident that identification of reversible recognition elements is essential for selective covalent inhibition, but this must also be achieved with the appropriate level of inherent reactivity of the reactive functionality (or "warhead"). Structural changes that increase or decrease warhead reactivity, guided by methods to predict the effect of those changes, have the potential to tune warhead reactivity and negate issues related to potency and/or toxicity. The half-life to adduct formation with glutathione (GSH t1/2) is a useful assay for measuring the reactivity of cysteine-targeting covalent warheads but is limited to synthesized mols. In this manuscript the authors assess the ability of several exptl. and computational approaches to predict GSH t1/2 for a range of cysteine targeting warheads, including a novel method based on pKa. Furthermore, matched mol. pairs anal. has been performed against the internal compd. collection, revealing structure-activity relationships between a selection of different covalent warheads. These observations and methods of prediction will be valuable in the design of new covalent inhibitors with desired levels of reactivity.
- 96Oballa, 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), 998– 1002, DOI: 10.1016/j.bmcl.2006.11.04496https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht12qtr4%253D&md5=b8fff00bdd6a746753f8f244d25b0cc8A generally applicable method for assessing the electrophilicity and reactivity of diverse nitrile-containing compoundsOballa, Renata M.; Truchon, Jean-Francois; Bayly, Christopher I.; Chauret, Nathalie; Day, Stephen; Crane, Sheldon; Berthelette, CarlBioorganic & Medicinal Chemistry Letters (2007), 17 (4), 998-1002CODEN: BMCLE8; ISSN:0960-894X. (Elsevier Ltd.)Nitrile-based inhibitors of cathepsin K have been known for some time and mechanism-of-action studies have demonstrated that cysteinyl proteases interact with nitriles in a reversible fashion. Three main classes of nitrile-contg. inhibitors have been published in the cathepsin K field: (i) cyanamides, (ii) arom. nitriles, and (iii) aminoacetonitriles. A computational approach was used to calc. the theor. reactivities of diverse nitriles and this was found to correlate with their extent of reactivity with free cysteine. Moreover, there is a tentative link between high reactivity with cysteine and the potential to lead to irreversible covalent binding to proteins.
- 97Copeland, 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), 730– 739, DOI: 10.1038/nrd208297https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XptVCltro%253D&md5=60ede2301584b10ac4e8fa18e1e6d107Drug-target residence time and its implications for lead optimizationCopeland, Robert A.; Pompliano, David L.; Meek, Thomas D.Nature Reviews Drug Discovery (2006), 5 (9), 730-739CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Much of drug discovery today is predicated on the concept of selective targeting of particular bioactive macromols. by low-mol.-mass drugs. The binding of drugs to their macromol. targets is therefore seen as paramount for pharmacol. activity. In vitro assessment of drug-target interactions is classically quantified in terms of binding parameters such as IC50 or Kd. This article presents an alternative perspective on drug optimization in terms of drug-target binary complex residence time, as quantified by the dissociative half-life of the drug-target binary complex. We describe the potential advantages of long residence time in terms of duration of pharmacol. effect and target selectivity.
- 98Bernetti, M.; Masetti, M.; Rocchia, W.; Cavalli, A. Kinetics of Drug Binding and Residence Time. Annu. Rev. Phys. Chem. 2019, 70, 143– 171, DOI: 10.1146/annurev-physchem-042018-05234098https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjsFSgsro%253D&md5=6016da9b20e539f43e627432018ae51bKinetics of Drug Binding and Residence TimeBernetti, Mattia; Masetti, Matteo; Rocchia, Walter; Cavalli, AndreaAnnual Review of Physical Chemistry (2019), 70 (), 143-171CODEN: ARPLAP; ISSN:0066-426X. (Annual Reviews)The kinetics of drug binding and unbinding is assuming an increasingly crucial role in the long, costly process of bringing a new medicine to patients. For example, the time a drug spends in contact with its biol. target is known as residence time (the inverse of the kinetic const. of the drug-target unbinding, 1/koff). Recent reports suggest that residence time could predict drug efficacy in vivo, perhaps even more effectively than conventional thermodn. parameters (free energy, enthalpy, entropy). There are many exptl. and computational methods for predicting drug-target residence time at an early stage of drug discovery programs. Here, we review and discuss the methodol. approaches to estg. drug binding kinetics and residence time. We first introduce the theor. background of drug binding kinetics from a physicochem. standpoint. We then analyze the recent literature in the field, starting from the exptl. methodologies and applications thereof and moving to theor. and computational approaches to the kinetics of drug binding and unbinding. We acknowledge the central role of mol. dynamics and related methods, which comprise a great no. of the computational methods and applications reviewed here. However, we also consider kinetic Monte Carlo. We conclude with the outlook that drug (un)binding kinetics may soon become a go/no go step in the discovery and development of new medicines.
- 99Ren, 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), 493– 510, DOI: 10.1007/s10928-022-09822-yThere is no corresponding record for this reference.
- 100Frühauf, A.; Wolff, B.; Schweipert, M.; Meyer-Almes, F.-J. Synthesis and Characterization of Reversible Covalent HDAC4 Inhibitors. Methods Mol. Biol. Clifton NJ. 2023, 2589, 207– 221, DOI: 10.1007/978-1-0716-2788-4_14There is no corresponding record for this reference.
- 101Forman, H. J.; Zhang, H.; Rinna, A. Glutathione: Overview of Its Protective Roles, Measurement, and Biosynthesis. Mol. Aspects Med. 2009, 30 (1–2), 1– 12, DOI: 10.1016/j.mam.2008.08.006101https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXltFelurY%253D&md5=c1b1aa2b03389bf1434af0762df78fe1Glutathione: Overview of its protective roles, measurement, and biosynthesisForman, Henry Jay; Zhang, Hongqiao; Rinna, AlessandraMolecular Aspects of Medicine (2009), 30 (1-2), 1-12CODEN: MAMED5; ISSN:0098-2997. (Elsevier B.V.)This review is the introduction to a special issue concerning, glutathione (GSH), the most abundant low mol. wt. thiol compd. synthesized in cells. GSH plays crit. roles in protecting cells from oxidative damage and the toxicity of xenobiotic electrophiles, and maintaining redox homeostasis. Here, the functions and GSH and the sources of oxidants and electrophiles, the elimination of oxidants by redn. and electrophiles by conjugation with GSH are briefly described. Methods of assessing GSH status in the cells are also described. GSH synthesis and its regulation are addressed along with therapeutic approaches for manipulating GSH content that have been proposed. The purpose here is to provide a brief overview of some of the important aspects of glutathione metab. as part of this special issue that will provide a more comprehensive review of the state of knowledge regarding this essential mol.
- 102Wu, G.; Fang, Y.-Z.; Yang, S.; Lupton, J. R.; Turner, N. D. Glutathione Metabolism and Its Implications for Health. J. Nutr. 2004, 134 (3), 489– 492, DOI: 10.1093/jn/134.3.489102https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXitlCrurc%253D&md5=98f0af4f642d488fbfa0b4f43632512cGlutathione metabolism and its implications for healthWu, Guoyao; Fang, Yun-Zhong; Yang, Sheng; Lupton, Joanne R.; Turner, Nancy D.Journal of Nutrition (2004), 134 (3), 489-492CODEN: JONUAI; ISSN:0022-3166. (American Society for Nutritional Sciences)A review. Glutathione (γ-glutamyl-cysteinyl-glycine; GSH) is the most abundant low-mol.-wt. thiol, and GSH/glutathione disulfide is the major redox couple in animal cells. The synthesis of GSH from glutamate, cysteine, and glycine is catalyzed sequentially by two cytosolic enzymes, γ-glutamylcysteine synthetase and GSH synthetase. Compelling evidence shows that GSH synthesis is regulated primarily by γ-glutamylcysteine synthetase activity, cysteine availability, and GSH feedback inhibition. Animal and human studies demonstrate that adequate protein nutrition is crucial for the maintenance of GSH homeostasis. In addn., enteral or parenteral cystine, methionine, N-acetyl-cysteine, and L-2-oxothiazolidine-4-carboxylate are effective precursors of cysteine for tissue GSH synthesis. Glutathione plays important roles in antioxidant defense, nutrient metab., and regulation of cellular events (including gene expression, DNA and protein synthesis, cell proliferation and apoptosis, signal transduction, cytokine prodn. and immune response, and protein glutathionylation). Glutathione deficiency contributes to oxidative stress, which plays a key role in aging and the pathogenesis of many diseases (including kwashiorkor, seizure, Alzheimer's disease, Parkinson's disease, liver disease, cystic fibrosis, sickle cell anemia, HIV, AIDS, cancer, heart attack, stroke, and diabetes). New knowledge of the nutritional regulation of GSH metab. is crit. for the development of effective strategies to improve health and to treat these diseases.
- 103Bansal, A.; Simon, M. C. Glutathione Metabolism in Cancer Progression and Treatment Resistance. J. Cell Biol. 2018, 217 (7), 2291– 2298, DOI: 10.1083/jcb.201804161103https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFGntb3N&md5=2d071bd5b51fa8628dd56a2ee683931dGlutathione metabolism in cancer progression and treatment resistanceBansal, Ankita; Simon, M. CelesteJournal of Cell Biology (2018), 217 (7), 2291-2298CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)Glutathione (GSH) is the most abundant antioxidant found in living organisms and has multiple functions, most of which maintain cellular redox homeostasis. GSH preserves sufficient levels of cysteine and detoxifies xenobiotics while also conferring therapeutic resistance to cancer cells. However, GSH metab. plays both beneficial and pathogenic roles in a variety of malignancies. It is crucial to the removal and detoxification of carcinogens, and alterations in this pathway can have a profound effect on cell survival. Excess GSH promotes tumor progression, where elevated levels correlate with increased metastasis. In this review, we discuss recent studies that focus on deciphering the role of GSH in tumor initiation and progression as well as mechanisms underlying how GSH imparts treatment resistance to growing cancers. Targeting GSH synthesis/utilization therefore represents a potential means of rendering tumor cells more susceptible to different treatment options such as chemotherapy and radiotherapy.
- 104Bajic, 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: 10.1155/2019/5028181There is no corresponding record for this reference.
- 105Fu, 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), 2891– 2919, DOI: 10.1038/s41596-020-0352-2105https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVWgt7bJ&md5=aebfe7aa84c68df1202c7f984b2672beA quantitative thiol reactivity profiling platform to analyze redox and electrophile reactive cysteine proteomesFu, Ling; Li, Zongmin; Liu, Keke; Tian, Caiping; He, Jixiang; He, Jingyang; He, Fuchu; Xu, Ping; Yang, JingNature Protocols (2020), 15 (9), 2891-2919CODEN: NPARDW; ISSN:1750-2799. (Nature Research)Cysteine is unique among all protein-coding amino acids, owing to its intrinsically high nucleophilicity. The cysteinyl thiol group can be covalently modified by a broad range of redox mechanisms or by various electrophiles derived from exogenous or endogenous sources. Measuring the response of protein cysteines to redox perturbation or electrophiles is crit. for understanding the underlying mechanisms involved. Activity-based protein profiling based on thiol-reactive probes has been the method of choice for such analyses. We therefore adapted this approach and developed a new chemoproteomic platform, termed 'QTRP' (quant. thiol reactivity profiling), that relies on the ability of a com. available thiol-reactive probe IPM (2-iodo-N-(prop-2-yn-1-yl)acetamide) to covalently label, enrich and quantify the reactive cysteinome in cells and tissues. Here, we provide a detailed and updated workflow of QTRP that includes procedures for (i) labeling of the reactive cysteinome from cell or tissue samples (e.g., control vs. treatment) with IPM, (ii) processing the protein samples into tryptic peptides and tagging the probe-modified peptides with isotopically labeled azido-biotin reagents contg. a photo-cleavable linker via click chem. reaction, (iii) capturing biotin-conjugated peptides with streptavidin beads, (iv) identifying and quantifying the photo-released peptides by mass spectrometry (MS)-based shotgun proteomics and (v) interpreting MS data by a streamlined informatic pipeline using a proteomics software, pFind 3, and an automatic post-processing algorithm. We also exemplified here how to use QTRP for mining H2O2-sensitive cysteines and for detg. the intrinsic reactivity of cysteines in a complex proteome. We anticipate that this protocol should find broad applications in redox biol., chem. biol. and the pharmaceutical industry. The protocol for sample prepn. takes 3 d, whereas MS measurements and data analyses require 75 min and <30 min, resp., per sample.
- 106Kuljanin, 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), 630– 641, DOI: 10.1038/s41587-020-00778-3106https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXkslSrug%253D%253D&md5=d256bc7ff87c75e4929fe4a6649242b4Reimagining high-throughput profiling of reactive cysteines for cell-based screening of large electrophile librariesKuljanin, Miljan; Mitchell, Dylan C.; Schweppe, Devin K.; Gikandi, Ajami S.; Nusinow, David P.; Bulloch, Nathan J.; Vinogradova, Ekaterina V.; Wilson, David L.; Kool, Eric T.; Mancias, Joseph D.; Cravatt, Benjamin F.; Gygi, Steven P.Nature Biotechnology (2021), 39 (5), 630-641CODEN: NABIF9; ISSN:1087-0156. (Nature Portfolio)Current methods used for measuring amino acid side-chain reactivity lack the throughput needed to screen large chem. libraries for interactions across the proteome. Here we redesigned the work flow for activity-based protein profiling of reactive cysteine residues by using a smaller desthiobiotin-based probe, sample multiplexing, reduced protein starting amts. and software to boost data acquisition in real time on the mass spectrometer. Our method, streamlined cysteine activity-based protein profiling (SLC-ABPP), achieved a 42-fold improvement in sample throughput, corresponding to profiling library members at a depth of >8,000 reactive cysteine sites at 18 min per compd. We applied it to identify proteome-wide targets of covalent inhibitors to mutant Kirsten rat sarcoma (KRAS)G12C and Bruton's tyrosine kinase (BTK). In addn., we created a resource of cysteine reactivity to 285 electrophiles in three human cell lines, which includes >20,000 cysteines from >6,000 proteins per line. The goal of proteome-wide profiling of cysteine reactivity across thousand-member libraries under several cellular contexts is now within reach.
- 107Yamamoto, 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), 1169– 1203, DOI: 10.1152/physrev.00023.2017107https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXkvFektrc%253D&md5=906947c8ad97c48f60e46bc44103664aThe KEAP1-NRF2 system: a thiol-based sensor-effector apparatus for maintaining redox homeostasisYamamoto, Masayuki; Kensler, Thomas W.; Motohashi, HozumiPhysiological Reviews (2018), 98 (3), 1169-1203CODEN: PHREA7; ISSN:1522-1210. (American Physiological Society)A review. The Kelch-like ECH-assocd. protein 1-NF-E2-related factor 2 (KEAP1-NRF2) system forms the major node of cellular and organismal defense against oxidative and electrophilic stresses of both exogenous and endogenous origins. KEAP1 acts as a cysteine thiol-rich sensor of redox insults, whereas NRF2 is a transcription factor that robustly transduces chem. signals to regulate a battery of cytoprotective genes. KEAP1 represses NRF2 activity under quiescent conditions, whereas NRF2 is liberated from KEAP1-mediated repression on exposure to stresses. The rapid inducibility of a response based on a derepression mechanism is an important feature of the KEAP1-NRF2 system. Recent studies have unveiled the complexities of the functional contributions of the KEAP1-NRF2 system and defined its broader involvement in biol. processes, including cell proliferation and differentiation, as well as cytoprotection. In this review, we describe historical milestones in the initial characterization of the KEAP1-NRF2 system and provide a comprehensive overview of the mol. mechanisms governing the functions of KEAP1 and NRF2, as well as their roles in physiol. and pathol. We also refer to the clin. significance of the KEAP1-NRF2 system as an important prophylactic and therapeutic target for various diseases, particularly aging-related disorders. We believe that controlled harnessing of the KEAP1-NRF2 system is a key to healthy aging and well-being in humans.
- 108Unoki, 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/ijms21020545There is no corresponding record for this reference.
- 109Backus, K. M.; Cao, J.; Maddox, S. M. Opportunities and Challenges for the Development of Covalent Chemical Immunomodulators. Bioorg. Med. Chem. 2019, 27 (15), 3421– 3439, DOI: 10.1016/j.bmc.2019.05.050109https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFOku73L&md5=fbec9cd36ed1c0f3e9dbacf4d613730bOpportunities and challenges for the development of covalent chemical immunomodulatorsBackus, Keriann M.; Cao, Jian; Maddox, Sean M.Bioorganic & Medicinal Chemistry (2019), 27 (15), 3421-3439CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A review. Compds. that react irreversibly with cysteines have reemerged as potent and selective tools for altering protein function, serving as chem. probes and even clin. approved drugs. The exquisite sensitivity of human immune cell signaling pathways to oxidative stress indicates the likely, yet still underexploited, general utility of covalent probes for selective chem. immunomodulation. Here, we provide an overview of immunomodulatory cysteines, including identification of electrophilic compds. available to label these residues. We focus our discussion on three protein classes essential for cell signaling, which span the 'druggability' spectrum from amenable to chem. probes (kinases), somewhat druggable (proteases), to inaccessible (phosphatases). Using existing inhibitors as a guide, we identify general strategies to guide the development of covalent probes for selected undruggable classes of proteins and propose the application of such compds. to alter immune cell functions.
- 110Lincoln, 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), 166– 174, DOI: 10.1021/acssensors.1c01940There is no corresponding record for this reference.
- 111Zheng, S.; Liu, G. Polymeric Emissive Materials Based on Dynamic Covalent Bonds. Mol. Basel Switz. 2022, 27 (19), 6635, DOI: 10.3390/molecules27196635There is no corresponding record for this reference.