Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 15(7): 1151-1158, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39015284

ABSTRACT

MUS81 is a structure-selective endonuclease that cleaves various branched DNA structures arising from natural physiological processes such as homologous recombination and mitosis. Due to this, MUS81 is able to relieve replication stress, and its function has been reported to be critical to the survival of many cancers, particularly those with dysfunctional DNA-repair machinery. There is therefore interest in MUS81 as a cancer drug target, yet there are currently few small molecule inhibitors of this enzyme reported, and no liganded crystal structures are available to guide hit optimization. Here we report the fragment-based discovery of novel small molecule MUS81 inhibitors with sub-µM biochemical activity. These inhibitors were used to develop a novel crystal system, providing the first structural insight into the inhibition of MUS81 with small molecules.

2.
Nat Commun ; 14(1): 7093, 2023 11 04.
Article in English | MEDLINE | ID: mdl-37925433

ABSTRACT

Human antigen R (HuR) is a ubiquitously expressed RNA-binding protein, which functions as an RNA regulator. Overexpression of HuR correlates with high grade tumours and poor patient prognosis, implicating it as an attractive therapeutic target. However, an effective small molecule antagonist to HuR for clinical use remains elusive. Here, a single domain antibody (VHH) that binds HuR with low nanomolar affinity was identified and shown to inhibit HuR binding to RNA. This VHH was used to engineer a TRIM21-based biological PROTAC (bioPROTAC) that could degrade endogenous HuR. Significantly, HuR degradation reverses the tumour-promoting properties of cancer cells in vivo by altering the HuR-regulated proteome, highlighting the benefit of HuR degradation and paving the way for the development of HuR-degrading therapeutics. These observations have broader implications for degrading intractable therapeutic targets, with bioPROTACs presenting a unique opportunity to explore targeted-protein degradation through a modular approach.


Subject(s)
ELAV-Like Protein 1 , Neoplasms , Proteolysis Targeting Chimera , Humans , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , RNA , RNA-Binding Proteins/metabolism
3.
J Med Chem ; 64(23): 17287-17303, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34787423

ABSTRACT

RAS proteins are central in the proliferation of many types of cancer, but a general approach toward the identification of pan-mutant RAS inhibitors has remained unresolved. In this work, we describe the application of a binding pharmacophore identified from analysis of known RAS binding peptides to the design of novel peptides. Using a chemically divergent approach, we generated a library of small stapled peptides from which we identified compounds with weak binding activity. Exploration of structure-activity relationships (SARs) and optimization of these early compounds led to low-micromolar binders of KRAS that block nucleotide exchange.


Subject(s)
Peptides/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Binding Sites , Cyclization , Humans , Hydrophobic and Hydrophilic Interactions , Peptides/chemistry , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/chemistry , Structure-Activity Relationship
4.
Wellcome Open Res ; 5: 20, 2020.
Article in English | MEDLINE | ID: mdl-32587898

ABSTRACT

Targeting the interaction of proteins with weak binding affinities or low solubility represents a particular challenge for drug screening. The NanoLuc â ® Binary Technology (NanoBiT â ®) was originally developed to detect protein-protein interactions in live mammalian cells. Here we report the successful translation of the NanoBit cellular assay into a biochemical, cell-free format using mammalian cell lysates. We show that the assay is suitable for the detection of both strong and weak protein interactions such as those involving the binding of RAS oncoproteins to either RAF or phosphoinositide 3-kinase (PI3K) effectors respectively, and that it is also effective for the study of poorly soluble protein domains such as the RAS binding domain of PI3K. Furthermore, the RAS interaction assay is sensitive and responds to both strong and weak RAS inhibitors. Our data show that the assay is robust, reproducible, cost-effective, and can be adapted for small and large-scale screening approaches. The NanoBit Biochemical Assay offers an attractive tool for drug screening against challenging protein-protein interaction targets, including the interaction of RAS with PI3K.

5.
ACS Chem Biol ; 14(10): 2134-2140, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31525019

ABSTRACT

Enhancer of zeste homologue 2 (EZH2), the catalytic subunit of polycomb repressive complex 2 (PRC2), regulates chromatin state and gene expression by methylating histone H3 lysine 27. EZH2 is overexpressed or mutated in various hematological malignancies and solid cancers. Our previous efforts to identify inhibitors of PRC2 methyltransferase activity by high-throughput screening (HTS) resulted in large numbers of false positives and thus a significant hit deconvolution challenge. More recently, others have reported compounds that bind to another PRC2 core subunit, EED, and allosterically inhibit EZH2 activity. This mechanism is particularly appealing as it appears to retain potency in cell lines that have acquired resistance to orthosteric EZH2 inhibition. By designing a fluorescence polarization probe based on the reported EED binding compounds, we were able to quickly and cleanly re-triage our previously challenging HTS hit list and identify novel allosteric PRC2 inhibitors.


Subject(s)
Benzofurans/chemistry , Enzyme Inhibitors/chemistry , Polycomb Repressive Complex 2/antagonists & inhibitors , Small Molecule Libraries/chemistry , Allosteric Regulation/drug effects , Benzofurans/metabolism , Carbocyanines/chemistry , Cell Line, Tumor , Enzyme Inhibitors/metabolism , Fluorescent Dyes/chemistry , High-Throughput Screening Assays , Humans , Ligands , Polycomb Repressive Complex 2/isolation & purification , Polycomb Repressive Complex 2/metabolism , Protein Binding , Small Molecule Libraries/metabolism
6.
ACS Chem Biol ; 12(12): 3113-3125, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29131570

ABSTRACT

The ubiquitin proteasome system is widely postulated to be a new and important field of drug discovery for the future, with the ubiquitin specific proteases (USPs) representing one of the more attractive target classes within the area. Many USPs have been linked to critical axes for therapeutic intervention, and the finding that USP28 is required for c-Myc stability suggests that USP28 inhibition may represent a novel approach to targeting this so far undruggable oncogene. Here, we describe the discovery of the first reported inhibitors of USP28, which we demonstrate are able to bind to and inhibit USP28, and while displaying a dual activity against the closest homologue USP25, these inhibitors show a high degree of selectivity over other deubiquitinases (DUBs). The utility of these compounds as valuable probes to investigate and further explore cellular DUB biology is highlighted by the demonstration of target engagement against both USP25 and USP28 in cells. Furthermore, we demonstrate that these inhibitors are able to elicit modulation of both the total levels and the half-life of the c-Myc oncoprotein in cells and also induce apoptosis and loss of cell viability in a range of cancer cell lines. We however observed a narrow therapeutic index compared to a panel of tissue-matched normal cell lines. Thus, it is hoped that these probes and data presented herein will further advance our understanding of the biology and tractability of DUBs as potential future therapeutic targets.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , HCT116 Cells , Humans
7.
J Med Chem ; 58(5): 2265-74, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25695162

ABSTRACT

Constitutively active mutant KRas displays a reduced rate of GTP hydrolysis via both intrinsic and GTPase-activating protein-catalyzed mechanisms, resulting in the perpetual activation of Ras pathways. We describe a fragment screening campaign using X-ray crystallography that led to the discovery of three fragment binding sites on the Ras:SOS complex. The identification of tool compounds binding at each of these sites allowed exploration of two new approaches to Ras pathway inhibition by stabilizing or covalently modifying the Ras:SOS complex to prevent the reloading of Ras with GTP. Initially, we identified ligands that bound reversibly to the Ras:SOS complex in two distinct sites, but these compounds were not sufficiently potent inhibitors to validate our stabilization hypothesis. We conclude by demonstrating that covalent modification of Cys118 on Ras leads to a novel mechanism of inhibition of the SOS-mediated interaction between Ras and Raf and is effective at inhibiting the exchange of labeled GDP in both mutant (G12C and G12V) and wild type Ras.


Subject(s)
Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/metabolism , Small Molecule Libraries/pharmacology , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Mutation/genetics , Protein Binding/drug effects , Protein Conformation , Proto-Oncogene Proteins p21(ras)/genetics , SOS1 Protein/chemistry , Small Molecule Libraries/chemistry
8.
J Med Chem ; 55(7): 3285-306, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22417091

ABSTRACT

Lactate dehydrogenase A (LDHA) catalyzes the conversion of pyruvate to lactate, utilizing NADH as a cofactor. It has been identified as a potential therapeutic target in the area of cancer metabolism. In this manuscript we report our progress using fragment-based lead generation (FBLG), assisted by X-ray crystallography to develop small molecule LDHA inhibitors. Fragment hits were identified through NMR and SPR screening and optimized into lead compounds with nanomolar binding affinities via fragment linking. Also reported is their modification into cellular active compounds suitable for target validation work.


Subject(s)
L-Lactate Dehydrogenase/antagonists & inhibitors , Animals , Catalytic Domain , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Enzyme Assays , Humans , Isoenzymes/antagonists & inhibitors , Lactate Dehydrogenase 5 , Magnetic Resonance Spectroscopy , Malonates/chemical synthesis , Malonates/chemistry , Malonates/pharmacology , Models, Molecular , Molecular Structure , Niacinamide/chemistry , Oxamic Acid/analogs & derivatives , Oxamic Acid/chemical synthesis , Oxamic Acid/chemistry , Oxamic Acid/pharmacology , Protein Binding , Rats , Structure-Activity Relationship , Surface Plasmon Resonance
9.
Ann Rheum Dis ; 70(4): 683-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21216815

ABSTRACT

OBJECTIVE: Aggrecan is a critical component of cartilage extracellular matrix. Several members of the 'a disintegrin and metalloproteinase with thrombospondin motifs' (ADAMTS) family have been characterised as aggrecanases by their ability to generate fragments containing the NITEGE neoepitope from aggrecan. Increased NITEGE fragments in synovial fluid and articular cartilage are a hallmark of osteoarthritis (OA) and it is hypothesised that the enhanced rate of aggrecan degradation is critical for cartilage destruction in OA. Recently, matrix metalloproteinase 17 (MMP17, also known as MT4-MMP) has been implicated in the activation of one of the key aggrecanases: ADAMTS4. In the present work, the hypothesis that MMP17 mediates the interleukin 1ß (IL-1ß) induced release of NITEGE neoepitope from human and murine articular cartilage is investigated. METHODS: MMP17 was quantified at the protein and RNA level and NITEGE neoepitope generation by immunohistochemistry. Human postmortem articular cartilage explants were treated with recombinant MMP17, or IL-1ß in the presence or absence of an MMP17 inhibitor. Glycosaminoglycan (GAG) loss into the media was quantified using the 1,9-dimethylmethylene blue (DMMB) assay. Intra-articular injection (IAI) of IL-1ß or meniscotibial ligament transaction was carried out in MMP17 null mice. RESULTS: The data reveal an association between increased MMP17 protein and NITEGE staining in areas of OA cartilage damage. Ex vivo treatment of normal human cartilage with recombinant MMP17 protein increased NITEGE generation in the cartilage and GAG loss into the media. In addition, IL-1ß mediated cartilage GAG loss, and increased NITEGE neoepitope expression, were attenuated with an MMP17 inhibitor. IAI of IL-1ß into C57BL6/Jax mice resulted in increased MMP17 expression in articular cartilage and increased GAG content in the synovial fluid. MMP17 null mice were protected against this increase. However, aggrecan loss driven by mechanical stress following medial meniscotibial ligament transection was not dependent on MMP17. CONCLUSION: These data further implicate MMP17 in the control of articular cartilage extracellular matrix aggrecan integrity in an inflammatory environment.


Subject(s)
Aggrecans/metabolism , Cartilage, Articular/metabolism , Matrix Metalloproteinase 17/physiology , Animals , Cartilage, Articular/drug effects , Endopeptidases/metabolism , Glycosaminoglycans/metabolism , Humans , Matrix Metalloproteinase 17/pharmacology , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Protease Inhibitors/pharmacology , Recombinant Proteins/pharmacology , Tissue Culture Techniques
10.
J Biol Chem ; 279(21): 22377-86, 2004 May 21.
Article in English | MEDLINE | ID: mdl-14970227

ABSTRACT

Tumor necrosis factor (TNF) alpha-converting enzyme (TACE/ADAM-17) has diverse roles in the proteolytic processing of cell surface molecules and, due to its ability to process TNFalpha, is a validated therapeutic target for anti-inflammatory therapies. Unlike a number of other ADAM proteins, which interact with integrin receptors via their disintegrin domains, there is currently no evidence for an ADAM-17-integrin association. By analyzing the adhesion of a series of cell lines with recombinant fragments of the extracellular domain of ADAM-17, we now demonstrate a functional interaction between ADAM-17 and alpha(5)beta(1) integrin in a trans orientation. Because ADAM-17-mediated adhesion was sensitive to RGD peptides and EDTA, and the integrin-binding site within ADAM-17 was narrowed down to the disintegrin/cysteine-rich region, the two molecules appear to have a ligand-receptor relationship mediated by the alpha(5)beta(1) ligand binding pocket. Intriguingly, ADAM-17 and alpha(5)beta(1) were found to co-localize in both membrane ruffles and focal adhesions in HeLa cells. When confluent HeLa cell monolayers were wounded, ADAM-17 and alpha(5)beta(1) redistributed to the leading edge and co-localized, which is suggestive of a cis orientation. We postulate that the interaction of ADAM-17 with alpha(5)beta(1) may target or modulate its metalloproteolytic activity.


Subject(s)
Integrin alpha5beta1/metabolism , Metalloendopeptidases/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Blotting, Western , COS Cells , Cations , Cell Adhesion , Cell Line , Cell Movement , Cell Separation , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Edetic Acid/chemistry , Fibroblasts/metabolism , Flow Cytometry , HeLa Cells , Humans , Inflammation , Ligands , Microscopy, Fluorescence , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Skin/cytology , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...