Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
2.
Chembiochem ; 24(11): e202300116, 2023 06 01.
Article En | MEDLINE | ID: mdl-37069799

While vaccines and antivirals are now being deployed for the current SARS-CoV-2 pandemic, we require additional antiviral therapeutics to not only effectively combat SARS-CoV-2 and its variants, but also future coronaviruses. All coronaviruses have relatively similar genomes that provide a potential exploitable opening to develop antiviral therapies that will be effective against all coronaviruses. Among the various genes and proteins encoded by all coronaviruses, one particularly "druggable" or relatively easy-to-drug target is the coronavirus Main Protease (3CLpro or Mpro), an enzyme that is involved in cleaving a long peptide translated by the viral genome into its individual protein components that are then assembled into the virus to enable viral replication in the cell. Inhibiting Mpro with a small-molecule antiviral would effectively stop the ability of the virus to replicate, providing therapeutic benefit. In this study, we have utilized activity-based protein profiling (ABPP)-based chemoproteomic approaches to discover and further optimize cysteine-reactive pyrazoline-based covalent inhibitors for the SARS-CoV-2 Mpro. Structure-guided medicinal chemistry and modular synthesis of di- and tri-substituted pyrazolines bearing either chloroacetamide or vinyl sulfonamide cysteine-reactive warheads enabled the expedient exploration of structure-activity relationships (SAR), yielding nanomolar potency inhibitors against Mpro from not only SARS-CoV-2, but across many other coronaviruses. Our studies highlight promising chemical scaffolds that may contribute to future pan-coronavirus inhibitors.


COVID-19 , SARS-CoV-2 , Humans , Cysteine , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Molecular Docking Simulation
3.
Cell Chem Biol ; 29(10): 1470-1481.e31, 2022 10 20.
Article En | MEDLINE | ID: mdl-36070758

Targeted protein degradation (TPD) uses small molecules to recruit E3 ubiquitin ligases into the proximity of proteins of interest, inducing ubiquitination-dependent degradation. A major bottleneck in the TPD field is the lack of accessible E3 ligase ligands for developing degraders. To expand the E3 ligase toolbox, we sought to convert the Kelch-like ECH-associated protein 1 (KEAP1) inhibitor KI696 into a recruitment handle for several targets. While we were able to generate KEAP1-recruiting degraders of BET family and murine focal adhesion kinase (FAK), we discovered that the target scope of KEAP1 was narrow, as targets easily degraded using a cereblon (CRBN)-recruiting degrader were refractory to KEAP1-mediated degradation. Linking the KEAP1-binding ligand to a CRBN-binding ligand resulted in a molecule that induced degradation of KEAP1 but not CRBN. In sum, we characterize tool compounds to explore KEAP1-mediated ubiquitination and delineate the challenges of exploiting new E3 ligases for generating bivalent degraders.


NF-E2-Related Factor 2 , Ubiquitin-Protein Ligases , Mice , Animals , Ubiquitin-Protein Ligases/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Ligands , NF-E2-Related Factor 2/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Ubiquitins/metabolism
4.
Nat Rev Drug Discov ; 21(12): 881-898, 2022 12.
Article En | MEDLINE | ID: mdl-36008483

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 addition 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 programmes and how an evolving toolbox of covalent drug discovery techniques facilitates success in this field.


COVID-19 Drug Treatment , Protein Kinase Inhibitors , Humans , Drug Discovery/methods , Ligands , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/metabolism , SARS-CoV-2 , Structure-Activity Relationship
5.
Nat Chem Biol ; 18(4): 412-421, 2022 04.
Article En | MEDLINE | ID: mdl-35210618

Many diseases are driven by proteins that are aberrantly ubiquitinated and degraded. These diseases would be therapeutically benefited by targeted protein stabilization (TPS). Here we present deubiquitinase-targeting chimeras (DUBTACs), heterobifunctional small molecules consisting of a deubiquitinase recruiter linked to a protein-targeting ligand, to stabilize the levels of specific proteins degraded in a ubiquitin-dependent manner. Using chemoproteomic approaches, we discovered the covalent ligand EN523 that targets a non-catalytic allosteric cysteine C23 in the K48-ubiquitin-specific deubiquitinase OTUB1. We showed that a DUBTAC consisting of our EN523 OTUB1 recruiter linked to lumacaftor, a drug used to treat cystic fibrosis that binds ΔF508-cystic fibrosis transmembrane conductance regulator (CFTR), robustly stabilized ΔF508-CFTR protein levels, leading to improved chloride channel conductance in human cystic fibrosis bronchial epithelial cells. We also demonstrated stabilization of the tumor suppressor kinase WEE1 in hepatoma cells. Our study showcases covalent chemoproteomic approaches to develop new induced proximity-based therapeutic modalities and introduces the DUBTAC platform for TPS.


Cystic Fibrosis , Chimera/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/therapeutic use , Humans , Ligands , Ubiquitin/metabolism
6.
J Am Chem Soc ; 144(2): 701-708, 2022 01 19.
Article En | MEDLINE | ID: mdl-34994556

Proteolysis-targeting chimeras (PROTACs), heterobifunctional compounds that consist of protein-targeting ligands linked to an E3 ligase recruiter, have arisen as a powerful therapeutic modality for targeted protein degradation (TPD). Despite the popularity of TPD approaches in drug discovery, only a small number of E3 ligase recruiters are available for the >600 E3 ligases that exist in human cells. Here, we have discovered a cysteine-reactive covalent ligand, EN106, that targets FEM1B, an E3 ligase recently discovered as the critical component of the cellular response to reductive stress. By targeting C186 in FEM1B, EN106 disrupts recognition of the key reductive stress substrate of FEM1B, FNIP1. We further establish that EN106 can be used as a covalent recruiter for FEM1B in TPD applications by demonstrating that a PROTAC linking EN106 to the BET bromodomain inhibitor JQ1 or the kinase inhibitor dasatinib leads to the degradation of BRD4 and BCR-ABL, respectively. Our study showcases a covalent ligand that targets a natural E3 ligase-substrate binding site and highlights the utility of covalent ligand screening in expanding the arsenal of E3 ligase recruiters suitable for TPD applications.


Acetamides/chemistry , Cell Cycle Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligase Complexes/metabolism , Animals , Azepines/chemistry , Binding Sites , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line , Cysteine/chemistry , Dasatinib/chemistry , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Humans , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Kinase Inhibitors/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Triazoles/chemistry , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/genetics
7.
Angew Chem Int Ed Engl ; 60(29): 15905-15911, 2021 07 12.
Article En | MEDLINE | ID: mdl-33915015

Aberrant activation of FGFR signaling occurs in many cancers, and ATP-competitive FGFR inhibitors have received regulatory approval. Despite demonstrating clinical efficacy, these inhibitors exhibit dose-limiting toxicity, potentially due to a lack of selectivity amongst the FGFR family and are poorly tolerated. Here, we report the discovery and characterization of DGY-09-192, a bivalent degrader that couples the pan-FGFR inhibitor BGJ398 to a CRL2VHL E3 ligase recruiting ligand, which preferentially induces FGFR1&2 degradation while largely sparing FGFR3&4. DGY-09-192 exhibited two-digit nanomolar DC50 s for both wildtype FGFR2 and several FGFR2-fusions, resulting in degradation-dependent antiproliferative activity in representative gastric cancer and cholangiocarcinoma cells. Importantly, DGY-09-192 induced degradation of a clinically relevant FGFR2 fusion protein in a xenograft model. Taken together, we demonstrate that DGY-09-192 has potential as a prototype FGFR degrader.


Drug Discovery , Proteolysis/drug effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Cell Line, Tumor , Humans
8.
Cell Chem Biol ; 28(1): 4-13.e17, 2021 01 21.
Article En | MEDLINE | ID: mdl-32966806

MYC is a major oncogenic transcriptional driver of most human cancers that has remained intractable to direct targeting because much of MYC is intrinsically disordered. Here, we have performed a cysteine-reactive covalent ligand screen to identify compounds that could disrupt the binding of MYC to its DNA consensus sequence in vitro and also impair MYC transcriptional activity in situ in cells. We have identified a covalent ligand, EN4, that targets cysteine 171 of MYC within a predicted intrinsically disordered region of the protein. We show that EN4 directly targets MYC in cells, reduces MYC and MAX thermal stability, inhibits MYC transcriptional activity, downregulates multiple MYC transcriptional targets, and impairs tumorigenesis. We also show initial structure-activity relationships of EN4 and identify compounds that show improved potency. Overall, we identify a unique ligandable site within an intrinsically disordered region of MYC that leads to inhibition of MYC transcriptional activity.


Cysteine/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Cells, Cultured , Cysteine/metabolism , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Structure , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
9.
Biochem Biophys Res Commun ; 533(4): 1298-1302, 2020 12 17.
Article En | MEDLINE | ID: mdl-33046246

Reacted with methylglyoxal (MGO), murine Aß(1-40) (mAß) produced significantly less superoxide anion (O2•-) compared to human Aß(1-40) (hAß). The reactions of MGO with mAß(R13H), hAß(H13F), Nα-acetyl-l-lysine, and Nα-acetyl-l-arginine implied that the lack of His13 in mAß prohibits its Lys16 residue from reacting to produce cross-linked reaction products and O2•-. Our results suggest that murine brains are under less oxidative stress than human brains, which may be one of the reasons why rodents do not develop AD-like symptoms, and which provides further insight into a chemical mechanism for the development of AD in humans.


Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Pyruvaldehyde/chemistry , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Mice , Peptide Fragments/metabolism , Pyruvaldehyde/metabolism , Superoxides/metabolism
10.
Front Mol Biosci ; 7: 81, 2020.
Article En | MEDLINE | ID: mdl-32509799

Unregulated Src activity promotes malignant processes in cancer, but no Src-directed targeted therapies are used clinically, possibly because early Src inhibitors produce off-target effects leading to toxicity. Improved selective Src inhibitors may enable Src-directed therapies. Previously, we reported an irreversible Src inhibitor, DGY-06-116, based on the hybridization of dasatinib and a promiscuous covalent kinase probe SM1-71. Here, we report biochemical and biophysical characterization of this compound. An x-ray co-crystal structure of DGY-06-116: Src shows a covalent interaction with the kinase p-loop and occupancy of the back hydrophobic kinase pocket, explaining its high potency, and selectivity. However, a reversible analog also shows similar potency. Kinetic analysis shows a slow inactivation rate compared to other clinically approved covalent kinase inhibitors, consistent with a need for p-loop movement prior to covalent bond formation. Overall, these results suggest that a strong reversible interaction is required to allow sufficient time for the covalent reaction to occur. Further optimization of the covalent linker may improve the kinetics of covalent bond formation.

11.
J Med Chem ; 63(4): 1624-1641, 2020 02 27.
Article En | MEDLINE | ID: mdl-31935084

SRC is a major regulator of many signaling pathways and contributes to cancer development. However, development of a selective SRC inhibitor has been challenging, and FDA-approved SRC inhibitors, dasatinib and bosutinib, are multitargeted kinase inhibitors. Here, we describe our efforts to develop a selective SRC covalent inhibitor by targeting cysteine 277 on the P-loop of SRC. Using a promiscuous covalent kinase inhibitor (CKI) SM1-71 as a starting point, we developed covalent inhibitor 15a, which discriminates SRC from other covalent targets of SM1-71 including TAK1 and FGFR1. As an irreversible covalent inhibitor, compound 15a exhibited sustained inhibition of SRC signaling both in vitro and in vivo. Moreover, 15a exhibited potent antiproliferative effects in nonsmall cell lung cancer cell lines harboring SRC activation, thus providing evidence that this approach may be promising for further drug development efforts.


Anilides/pharmacology , Cysteine/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , src-Family Kinases/antagonists & inhibitors , AAA Domain , Amino Acid Sequence , Anilides/chemical synthesis , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Humans , Mice, Inbred C57BL , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/chemical synthesis , Signal Transduction/drug effects , Structure-Activity Relationship , src-Family Kinases/chemistry
12.
J Biol Inorg Chem ; 22(8): 1211-1222, 2017 Dec.
Article En | MEDLINE | ID: mdl-29038915

Extensive research has linked the amyloid-beta (Aß) peptide to neurological dysfunction in Alzheimer's disease (AD). Insoluble Aß plaques in the AD patient brain contain high concentrations of advanced glycation end-products (AGEs) as well as transition metal ions. This research elucidated the roles of Aß, sugars, and Cu2+ in the oxidative stress mechanism of AD at the molecular level. Mass spectral (MS) analysis of the reactions of Aß with two representative sugars, ribose-5-phosphate (R5P) and methylglyoxal (MG), revealed Lys-16 and Arg-5 as the primary glycation sites. Quantitative analysis of superoxide [Formula: see text] production by a cyt c assay showed that Lys-16 generated four times as much [Formula: see text] as Arg-5. Lys-16 and Arg-5 in Aß1-40 are both adjacent to histidine residues, which are suggested to catalyze glycation. Additionally, Lys-16 is close to the central hydrophobic core (Leu-17-Ala-21) and to His-13, both of which are known to lower the pKa of the residue, leading to increased deprotonation of the amine and an enhanced glycation reactivity compared to Arg-5. Gel electrophoresis results indicated that all three components of AD plaques-Aß1-40, sugars, and Cu2+-are necessary for DNA damage. It is concluded that the glycation of Aß1-40 with sugars generates significant amounts of [Formula: see text], owing to the rapid glycation of Lys-16 and Arg-5. In the presence of Cu2+, [Formula: see text] converts to hydroxyl radical (HO·), the source of oxidative stress in AD.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Arginine/metabolism , Copper/pharmacology , Lysine/metabolism , Oxidative Stress/drug effects , Amino Acid Sequence , Cytochromes c/metabolism , DNA Damage , Deoxyguanine Nucleotides/metabolism , Glycosylation/drug effects , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/metabolism , Models, Molecular , Oxidation-Reduction/drug effects , Protein Conformation
...