Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Cell Chem Biol ; 29(10): 1470-1481.e31, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36070758

ABSTRACT

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.


Subject(s)
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
2.
Nat Cancer ; 3(5): 595-613, 2022 05.
Article in English | MEDLINE | ID: mdl-35534777

ABSTRACT

Acute myeloid leukemia (AML) remains difficult to treat and requires new therapeutic approaches. Potent inhibitors of the chromatin-associated protein MENIN have recently entered human clinical trials, opening new therapeutic opportunities for some genetic subtypes of this disease. Using genome-scale functional genetic screens, we identified IKAROS (encoded by IKZF1) as an essential transcription factor in KMT2A (MLL1)-rearranged (MLL-r) AML that maintains leukemogenic gene expression while also repressing pathways for tumor suppression, immune regulation and cellular differentiation. Furthermore, IKAROS displays an unexpected functional cooperativity and extensive chromatin co-occupancy with mixed lineage leukemia (MLL)1-MENIN and the regulator MEIS1 and an extensive hematopoietic transcriptional complex involving homeobox (HOX)A10, MEIS1 and IKAROS. This dependency could be therapeutically exploited by inducing IKAROS protein degradation with immunomodulatory imide drugs (IMiDs). Finally, we demonstrate that combined IKAROS degradation and MENIN inhibition effectively disrupts leukemogenic transcriptional networks, resulting in synergistic killing of leukemia cells and providing a paradigm for improved drug targeting of transcription and an opportunity for rapid clinical translation.


Subject(s)
Leukemia, Myeloid, Acute , Chromatin , Gene Expression , Humans , Ikaros Transcription Factor/metabolism , Leukemia, Myeloid, Acute/drug therapy , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Transcription Factors/genetics
3.
J Med Chem ; 64(15): 11637-11650, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34279939

ABSTRACT

Chemical biology tools to modulate protein levels in cells are critical to decipher complex biology. Targeted protein degradation offers the potential for rapid and dose-dependent protein depletion through the use of protein fusion tags toward which protein degraders have been established. Here, we present a newly developed protein degradation tag BRD4BD1L94V along with the corresponding cereblon (CRBN)-based heterobifunctional degrader based on a "bump-and-hole" approach. The resulting compound XY-06-007 shows a half-degradation concentration (DC50, 6 h) of 10 nM against BRD4BD1L94V with no degradation of off-targets, as assessed by whole proteome mass spectrometry, and demonstrates suitable pharmacokinetics for in vivo studies. We demonstrate that BRD4BD1L94V can be combined with the dTAG approach to achieve simultaneous degrader-mediated depletion of their respective protein fusions. This orthogonal system complements currently available protein degradation tags and enables investigation into the consequences resulting from rapid degradation of previously undruggable disease codependencies.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Design , Transcription Factors/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , Transcription Factors/metabolism
4.
Cell Chem Biol ; 28(10): 1514-1527.e4, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34314730

ABSTRACT

Targeted protein degradation refers to the use of small molecules that recruit a ubiquitin ligase to a target protein for ubiquitination and subsequent proteasome-dependent degradation. While degraders have been developed for many targets, key questions regarding degrader development and the consequences of acute pharmacological degradation remain, specifically for targets that exist in obligate multi-protein complexes. Here, we synthesize a pan-histone deacetylase (HDAC) degrader library for the chemo-proteomic exploration of acute degradation of a key class of chromatin-modifying enzymes. Using chemo-proteomics, we not only map the degradability of the zinc-dependent HDAC family identifying leads for targeting HDACs 1-8 and 10 but also explore important aspects of degrading epigenetic enzymes. We discover cell line-driven target specificity and that HDAC degradation often results in collateral loss of HDAC-containing repressive complexes. These findings potentially offer a new mechanism toward controlling chromatin structure, and our resource will facilitate accelerated degrader design and development for HDACs.


Subject(s)
Histone Deacetylases/metabolism , Small Molecule Libraries/metabolism , Cell Line , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylases/chemistry , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Proteolysis , Proteomics/methods , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
5.
Nat Chem Biol ; 17(6): 711-717, 2021 06.
Article in English | MEDLINE | ID: mdl-34035522

ABSTRACT

The zinc-finger transcription factor Helios is critical for maintaining the identity, anergic phenotype and suppressive activity of regulatory T (Treg) cells. While it is an attractive target to enhance the efficacy of currently approved immunotherapies, no existing approaches can directly modulate Helios activity or abundance. Here, we report the structure-guided development of small molecules that recruit the E3 ubiquitin ligase substrate receptor cereblon to Helios, thereby promoting its degradation. Pharmacological Helios degradation destabilized the anergic phenotype and reduced the suppressive activity of Treg cells, establishing a route towards Helios-targeting therapeutics. More generally, this study provides a framework for the development of small-molecule degraders for previously unligandable targets by reprogramming E3 ligase substrate specificity.


Subject(s)
DNA-Binding Proteins/drug effects , Ikaros Transcription Factor/drug effects , T-Lymphocytes, Regulatory/drug effects , Transcription Factors/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , DNA-Binding Proteins/genetics , Humans , Ikaros Transcription Factor/genetics , Jurkat Cells , Mice , Models, Molecular , Molecular Structure , Mutation/genetics , Small Molecule Libraries , Substrate Specificity , Transcription Factors/genetics , Ubiquitin-Protein Ligases/metabolism
6.
STAR Protoc ; 2(2): 100530, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34041503

ABSTRACT

Transcriptional changes happen within minutes; however, RNAi or genetic deletion requires days to weeks before transcription networks can be analyzed. This limitation has made it challenging to distinguish direct from indirect targets of sequence-specific transcription factors. This inability to define direct transcriptional targets hinders detailed studies of transcriptional mechanisms. This protocol combines rapid degradation of endogenous transcription factors with nascent transcript analysis to define the earliest, and likely direct, regulatory targets of transcription factors. For complete details on the use and execution of this protocol, please refer to Stengel et al., 2021).


Subject(s)
Transcription Factors , Transcription, Genetic , Animals , Cells, Cultured , RNA Interference , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Transcription Factors/analysis , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/physiology
7.
Cell ; 183(6): 1714-1731.e10, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33275901

ABSTRACT

Targeted protein degradation (TPD) refers to the use of small molecules to induce ubiquitin-dependent degradation of proteins. TPD is of interest in drug development, as it can address previously inaccessible targets. However, degrader discovery and optimization remains an inefficient process due to a lack of understanding of the relative importance of the key molecular events required to induce target degradation. Here, we use chemo-proteomics to annotate the degradable kinome. Our expansive dataset provides chemical leads for ∼200 kinases and demonstrates that the current practice of starting from the highest potency binder is an ineffective method for discovering active compounds. We develop multitargeted degraders to answer fundamental questions about the ubiquitin proteasome system, uncovering that kinase degradation is p97 dependent. This work will not only fuel kinase degrader discovery, but also provides a blueprint for evaluating targeted degradation across entire gene families to accelerate understanding of TPD beyond the kinome.


Subject(s)
Protein Kinases/metabolism , Proteolysis , Proteome/metabolism , Adult , Cell Line , Databases, Protein , Female , Humans , Male , Middle Aged , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/genetics , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/metabolism , Young Adult
8.
Nat Chem Biol ; 16(1): 7-14, 2020 01.
Article in English | MEDLINE | ID: mdl-31686031

ABSTRACT

The investigational drugs E7820, indisulam and tasisulam (aryl-sulfonamides) promote the degradation of the splicing factor RBM39 in a proteasome-dependent mechanism. While the activity critically depends on the cullin RING ligase substrate receptor DCAF15, the molecular details remain elusive. Here we present the cryo-EM structure of the DDB1-DCAF15-DDA1 core ligase complex bound to RBM39 and E7820 at a resolution of 4.4 Å, together with crystal structures of engineered subcomplexes. We show that DCAF15 adopts a new fold stabilized by DDA1, and that extensive protein-protein contacts between the ligase and substrate mitigate low affinity interactions between aryl-sulfonamides and DCAF15. Our data demonstrate how aryl-sulfonamides neo-functionalize a shallow, non-conserved pocket on DCAF15 to selectively bind and degrade RBM39 and the closely related splicing factor RBM23 without the requirement for a high-affinity ligand, which has broad implications for the de novo discovery of molecular glue degraders.


Subject(s)
Indoles/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Proteolysis/drug effects , RNA Recognition Motif Proteins/chemistry , Sulfonamides/chemistry , Amino Acid Motifs , Animals , Benzamides/chemistry , Benzamides/pharmacology , Cryoelectron Microscopy , Fluorescence Resonance Energy Transfer , Humans , Indoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Protein Binding , Protein Domains , Protein Interaction Mapping , Protein Structure, Secondary , RNA Recognition Motif Proteins/metabolism , RNA-Binding Proteins , Spodoptera , Sulfonamides/pharmacology , Ubiquitin-Protein Ligases/chemistry , Xenopus
9.
Cell Chem Biol ; 27(1): 57-65.e9, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31735695

ABSTRACT

The G1/S cell cycle checkpoint is frequently dysregulated in cancer, leaving cancer cells reliant on a functional G2/M checkpoint to prevent excessive DNA damage. Wee1 regulates the G2/M checkpoint by phosphorylating CDK1 at Tyr15 to prevent mitotic entry. Previous drug development efforts targeting Wee1 resulted in the clinical-grade inhibitor, AZD1775. However, AZD1775 is burdened by dose-limiting adverse events, and has off-target PLK1 activity. In an attempt to overcome these limitations, we developed Wee1 degraders by conjugating AZD1775 to the cereblon (CRBN)-binding ligand, pomalidomide. The resulting lead compound, ZNL-02-096, degrades Wee1 while sparing PLK1, induces G2/M accumulation at 10-fold lower doses than AZD1775, and synergizes with Olaparib in ovarian cancer cells. We demonstrate that ZNL-02-096 has CRBN-dependent pharmacology that is distinct from AZD1775, which justifies further evaluation of selective Wee1 degraders.


Subject(s)
Cell Cycle Proteins/metabolism , Drug Development , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Proteolysis/drug effects , Pyrazoles/pharmacology , Pyrimidinones/pharmacology , Thalidomide/analogs & derivatives , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage , Female , Humans , Molecular Structure , Phthalazines/chemistry , Phthalazines/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrimidinones/chemistry , Thalidomide/chemistry , Thalidomide/pharmacology
10.
Cell Chem Biol ; 27(1): 66-73.e7, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31859249

ABSTRACT

The PI3K/AKT signaling cascade is one of the most commonly dysregulated pathways in cancer, with over half of tumors exhibiting aberrant AKT activation. Although potent small-molecule AKT inhibitors have entered clinical trials, robust and durable therapeutic responses have not been observed. As an alternative strategy to target AKT, we report the development of INY-03-041, a pan-AKT degrader consisting of the ATP-competitive AKT inhibitor GDC-0068 conjugated to lenalidomide, a recruiter of the E3 ubiquitin ligase substrate adaptor Cereblon (CRBN). INY-03-041 induced potent degradation of all three AKT isoforms and displayed enhanced anti-proliferative effects relative to GDC-0068. Notably, INY-03-041 promoted sustained AKT degradation and inhibition of downstream signaling effects for up to 96 h, even after compound washout. Our findings suggest that AKT degradation may confer prolonged pharmacological effects compared with inhibition, and highlight the potential advantages of AKT-targeted degradation.


Subject(s)
Drug Discovery , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Humans , Molecular Conformation , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry
11.
Nat Commun ; 10(1): 3468, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31371704

ABSTRACT

Targeted protein degradation is a promising drug development paradigm. Here we leverage this strategy to develop a new class of small molecule antivirals that induce proteasomal degradation of viral proteins. Telaprevir, a reversible-covalent inhibitor that binds to the hepatitis C virus (HCV) protease active site is conjugated to ligands that recruit the CRL4CRBN ligase complex, yielding compounds that can both inhibit and induce the degradation of the HCV NS3/4A protease. An optimized degrader, DGY-08-097, potently inhibits HCV in a cellular infection model, and we demonstrate that protein degradation contributes to its antiviral activity. Finally, we show that this new class of antiviral agents can overcome viral variants that confer resistance to traditional enzymatic inhibitors such as telaprevir. Overall, our work provides proof-of-concept that targeted protein degradation may provide a new paradigm for the development of antivirals with superior resistance profiles.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Antiviral Agents/chemistry , Cell Line, Tumor , Drug Design , Drug Resistance, Viral/genetics , Gene Knockdown Techniques , HEK293 Cells , Hepacivirus/drug effects , Hepacivirus/metabolism , Hepatitis C/drug therapy , Hepatitis C/genetics , Hepatitis C/virology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/pharmacology , Proof of Concept Study , Protease Inhibitors/chemistry , Proteolysis/drug effects , Ubiquitin-Protein Ligases/metabolism , Viral Nonstructural Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL