Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Cell Chem Biol ; 30(11): 1354-1365.e6, 2023 11 16.
Article En | MEDLINE | ID: mdl-37643616

RAF dimer inhibitors offer therapeutic potential in RAF- and RAS-driven cancers. The utility of such drugs is predicated on their capacity to occupy both RAF protomers in the RAS-RAF signaling complex. Here we describe a method to conditionally quantify drug-target occupancy at selected RAF protomers within an active RAS-RAF complex in cells. RAF target engagement can be measured in the presence or absence of any mutant KRAS allele, enabling the high-affinity state of RAF dimer inhibitors to be quantified in the cellular milieu. The intracellular protomer selectivity of clinical-stage type II RAF inhibitors revealed that ARAF protomer engagement, but not engagement of BRAF or CRAF, is commensurate with inhibition of MAPK signaling in various mutant RAS cell lines. Our results support a fundamental role for ARAF in mutant RAS signaling and reveal poor ARAF protomer vulnerability for a cohort of RAF inhibitors undergoing clinical evaluation.


Proto-Oncogene Proteins B-raf , Signal Transduction , Humans , Protein Subunits/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Mutation , MAP Kinase Signaling System
2.
Nat Chem Biol ; 19(2): 230-238, 2023 02.
Article En | MEDLINE | ID: mdl-36302899

Small-molecule tools have enabled mechanistic investigations and therapeutic targeting of the protein kinase-like (PKL) superfamily. However, such tools are still lacking for many PKL members, including the highly conserved and disease-related UbiB family. Here, we sought to develop and characterize an inhibitor for the archetypal UbiB member COQ8, whose function is essential for coenzyme Q (CoQ) biosynthesis. Guided by crystallography, activity assays and cellular CoQ measurements, we repurposed the 4-anilinoquinoline scaffold to selectively inhibit human COQ8A in cells. Our chemical tool promises to lend mechanistic insights into the activities of these widespread and understudied proteins and to offer potential therapeutic strategies for human diseases connected to their dysfunction.


Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Humans , Saccharomyces cerevisiae/metabolism , Ubiquinone/pharmacology , Ubiquinone/chemistry , Saccharomyces cerevisiae Proteins/metabolism
3.
Nat Chem Biol ; 18(6): 596-604, 2022 06.
Article En | MEDLINE | ID: mdl-35314814

Current small-molecule inhibitors of KRAS(G12C) bind irreversibly in the switch-II pocket (SII-P), exploiting the strong nucleophilicity of the acquired cysteine as well as the preponderance of the GDP-bound form of this mutant. Nevertheless, many oncogenic KRAS mutants lack these two features, and it remains unknown whether targeting the SII-P is a practical therapeutic approach for KRAS mutants beyond G12C. Here we use NMR spectroscopy and a cellular KRAS engagement assay to address this question by examining a collection of SII-P ligands from the literature and from our own laboratory. We show that the SII-Ps of many KRAS hotspot (G12, G13, Q61) mutants are accessible using noncovalent ligands, and that this accessibility is not necessarily coupled to the GDP state of KRAS. The results we describe here emphasize the SII-P as a privileged drug-binding site on KRAS and unveil new therapeutic opportunities in RAS-driven cancer.


Multiple Myeloma , Proto-Oncogene Proteins p21(ras) , Humans , Ligands , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
4.
J Med Chem ; 64(8): 4810-4840, 2021 04 22.
Article En | MEDLINE | ID: mdl-33830764

Histone deacetylase 6 (HDAC6) is a promising therapeutic target for the treatment of neurodegenerative disorders. SW-100 (1a), a phenylhydroxamate-based HDAC6 inhibitor (HDAC6i) bearing a tetrahydroquinoline (THQ) capping group, is a highly potent and selective HDAC6i that was shown to be effective in mouse models of Fragile X syndrome and Charcot-Marie-Tooth disease type 2A (CMT2A). In this study, we report the discovery of a new THQ-capped HDAC6i, termed SW-101 (1s), that possesses excellent HDAC6 potency and selectivity, together with markedly improved metabolic stability and druglike properties compared to SW-100 (1a). X-ray crystallography data reveal the molecular basis of HDAC6 inhibition by SW-101 (1s). Importantly, we demonstrate that SW-101 (1s) treatment elevates the impaired level of acetylated α-tubulin in the distal sciatic nerve, counteracts progressive motor dysfunction, and ameliorates neuropathic symptoms in a CMT2A mouse model bearing mutant MFN2. Taken together, these results bode well for the further development of SW-101 (1s) as a disease-modifying HDAC6i.


Charcot-Marie-Tooth Disease/drug therapy , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Quinolines/chemistry , Acetylation , Animals , Benzamides/chemistry , Benzamides/metabolism , Binding Sites , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Crystallography, X-Ray , Disease Models, Animal , Half-Life , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Phenotype , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Quinolines/metabolism , Quinolines/therapeutic use , Structure-Activity Relationship , Tubulin/metabolism
5.
Nat Commun ; 11(1): 2743, 2020 06 02.
Article En | MEDLINE | ID: mdl-32488087

Concerted multidisciplinary efforts have led to the development of Cyclin-Dependent Kinase inhibitors (CDKi's) as small molecule drugs and chemical probes of intracellular CDK function. However, conflicting data has been reported on the inhibitory potency of CDKi's and a systematic characterization of affinity and selectivity against intracellular CDKs is lacking. We have developed a panel of cell-permeable energy transfer probes to quantify target occupancy for all 21 human CDKs in live cells, and present a comprehensive evaluation of intracellular isozyme potency and selectivity for a collection of 46 clinically-advanced CDKi's and tool molecules. We observed unexpected intracellular activity profiles for a number of CDKi's, offering avenues for repurposing of highly potent molecules as probes for previously unreported targets. Overall, we provide a broadly applicable method for evaluating the selectivity of CDK inhibitors in living cells, and present a refined set of tool molecules to study CDK function.


Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor Proteins/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , CDC2 Protein Kinase , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase 9 , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Phosphorylation , Structure-Activity Relationship
6.
J Med Chem ; 62(18): 8557-8577, 2019 09 26.
Article En | MEDLINE | ID: mdl-31414801

Isoxazole is a five-membered heterocycle that is widely used in drug discovery endeavors. Here, we report the design, synthesis, and structural and biological characterization of SS-208, a novel HDAC6-selective inhibitor containing the isoxazole-3-hydroxamate moiety as a zinc-binding group as well as a hydrophobic linker. A crystal structure of the Danio rerio HDAC6/SS-208 complex reveals a bidentate coordination of the active-site zinc ion that differs from the preferred monodentate coordination observed for HDAC6 complexes with phenylhydroxamate-based inhibitors. While SS-208 has minimal effects on the viability of murine SM1 melanoma cells in vitro, it significantly reduced in vivo tumor growth in a murine SM1 syngeneic melanoma mouse model. These findings suggest that the antitumor activity of SS-208 is mainly mediated by immune-related antitumor activity as evidenced by the increased infiltration of CD8+ and NK+ T cells and the enhanced ratio of M1 and M2 macrophages in the tumor microenvironment.


Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Isoxazoles/pharmacology , Melanoma/drug therapy , Animals , CD8-Positive T-Lymphocytes/cytology , Catalytic Domain , Cell Line, Tumor , Drug Discovery , Histone Deacetylase Inhibitors/chemistry , Humans , Hydroxamic Acids/chemistry , Isoxazoles/chemistry , Macrophages/cytology , Mice , Microsomes/chemistry , Natural Killer T-Cells/cytology , Transplantation, Isogeneic , Zebrafish , Zinc/chemistry
7.
ACS Chem Neurosci ; 10(3): 1679-1695, 2019 03 20.
Article En | MEDLINE | ID: mdl-30511829

Disease-modifying therapies are needed for Fragile X Syndrome (FXS), as at present there are no effective treatments or cures. Herein, we report on a tetrahydroquinoline-based selective histone deacetylase 6 (HDAC6) inhibitor SW-100, its pharmacological and ADMET properties, and its ability to improve upon memory performance in a mouse model of FXS, Fmr1-/- mice. This small molecule demonstrates good brain penetrance, low-nanomolar potency for the inhibition of HDAC6 (IC50 = 2.3 nM), with at least a thousand-fold selectivity over all other class I, II, and IV HDAC isoforms. Moreover, through its inhibition of the α-tubulin deacetylase domain of HDAC6 (CD2), in cells SW-100 upregulates α-tubulin acetylation with no effect on histone acetylation and selectively restores the impaired acetylated α-tubulin levels in the hippocampus of Fmr1-/- mice. Lastly, SW-100 ameliorates several memory and learning impairments in Fmr1-/- mice, thus modeling the intellectual deficiencies associated with FXS, and hence providing a strong rationale for pursuing HDAC6-based therapies for the treatment of this rare disease.


Benzamides/pharmacology , Fragile X Syndrome/physiopathology , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Learning/drug effects , Memory/drug effects , Quinolines/pharmacology , Animals , Cognition/drug effects , Disease Models, Animal , Fragile X Syndrome/enzymology , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Protein Processing, Post-Translational/drug effects
8.
Cell Chem Biol ; 25(2): 206-214.e11, 2018 02 15.
Article En | MEDLINE | ID: mdl-29174542

For kinase inhibitors, intracellular target selectivity is fundamental to pharmacological mechanism. Although a number of acellular techniques have been developed to measure kinase binding or enzymatic inhibition, such approaches can fail to accurately predict engagement in cells. Here we report the application of an energy transfer technique that enabled the first broad-spectrum, equilibrium-based approach to quantitatively profile target occupancy and compound affinity in live cells. Using this method, we performed a selectivity profiling for clinically relevant kinase inhibitors against 178 full-length kinases, and a mechanistic interrogation of the potency offsets observed between cellular and biochemical analysis. For the multikinase inhibitor crizotinib, our approach accurately predicted cellular potency and revealed improved target selectivity compared with biochemical measurements. Due to cellular ATP, a number of putative crizotinib targets are unexpectedly disengaged in live cells at a clinically relevant drug dose.


Adenosine Triphosphate/metabolism , Phosphotransferases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cell Survival , Dose-Response Relationship, Drug , Energy Transfer , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Mass Spectrometry , Molecular Structure , Phosphotransferases/metabolism , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
9.
ACS Chem Biol ; 12(4): 1028-1037, 2017 04 21.
Article En | MEDLINE | ID: mdl-28195704

Novel engineered NanoLuc (Nluc) luciferase being smaller, brighter, and superior to traditional firefly (Fluc) or Renilla (Rluc) provides a great opportunity for the development of numerous biological, biomedical, clinical, and food and environmental safety applications. This new platform created an urgent need for Nluc inhibitors that could allow selective bioluminescent suppression and multiplexing compatibility with existing luminescence or fluorescence assays. Starting from thienopyrrole carboxylate 1, a hit from a 42 000 PubChem compound library with a low micromolar IC50 against Nluc, we derivatized four different structural fragments to discover a family of potent, single digit nanomolar, cell permeable inhibitors. Further elaboration revealed a channel that allowed access to the external Nluc surface, resulting in a series of highly potent cell impermeable Nluc inhibitors with negatively charged groups likely extending to the protein surface. The permeability was evaluated by comparing EC50 shifts calculated from both live and lysed cells expressing Nluc cytosolically. Luminescence imaging further confirmed that cell permeable compounds inhibit both intracellular and extracellular Nluc, whereas less permeable compounds differentially inhibit extracellular Nluc and Nluc on the cell surface. The compounds displayed little to no toxicity to cells and high luciferase specificity, showing no activity against firefly luciferase or even the closely related NanoBit system. Looking forward, the structural motifs used to gain access to the Nluc surface can also be appended with other functional groups, and therefore interesting opportunities for developing assays based on relief-of-inhibition can be envisioned.


Cell Membrane Permeability , Enzyme Inhibitors/pharmacology , Luciferases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Structure-Activity Relationship
10.
Nat Commun ; 6: 10091, 2015 Dec 03.
Article En | MEDLINE | ID: mdl-26631872

The therapeutic action of drugs is predicated on their physical engagement with cellular targets. Here we describe a broadly applicable method using bioluminescence resonance energy transfer (BRET) to reveal the binding characteristics of a drug with selected targets within intact cells. Cell-permeable fluorescent tracers are used in a competitive binding format to quantify drug engagement with the target proteins fused to Nanoluc luciferase. The approach enabled us to profile isozyme-specific engagement and binding kinetics for a panel of histone deacetylase (HDAC) inhibitors. Our analysis was directed particularly to the clinically approved prodrug FK228 (Istodax/Romidepsin) because of its unique and largely unexplained mechanism of sustained intracellular action. Analysis of the binding kinetics by BRET revealed remarkably long intracellular residence times for FK228 at HDAC1, explaining the protracted intracellular behaviour of this prodrug. Our results demonstrate a novel application of BRET for assessing target engagement within the complex milieu of the intracellular environment.


Cells/drug effects , Fluorescence Resonance Energy Transfer/methods , Histone Deacetylase Inhibitors/chemistry , Pharmaceutical Preparations/chemistry , Cell Proliferation , Cells/chemistry , Cells/cytology , HeLa Cells , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Luciferases/chemistry , Luciferases/genetics , Luciferases/metabolism , Luminescence
11.
J Neurosci Methods ; 228: 67-78, 2014 May 15.
Article En | MEDLINE | ID: mdl-24675049

BACKGROUND: Stem cell therapies appear promising for treating certain neurodegenerative disorders and molecular imaging methods that track these cells in vivo could answer some key questions regarding their survival and migration. Bioluminescence imaging (BLI), which relies on luciferase expression in these cells, has been used for this purpose due to its high sensitivity. NEW METHOD: In this study, we employ BLI to track luciferase-expressing human neural progenitor cells (hNPC(Luc2)) in the rat striatum long-term. RESULTS: We show that hNPC(Luc2) are detectable in the rat striatum. Furthermore, we demonstrate that using this tracking method, surviving grafts can be detected in vivo for up to 12 weeks, while those that were rejected do not produce bioluminescence signal. We also demonstrate the ability to discern hNPC(Luc2) contralateral migration. COMPARISON WITH EXISTING METHODS: Some of the advantages of BLI compared to other imaging methods used to track progenitor/stem cells include its sensitivity and specificity, low background signal and ability to distinguish surviving grafts from rejected ones over the long term while the blood-brain barrier remains intact. CONCLUSIONS: These new findings may be useful in future preclinical applications developing cell-based treatments for neurodegenerative disorders.


Corpus Striatum/cytology , Luminescent Measurements , Neural Stem Cells/physiology , Neuroimaging/methods , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Count , Cell Line, Transformed , Cell Movement , Corpus Striatum/surgery , Cyclosporine/pharmacology , Humans , Luciferases/genetics , Luciferases/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/surgery , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Stem Cell Transplantation , Time Factors , Transfection
...