Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
J Med Chem ; 67(15): 13147-13173, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39078366

ABSTRACT

Fungi have historically been the source of numerous important medicinal compounds, but full exploitation of their genetic potential for drug development has been hampered in traditional discovery paradigms. Here we describe a radically different approach, top-down drug discovery (TD3), starting with a massive digital search through a database of over 100,000 fully genomicized fungi to identify loci encoding molecules with a predetermined human target. We exemplify TD3 by the selection of cyclin-dependent kinases (CDKs) as targets and the discovery of two molecules, 1 and 2, which inhibit therapeutically important human CDKs. 1 and 2 exhibit a remarkable mechanism, forming a site-selective covalent bond to the CDK active site Lys. We explored the structure-activity relationship via semi- and total synthesis, generating an analog, 43, with improved kinase selectivity, bioavailability, and efficacy. This work highlights the power of TD3 to identify mechanistically and structurally novel molecules for the development of new medicines.


Subject(s)
Cyclin-Dependent Kinases , Drug Discovery , Protein Kinase Inhibitors , Humans , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Animals , Genomics/methods , Models, Molecular
2.
Mol Cancer Ther ; 22(5): 630-645, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36912782

ABSTRACT

Antitumor immunity can be hampered by immunosuppressive mechanisms in the tumor microenvironment, including recruitment of arginase (ARG) expressing myeloid cells that deplete l-arginine essential for optimal T-cell and natural killer cell function. Hence, ARG inhibition can reverse immunosuppression enhancing antitumor immunity. We describe AZD0011, a novel peptidic boronic acid prodrug to deliver an orally available, highly potent, ARG inhibitor payload (AZD0011-PL). We demonstrate that AZD0011-PL is unable to permeate cells, suggesting that this compound will only inhibit extracellular ARG. In vivo, AZD0011 monotherapy leads to arginine increases, immune cell activation, and tumor growth inhibition in various syngeneic models. Antitumor responses increase when AZD0011 is combined with anti-PD-L1 treatment, correlating with increases in multiple tumor immune cell populations. We demonstrate a novel triple combination of AZD0011, anti-PD-L1, and anti-NKG2A, and combination benefits with type I IFN inducers, including polyI:C and radiotherapy. Our preclinical data demonstrate AZD0011's ability to reverse tumor immunosuppression and enhance immune stimulation and antitumor responses with diverse combination partners providing potential strategies to increase immuno-oncology therapies clinically.


Subject(s)
Arginase , T-Lymphocytes , Humans , Cell Line, Tumor , Immunosuppression Therapy , Immune Tolerance , Tumor Microenvironment
3.
ACS Chem Biol ; 18(2): 296-303, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36602435

ABSTRACT

Lactic acid transport is a key process maintaining glycolytic flux in tumors. Inhibition of this process will result in glycolytic shutdown, impacting on cell growth and survival and thus has been pursued as a therapeutic approach for cancers. Using a cell-based screen in a MCT4-dependent cell line, we identified and optimized compounds for their ability to inhibit the efflux of intracellular lactic acid with good physical and pharmacokinetic properties. To deconvolute the mechanism of lactic acid efflux inhibition, we have developed three assays to measure cellular target engagement. Specifically, we synthesized a biologically active photoaffinity probe (IC50 < 10 nM), and using this probe, we demonstrated selective engagement of MCT4 of our parent molecule through a combination of confocal microscopy and in-cell chemoproteomics. As an orthogonal assay, the cellular thermal shift assay (CETSA) confirmed binding to MCT4 in the cellular system. Comparisons of lactic acid efflux potencies in cells with differential expression of MCT family members further confirmed that the optimized compounds inhibit the efflux of lactic acid through the inhibition of MCT4. Taken together, these data demonstrate the power of orthogonal chemical biology methods to determine cellular target engagement, particularly for proteins not readily amenable to traditional biophysical methods.


Subject(s)
Biology , Lactic Acid , Lactic Acid/metabolism , Biological Transport , Cell Line, Tumor , Cell Proliferation
4.
Nat Commun ; 13(1): 4146, 2022 07 16.
Article in English | MEDLINE | ID: mdl-35842429

ABSTRACT

Enhancing the removal of aggregate-prone toxic proteins is a rational therapeutic strategy for a number of neurodegenerative diseases, especially Huntington's disease and various spinocerebellar ataxias. Ideally, such approaches should preferentially clear the mutant/misfolded species, while having minimal impact on the stability of wild-type/normally-folded proteins. Furthermore, activation of both ubiquitin-proteasome and autophagy-lysosome routes may be advantageous, as this would allow effective clearance of both monomeric and oligomeric species, the latter which are inaccessible to the proteasome. Here we find that compounds that activate the D1 ATPase activity of VCP/p97 fulfill these requirements. Such effects are seen with small molecule VCP activators like SMER28, which activate autophagosome biogenesis by enhancing interactions of PI3K complex components to increase PI(3)P production, and also accelerate VCP-dependent proteasomal clearance of such substrates. Thus, this mode of VCP activation may be a very attractive target for many neurodegenerative diseases.


Subject(s)
Adenosine Triphosphatases , Neurodegenerative Diseases , Valosin Containing Protein , Adenosine Triphosphatases/metabolism , Autophagy , Cell Cycle Proteins/metabolism , Humans , Neurodegenerative Diseases/genetics , Phosphatidylinositol Phosphates , Proteasome Endopeptidase Complex/metabolism , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
5.
ACS Chem Biol ; 17(1): 54-67, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34955012

ABSTRACT

Cyclin-dependent-kinases (CDKs) are members of the serine/threonine kinase family and are highly regulated by cyclins, a family of regulatory subunits that bind to CDKs. CDK9 represents one of the most studied examples of these transcriptional CDKs. CDK9 forms a heterodimeric complex with its regulatory subunit cyclins T1, T2 and K to form the positive transcription elongation factor b (P-TEFb). This complex regulates transcription via the phosphorylation of RNA polymerase II (RNAPolII) on Ser-2, facilitating promoter clearance and transcription elongation and thus remains an attractive therapeutic target. Herein, we have utilized classical affinity purification chemical proteomics, kinobeads assay, compressed CEllular Thermal Shift Assay (CETSA)-MS and Limited Proteolysis (LiP) to study the selectivity, target engagement and downstream mechanistic insights of a CDK9 tool compound. The above experiments highlight the value of quantitative mass spectrometry approaches to drug discovery, specifically proteome wide target identification and selectivity profiling. The approaches utilized in this study unanimously indicated that the CDK family of kinases are the main target of the compound of interest, with CDK9, showing the highest target affinity with remarkable consistency across approaches. We aim to provide guidance to the scientific community on the available chemical biology/proteomic tools to study advanced lead molecules and to highlight pros and cons of each technology while describing our findings in the context of the CDKs biology.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Proteomics , Cell Line, Tumor , Chemical Fractionation , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mass Spectrometry
7.
Int J Mol Sci ; 21(6)2020 Mar 16.
Article in English | MEDLINE | ID: mdl-32188029

ABSTRACT

The ability to quantitatively probe diverse panels of proteins and their post-translational modifications (PTMs) across multiple samples would aid a broad spectrum of biological, biochemical and pharmacological studies. We report a novel, microarray analytical technology that combines immuno-affinity capture with Matrix Assisted Laser Desorption Ionization Mass Spectrometry (MALDI MS), which is capable of supporting highly multiplexed, targeted proteomic assays. Termed "Affinity-Bead Assisted Mass Spectrometry" (Affi-BAMS), this LC-free technology enables development of highly specific and customizable assay panels for simultaneous profiling of multiple proteins and PTMs. While affinity beads have been used previously in combination with MS, the Affi-BAMS workflow uses enrichment on a single bead that contains one type of antibody, generally capturing a single analyte (protein or PTM) while having enough binding capacity to enable quantification within approximately 3 orders of magnitude. The multiplexing capability is achieved by combining Affi-BAMS beads with different protein specificities. To enable screening of bead-captured analytes by MS, we further developed a novel method of performing spatially localized elution of targets from individual beads arrayed on a microscope slide. The resulting arrays of micro spots contain highly concentrated analytes localized within 0.5 mm diameter spots that can be directly measured using MALDI MS. While both intact proteins and protein fragments can be monitored by Affi-BAMS, we initially focused on applying this technology for bottom-up proteomics to enable screening of hundreds of samples per day by combining the robust magnetic bead-based workflow with the high throughput nature of MALDI MS acquisition. To demonstrate the variety of applications and robustness of Affi-BAMS, several studies are presented that focus on the response of 4EBP1, RPS6, ERK1/ERK2, mTOR, Histone H3 and C-MET to stimuli including rapamycin, H2O2, EPO, SU11274, Staurosporine and Vorinostat.


Subject(s)
Microarray Analysis/methods , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Chromatin , Humans , Hydrogen Peroxide , Isotopes , Peptide Hydrolases/chemistry , Point Mutation , Protein Processing, Post-Translational , Proteins/chemistry , Reproducibility of Results , Sensitivity and Specificity , Signal Transduction
8.
Br J Pharmacol ; 177(8): 1709-1718, 2020 04.
Article in English | MEDLINE | ID: mdl-32022252

ABSTRACT

Proteolysis-targeting chimeras are a new drug modality that exploits the endogenous ubiquitin proteasome system to degrade a protein of interest for therapeutic benefit. As the first-generation of proteolysis-targeting chimeras have now entered clinical trials for oncology indications, it is timely to consider the theoretical safety risks inherent with this modality which include off-target degradation, intracellular accumulation of natural substrates for the E3 ligases used in the ubiquitin proteasome system, proteasome saturation by ubiquitinated proteins, and liabilities associated with the "hook effect" of proteolysis-targeting chimeras This review describes in vitro and non-clinical in vivo data that provide mechanistic insight of these safety risks and approaches being used to mitigate these risks in the next generation of proteolysis-targeting chimera molecules to extend therapeutic applications beyond life-threatening diseases.


Subject(s)
Chimera , Pharmaceutical Preparations , Chimera/metabolism , Proteasome Endopeptidase Complex , Proteolysis , Ubiquitin-Protein Ligases/metabolism
9.
Cell Chem Biol ; 27(1): 41-46.e17, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31786184

ABSTRACT

Deregulation of the PRC2 complex, comprised of the core subunits EZH2, SUZ12, and EED, drives aberrant hypermethylation of H3K27 and tumorigenicity of many cancers. Although inhibitors of EZH2 have shown promising clinical activity, preclinical data suggest that resistance can be acquired through secondary mutations in EZH2 that abrogate drug target engagement. To address these limitations, we have designed several hetero-bifunctional PROTACs (proteolysis-targeting chimera) to efficiently target EED for elimination. Our PROTACs bind to EED (pKD ∼ 9.0) and promote ternary complex formation with the E3 ubiquitin ligase. The PROTACs potently inhibit PRC2 enzyme activity (pIC50 ∼ 8.1) and induce rapid degradation of not only EED but also EZH2 and SUZ12 within the PRC2 complex. Furthermore, the PROTACs selectively inhibit proliferation of PRC2-dependent cancer cells (half maximal growth inhibition [GI50] = 49-58 nM). In summary, our data demonstrate a therapeutic modality to target PRC2-dependent cancer through a PROTAC-mediated degradation mechanism.


Subject(s)
Polycomb Repressive Complex 2/metabolism , Proteolysis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Polycomb Repressive Complex 2/antagonists & inhibitors , Structure-Activity Relationship
10.
ACS Chem Biol ; 14(9): 1913-1920, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31329413

ABSTRACT

Demonstration of target binding is a key requirement for understanding the mode of action of new therapeutics. The cellular thermal shift assay (CETSA) has been introduced as a powerful label-free method to assess target engagement in physiological environments. Here, we present the application of live-cell CETSA to different classes of integral multipass transmembrane proteins using three case studies, the first showing a large and robust stabilization of the outer mitochondrial five-pass transmembrane protein TSPO, the second being a modest stabilization of SERCA2, and the last describing an atypical compound-driven stabilization of the GPCR PAR2. Our data demonstrated that using modified protocols with detergent extraction after the heating step, CETSA can reliably be applied to several membrane proteins of different complexity. By showing examples with distinct CETSA behaviors, we aim to provide the scientific community with an overview of different scenarios to expect during CETSA experiments, especially for challenging, membrane bound targets.


Subject(s)
Receptor, PAR-2/metabolism , Receptors, GABA/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Aminoquinolines/pharmacology , Benzamides/pharmacology , Benzimidazoles/pharmacology , Benzodiazepinones/pharmacology , Benzodioxoles/pharmacology , Benzyl Alcohols/pharmacology , Biological Assay , Cell Line, Tumor , GABA Antagonists/pharmacology , HEK293 Cells , Hot Temperature , Humans , Imidazoles/pharmacology , Phase Transition/drug effects , Protein Multimerization/drug effects , Pyridines/pharmacology , Receptor, PAR-2/antagonists & inhibitors , Receptor, PAR-2/chemistry , Receptors, GABA/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Thapsigargin/pharmacology
11.
ACS Chem Biol ; 13(11): 3131-3141, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30335946

ABSTRACT

B-cell lymphoma 6 (BCL6) inhibition is a promising mechanism for treating hematological cancers but high quality chemical probes are necessary to evaluate its therapeutic potential. Here we report potent BCL6 inhibitors that demonstrate cellular target engagement and exhibit exquisite selectivity for BCL6 based on mass spectrometry analyses following chemical proteomic pull down. Importantly, a proteolysis-targeting chimera (PROTAC) was also developed and shown to significantly degrade BCL6 in a number of diffuse large B-cell lymphoma (DLBCL) cell lines, but neither BCL6 inhibition nor degradation selectively induced marked phenotypic response. To investigate, we monitored PROTAC directed BCL6 degradation in DLBCL OCI-Ly1 cells by immunofluorescence and discovered a residual BCL6 population. Analysis of subcellular fractions also showed incomplete BCL6 degradation in all fractions despite having measurable PROTAC concentrations, together providing a rationale for the weak antiproliferative response seen with both BCL6 inhibitor and degrader. In summary, we have developed potent and selective BCL6 inhibitors and a BCL6 PROTAC that effectively degraded BCL6, but both modalities failed to induce a significant phenotypic response in DLBCL despite achieving cellular concentrations.


Subject(s)
Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , Quinolones/pharmacology , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Ligands , Lymphoma, Large B-Cell, Diffuse/drug therapy , Peptide Hydrolases/metabolism , Protein Binding , Proteolysis , Proto-Oncogene Proteins c-bcl-6/chemistry , Proto-Oncogene Proteins c-bcl-6/metabolism , Quinolones/chemical synthesis , Quinolones/metabolism , Thalidomide/chemical synthesis , Thalidomide/metabolism , Ubiquitin-Protein Ligases/metabolism
12.
ACS Chem Biol ; 13(10): 2849-2854, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30216042

ABSTRACT

Targeting the protein-protein interaction between p53 and MDM2/MDMX (MDM4) represents an attractive anticancer strategy for the treatment of p53-competent tumors. Several selective and potent MDM2 inhibitors have been developed and entered the clinic; however, the repertoire of MDMX antagonists is still limited. The arylmethylidenepyrazolinone SJ-172550 has been reported as a selective MDMX antagonist; yet, uncertainties about its mechanism of action have raised doubts about its use as a chemical probe. Here, we show that, in addition to its unclear mode of action, SJ-172550 is unstable in aqueous buffers, giving rise to side products of unknown biological activity. Using an SJ-172550-derived affinity probe, we observed promiscuous binding to cellular proteins whereas cellular thermal shift assays did not reveal a stabilizing effect on MDMX. Overall, our results raise further questions about the interpretation of data using SJ-172550 and related compounds to investigate cellular phenotypes.


Subject(s)
Acetates/metabolism , Enzyme Inhibitors/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Pyrazoles/metabolism , Acetates/chemistry , Affinity Labels/chemistry , Alkynes/chemistry , Binding Sites , Carbocyanines/chemistry , Cell Cycle Proteins , Cell Line, Tumor , Click Chemistry , Drug Stability , Enzyme Inhibitors/chemistry , Humans , Nuclear Proteins/chemistry , Protein Binding , Protein Stability/drug effects , Proto-Oncogene Proteins/chemistry , Pyrazoles/chemistry
13.
Cell Chem Biol ; 25(6): 705-717.e11, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29628435

ABSTRACT

Activating KRAS mutations are major oncogenic drivers in multiple tumor types. Synthetic lethal screens have previously been used to identify targets critical for the survival of KRAS mutant cells, but their application to drug discovery has proven challenging, possibly due in part to a failure of monolayer cultures to model tumor biology. Here, we report the results of a high-throughput synthetic lethal screen for small molecules that selectively inhibit the growth of KRAS mutant cell lines in soft agar. Chemoproteomic profiling identifies the target of the most KRAS-selective chemical series as dihydroorotate dehydrogenase (DHODH). DHODH inhibition is shown to perturb multiple metabolic pathways. In vivo preclinical studies demonstrate strong antitumor activity upon DHODH inhibition in a pancreatic tumor xenograft model.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrimidines/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Dihydroorotate Dehydrogenase , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Humans , Mice , Mice, SCID , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured
14.
ACS Synth Biol ; 7(4): 1152-1162, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29609459

ABSTRACT

Monoclonal antibody therapeutics have revolutionized the treatment of diseases such as cancer and autoimmune disorders, and also serve as research reagents for diverse and unparalleled applications. To extend their utility in both contexts, we have begun development of tunable antibodies, whose activity can be controlled by addition of a small molecule. Conceptually, we envision that incorporating cavity-forming mutations into an antibody can disrupt its structure, thereby reducing its affinity for antigen; addition of a small molecule may then restore the active structure, and thus rescue antigen binding. As a first proof of concept toward implementing this strategy, we have incorporated individual tryptophan to glycine mutations into FITC-E2, an anti-fluorescein single-chain variable fragment (scFv). We find that these can disrupt the protein structure and diminish antigen binding, and further that both structure and function can be rescued by addition of indole to complement the deleted side chain. While the magnitude of the affinity difference triggered by indole is modest in this first model system, it nonetheless provides a framework for future mutation/ligand pairs that may induce more dramatic responses. Disrupting and subsequently rescuing antibody activity, as exemplified by this first example, may represent a new approach to "design in" fine-tuned control of antibody activity for a variety of future applications.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Protein Engineering/methods , Amino Acid Substitution , Antibodies, Monoclonal/genetics , Fluorescein-5-isothiocyanate/chemistry , Fluorescein-5-isothiocyanate/metabolism , Fluorescence , Glycine/genetics , Indoles/chemistry , Models, Molecular , Mutagenesis, Site-Directed/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , Structure-Activity Relationship , Tryptophan/genetics
15.
ChemMedChem ; 12(12): 917-924, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28371485

ABSTRACT

Wnt signaling is critical for development, cell proliferation and differentiation, and mutations in this pathway resulting in constitutive signaling have been implicated in various cancers. A pathway screen using a Wnt-dependent reporter identified a chemical series based on a 1,2,3-thiadiazole-5-carboxamide (TDZ) core with sub-micromolar potency. Herein we report a comprehensive mechanism-of-action deconvolution study toward identifying the efficacy target(s) and biological implication of this chemical series involving bottom-up quantitative chemoproteomics, cell biology, and biochemical methods. Through observing the effects of our probes on metabolism and performing confirmatory cellular and biochemical assays, we found that this chemical series inhibits ATP synthesis by uncoupling the mitochondrial potential. Affinity chemoproteomics experiments identified sarco(endo)plasmic reticulum Ca2+ -dependent ATPase (SERCA2) as a binding partner of the TDZ series, and subsequent validation studies suggest that the TDZ series can act as ionophores through SERCA2 toward Wnt pathway inhibition.


Subject(s)
Oxidative Phosphorylation/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thiadiazoles/pharmacology , Wnt Signaling Pathway/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , Thiadiazoles/chemical synthesis , Thiadiazoles/chemistry
16.
Org Lett ; 11(15): 3362-5, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19719184

ABSTRACT

The synthesis of spirocyclic oxindole pyran and oxepene frameworks using highly stereoselective Prins cyclizations of homoallylic and bis-homoallylic alcohols and isatin ketals is described.


Subject(s)
Alkenes/chemical synthesis , Indoles/chemical synthesis , Oxepins/chemical synthesis , Pyrans/chemical synthesis , Silanes/chemical synthesis , Alkenes/chemistry , Cyclization , Indoles/chemistry , Isatin/chemistry , Oxepins/chemistry , Oxindoles , Propanols/chemistry , Pyrans/chemistry , Silanes/chemistry , Spiro Compounds/chemistry , Stereoisomerism
17.
Nat Chem Biol ; 5(11): 823-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19767736

ABSTRACT

The internal ribosome entry site (IRES), a highly conserved structured element of the hepatitis C virus (HCV) genomic RNA, is an attractive target for antiviral drugs. Here we show that benzimidazole inhibitors of the HCV replicon act by conformational induction of a widened interhelical angle in the IRES subdomain IIa, which facilitates the undocking of subdomain IIb from the ribosome and ultimately leads to inhibition of IRES-driven translation in HCV-infected cells.


Subject(s)
Hepacivirus/metabolism , Ribosomes/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Base Sequence , Benzimidazoles/pharmacology , Cytosine/chemistry , Genome, Viral/drug effects , Genotype , Hepacivirus/chemistry , Hepacivirus/drug effects , Hepacivirus/genetics , Magnesium/pharmacology , Models, Molecular , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Replicon/drug effects , Replicon/genetics , Ribosomes/genetics , Ribosomes/virology , Signal Transduction
18.
Chem Commun (Camb) ; (29): 3045-62, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16855685

ABSTRACT

Throughout history symmetry and chirality have inspired artists and scientists alike. Given that rotational axes are the only elements of symmetry compatible with chirality, it is not surprising that C2- and C3-symmetrical molecules have attracted considerable attention. In recent years, the aesthetic appeal of C2-symmetrical molecules has been translated into many widely-used applications some of which are of commercial importance by its exploitation in the area of asymmetric catalysis. In contrast, exploitation of the arguably greater aesthetic appeal of C3-symmetric molecules is still in its infancy. This review, which surveys the applications of chiral C3-symmetrical molecules in the areas of asymmetric catalysis, molecular recognition and nanoarchitecture, has been designed with a view to identifying some of the most promising areas of application of these very beautiful molecules.

20.
Chemistry ; 12(1): 138-48, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16278918

ABSTRACT

Chiral base chemistry has been used to create three chiral centres in one pot on a C3-symmetric substrate. The potential of this new approach to C3-symmetric molecules is exemplified by the creation of an enantiopure C3v-symmetric triol, triphosphane and tripyridine. A ruthenium complex of the last compound has been studied by X-ray crystallography.

SELECTION OF CITATIONS
SEARCH DETAIL