Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 34(9): 1679-1687, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37657082

ABSTRACT

Protein arylation has attracted much attention for developing new classes of bioconjugates with improved properties. Here, we have evaluated 2-sulfonylpyrimidines as covalent warheads for the mild, chemoselective, and metal free cysteine S-arylation. 2-Sulfonylpyrimidines react rapidly with cysteine, resulting in stable S-heteroarylated adducts at neutral pH. Fine tuning the heterocyclic core and exocyclic leaving group allowed predictable SNAr reactivity in vitro, covering >9 orders of magnitude. Finally, we achieved fast chemo- and regiospecific arylation of a mutant p53 protein and confirmed arylation sites by protein X-ray crystallography. Hence, we report the first example of a protein site specifically S-arylated with iodo-aromatic motifs. Overall, this study provides the most comprehensive structure-reactivity relationship to date on heteroaryl sulfones and highlights 2-sulfonylpyrimidine as a synthetically tractable and protein compatible covalent motif for targeting reactive cysteines, expanding the arsenal of tunable warheads for modern covalent ligand discovery.


Subject(s)
Cysteine , Sulfones , Mutant Proteins , Crystallography, X-Ray
2.
EMBO Mol Med ; 15(3): e14837, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36789546

ABSTRACT

Multiple sulfatase deficiency (MSD, MIM #272200) results from pathogenic variants in the SUMF1 gene that impair proper function of the formylglycine-generating enzyme (FGE). FGE is essential for the posttranslational activation of cellular sulfatases. MSD patients display reduced or absent sulfatase activities and, as a result, clinical signs of single sulfatase disorders in a unique combination. Up to date therapeutic options for MSD are limited and mostly palliative. We performed a screen of FDA-approved drugs using immortalized MSD patient fibroblasts. Recovery of arylsulfatase A activity served as the primary readout. Subsequent analysis confirmed that treatment of primary MSD fibroblasts with tazarotene and bexarotene, two retinoids, led to a correction of MSD pathophysiology. Upon treatment, sulfatase activities increased in a dose- and time-dependent manner, reduced glycosaminoglycan content decreased and lysosomal position and size normalized. Treatment of MSD patient derived induced pluripotent stem cells (iPSC) differentiated into neuronal progenitor cells (NPC) resulted in a positive treatment response. Tazarotene and bexarotene act to ultimately increase the stability of FGE variants. The results lay the basis for future research on the development of a first therapeutic option for MSD patients.


Subject(s)
Multiple Sulfatase Deficiency Disease , Humans , Multiple Sulfatase Deficiency Disease/diagnosis , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/pathology , Bexarotene , Drug Evaluation, Preclinical , Sulfatases/genetics , Oxidoreductases Acting on Sulfur Group Donors
3.
ACS Pharmacol Transl Sci ; 5(11): 1169-1180, 2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36407959

ABSTRACT

The tumor suppressor protein p53 is inactivated in the majority of human cancers and remains a prime target for developing new drugs to reactivate its tumor suppressing activity for anticancer therapies. The oncogenic p53 mutant Y220C accounts for approximately 125,000 new cancer cases per annum and is one of the most prevalent p53 mutants overall. It harbors a narrow, mutationally induced pocket at the surface of the DNA-binding domain that destabilizes p53, leading to its rapid denaturation and aggregation. Here, we present the structure-guided development of high-affinity small molecules stabilizing p53-Y220C in vitro, along with the synthetic routes developed in the process, in vitro structure-activity relationship data, and confirmation of their binding mode by protein X-ray crystallography. We disclose two new chemical probes displaying sub-micromolar binding affinity in vitro, marking an important milestone since the discovery of the first small-molecule ligand of Y220C in 2008. New chemical probe JC744 displayed a K d = 320 nM, along with potent in vitro protein stabilization. This study, therefore, represents a significant advance toward high-affinity Y220C ligands for clinical evaluation.

4.
J Med Chem ; 65(10): 7246-7261, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35581674

ABSTRACT

The canonical Wingless-related integration site signaling pathway plays a critical role in human physiology, and its dysregulation can lead to an array of diseases. ß-Catenin is a multifunctional protein within this pathway and an attractive yet challenging therapeutic target, most notably in oncology. This has stimulated the search for potent small-molecule inhibitors binding directly to the ß-catenin surface to inhibit its protein-protein interactions and downstream signaling. Here, we provide an account of the claimed (and some putative) small-molecule ligands of ß-catenin from the literature. Through in silico analysis, we show that most of these molecules contain promiscuous chemical substructures notorious for interfering with screening assays. Finally, and in line with this analysis, we demonstrate using orthogonal biophysical techniques that none of the examined small molecules bind at the surface of ß-catenin. While shedding doubts on their reported mode of action, this study also reaffirms ß-catenin as a prominent target in drug discovery.


Subject(s)
Small Molecule Libraries , Wnt Signaling Pathway , beta Catenin , Animals , Biophysical Phenomena , Drug Discovery , Humans , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
5.
Front Oncol ; 10: 1460, 2020.
Article in English | MEDLINE | ID: mdl-32974171

ABSTRACT

The transcription factor p53 is a key tumor suppressor that is inactivated in almost all cancers due to either point mutations in the TP53 gene or overexpression of its negative regulators. The p53 protein is known as the "cellular gatekeeper" for its roles in facilitating DNA repair, cell cycle arrest or apoptosis upon DNA damage. Most p53 mutations are missense and result in either structural destabilization of the protein, causing its partial unfolding and deactivation under physiological conditions, or impairment of its DNA-binding properties. Tumor cells with p53 mutations are generally more immunogenic due to "hot spot" neoantigens that instigate the immune system response. In this review, we discuss the key therapeutic strategies targeting mutant p53 tumors, including classical approaches based on small molecule intervention and emerging technologies such as gene editing and T cell immunotherapy.

6.
Explor Target Antitumor Ther ; 1(5): 273-312, 2020.
Article in English | MEDLINE | ID: mdl-36046485

ABSTRACT

PROteolysis TArgeting Chimeras (PROTACs) are heterobifunctional molecules consisting of two ligands; an "anchor" to bind to an E3 ubiquitin ligase and a "warhead" to bind to a protein of interest, connected by a chemical linker. Targeted protein degradation by PROTACs has emerged as a new modality for the knock down of a range of proteins, with the first agents now reaching clinical evaluation. It has become increasingly clear that the length and composition of the linker play critical roles on the physicochemical properties and bioactivity of PROTACs. While linker design has historically received limited attention, the PROTAC field is evolving rapidly and currently undergoing an important shift from synthetically tractable alkyl and polyethylene glycol to more sophisticated functional linkers. This promises to unlock a wealth of novel PROTAC agents with enhanced bioactivity for therapeutic intervention. Here, the authors provide a timely overview of the diverse linker classes in the published literature, along with their underlying design principles and overall influence on the properties and bioactivity of the associated PROTACs. Finally, the authors provide a critical analysis of current strategies for PROTAC assembly. The authors highlight important limitations associated with the traditional "trial and error" approach around linker design and selection, and suggest potential future avenues to further inform rational linker design and accelerate the identification of optimised PROTACs. In particular, the authors believe that advances in computational and structural methods will play an essential role to gain a better understanding of the structure and dynamics of PROTAC ternary complexes, and will be essential to address the current gaps in knowledge associated with PROTAC design.

7.
Eur J Med Chem ; 152: 101-114, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29702446

ABSTRACT

Many cancers have the tumor suppressor p53 inactivated by mutation, making reactivation of mutant p53 with small molecules a promising strategy for the development of novel anticancer therapeutics. The oncogenic p53 mutation Y220C, which accounts for approximately 100,000 cancer cases per year, creates an extended surface crevice in the DNA-binding domain, which destabilizes p53 and causes denaturation and aggregation. Here, we describe the structure-guided design of a novel class of small-molecule Y220C stabilizers and the challenging synthetic routes developed in the process. The synthesized chemical probe MB710, an aminobenzothiazole derivative, binds tightly to the Y220C pocket and stabilizes p53-Y220C in vitro. MB725, an ethylamide analogue of MB710, induced selective viability reduction in several p53-Y220C cancer cell lines while being well tolerated in control cell lines. Reduction of viability correlated with increased and selective transcription of p53 target genes such as BTG2, p21, PUMA, FAS, TNF, and TNFRSF10B, which promote apoptosis and cell cycle arrest, suggesting compound-mediated transcriptional activation of the Y220C mutant. Our data provide a framework for the development of a class of potent, non-toxic compounds for reactivating the Y220C mutant in anticancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Structure-Activity Relationship , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
8.
ACS Omega ; 2(8): 4328-4332, 2017 Aug 31.
Article in English | MEDLINE | ID: mdl-31457724

ABSTRACT

TC AC 28, 6-(1H-Indol-4-yl)-8-methoxy-1-methyl-4H-[1,2,4]triazolo[4,3-a][1,4]benzodiazepine-4-acetic acid methyl ester, has been synthesized on a near-gram scale in seven steps with notable improvements in the reported poor-yielding last two steps enabling this key chemical probe compound to be available for researchers.

9.
ACS Infect Dis ; 3(2): 152-161, 2017 02 10.
Article in English | MEDLINE | ID: mdl-27756124

ABSTRACT

Photopharmacological agents exhibit light-dependent biological activity and may have potential in the development of new antimicrobial agents/modalities. Amidohydrolase enzymes homologous to the well-known human histone deacetylases (HDACs) are present in bacteria, including resistant organisms responsible for a significant number of hospital-acquired infections and deaths. We report photopharmacological inhibitors of these enzymes, using two classes of photoswitches embedded in the inhibitor pharmacophore: azobenzenes and arylazopyrazoles. Although both classes of inhibitor show excellent inhibitory activity (nM IC50 values) of the target enzymes and promising differential activity of the switchable E- and Z-isomeric forms, the arylazopyrazoles exhibit better intrinsic photoswitch performance (more complete switching, longer thermal lifetime of the Z-isomer). We also report protein-ligand crystal structures of the E-isomers of both an azobenzene and an arylazopyrazole inhibitor, bound to bacterial histone deacetylase-like amidohydrolases (HDAHs). These structures not only uncover interactions important for inhibitor binding but also reveal conformational differences between the two photoswitch inhibitor classes. As such, our data may pave the way for the design of improved photopharmacological agents targeting the HDAC superfamily.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Infective Agents/chemistry , Enzyme Inhibitors/chemistry , Photosensitizing Agents/chemistry , Anti-Infective Agents/pharmacology , Azo Compounds/chemistry , Bacteria/drug effects , Bacteria/enzymology , Bacterial Proteins/antagonists & inhibitors , Catalytic Domain/drug effects , Enzyme Inhibitors/pharmacology , Photosensitizing Agents/pharmacology , Pyrazoles/chemistry
10.
ACS Chem Biol ; 11(8): 2265-74, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27267810

ABSTRACT

Many oncogenic mutants of the tumor suppressor p53 are conformationally unstable, including the frequently occurring Y220C mutant. We have previously developed several small-molecule stabilizers of this mutant. One of these molecules, PhiKan083, 1-(9-ethyl-9H-carbazole-3-yl)-N-methylmethanamine, binds to a mutation-induced surface crevice with a KD = 150 µM, thereby increasing the melting temperature of the protein and slowing its rate of aggregation. Incorporation of fluorine atoms into small molecule ligands can substantially improve binding affinity to their protein targets. We have, therefore, harnessed fluorine-protein interactions to improve the affinity of this ligand. Step-wise introduction of fluorines at the carbazole ethyl anchor, which is deeply buried within the binding site in the Y220C-PhiKan083 complex, led to a 5-fold increase in affinity for a 2,2,2-trifluoroethyl anchor (ligand efficiency of 0.3 kcal mol(-1) atom(-1)). High-resolution crystal structures of the Y220C-ligand complexes combined with quantum chemical calculations revealed favorable interactions of the fluorines with protein backbone carbonyl groups (Leu145 and Trp146) and the sulfur of Cys220 at the mutation site. Affinity gains were, however, only achieved upon trifluorination, despite favorable interactions of the mono- and difluorinated anchors with the binding pocket, indicating a trade-off between energetically favorable protein-fluorine interactions and increased desolvation penalties. Taken together, the optimized carbazole scaffold provides a promising starting point for the development of high-affinity ligands to reactivate the tumor suppressor function of the p53 mutant Y220C in cancer cells.


Subject(s)
Drug Design , Fluorine/chemistry , Mutation , Sulfur/chemistry , Tumor Suppressor Protein p53/chemistry , Biophysics , Crystallography, X-Ray , Models, Molecular , Molecular Structure , Quantum Theory , Tumor Suppressor Protein p53/genetics
11.
J Med Chem ; 59(4): 1492-500, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26367539

ABSTRACT

We describe new synthetic routes developed toward a range of substituted analogues of bromo and extra-terminal (BET) bromodomain inhibitors I-BET762/JQ1 based on the triazolo-benzodiazepine scaffold. These new routes allow for the derivatization of the methoxyphenyl and chlorophenyl rings, in addition to the diazepine ternary center and the side chain methylene moiety. Substitution at the level of the side chain methylene afforded compounds targeting specifically and potently engineered BET bromodomains designed as part of a bump and hole approach. We further demonstrate that marked selectivity for the second over the first bromodomain can be achieved with an indole derivative that exploits differential interaction with an aspartate/histidine conservative substitution on the BC loop of BET bromodomains.


Subject(s)
Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Nuclear Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Triazoles/chemistry , Triazoles/pharmacology , Cell Cycle Proteins , Drug Design , Humans , Molecular Docking Simulation , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
12.
Structure ; 23(12): 2246-2255, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26636255

ABSTRACT

The destabilizing p53 cancer mutation Y220C creates an extended crevice on the surface of the protein that can be targeted by small-molecule stabilizers. Here, we identify different classes of small molecules that bind to this crevice and determine their binding modes by X-ray crystallography. These structures reveal two major conformational states of the pocket and a cryptic, transiently open hydrophobic subpocket that is modulated by Cys220. In one instance, specifically targeting this transient protein state by a pyrrole moiety resulted in a 40-fold increase in binding affinity. Molecular dynamics simulations showed that both open and closed states of this subsite were populated at comparable frequencies along the trajectories. Our data extend the framework for the design of high-affinity Y220C mutant binders for use in personalized anticancer therapy and, more generally, highlight the importance of implementing protein dynamics and hydration patterns in the drug-discovery process.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Dynamics Simulation , Tumor Suppressor Protein p53/chemistry , Amino Acid Sequence , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Mutation , Protein Binding , Protein Stability , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Chem Sci ; 6(7): 3853-3862, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-29218156

ABSTRACT

Asymmetric photochemical synthesis using circularly polarized (CP) light is theoretically attractive as a means of absolute asymmetric synthesis and postulated as an explanation for homochirality on Earth. Using an asymmetric photochemical synthesis of a dihydrohelicene as an example, we demonstrate the principle that two wavelengths of CP light can be used to control separate reactions. In doing so, a photostationary state (PSS) is set up in such a way that the enantiomeric induction intrinsic to each step can combine additively, significantly increasing the asymmetric induction possible in these reactions. Moreover, we show that the effects of this dual wavelength approach can be accurately determined by kinetic modelling of the PSS. Finally, by coupling a PSS to a thermal reaction to trap the photoproduct, we demonstrate that higher enantioselectivity can be achieved than that obtainable with single wavelength irradiation, without compromising the yield of the final product.

14.
Science ; 346(6209): 638-641, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25323695

ABSTRACT

Small molecules are useful tools for probing the biological function and therapeutic potential of individual proteins, but achieving selectivity is challenging when the target protein shares structural domains with other proteins. The Bromo and Extra-Terminal (BET) proteins have attracted interest because of their roles in transcriptional regulation, epigenetics, and cancer. The BET bromodomains (protein interaction modules that bind acetyl-lysine) have been targeted by potent small-molecule inhibitors, but these inhibitors lack selectivity for individual family members. We developed an ethyl derivative of an existing small-molecule inhibitor, I-BET/JQ1, and showed that it binds leucine/alanine mutant bromodomains with nanomolar affinity and achieves up to 540-fold selectivity relative to wild-type bromodomains. Cell culture studies showed that blockade of the first bromodomain alone is sufficient to displace a specific BET protein, Brd4, from chromatin. Expansion of this approach could help identify the individual roles of single BET proteins in human physiology and disease.


Subject(s)
Molecular Probes/chemistry , Nuclear Proteins/chemistry , Protein Engineering/methods , Transcription Factors/chemistry , Amino Acid Sequence , Azepines/chemistry , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Chromatin/chemistry , Crystallography, X-Ray , Humans , Leucine/genetics , Models, Molecular , Mutation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Structure, Tertiary , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Triazoles/chemistry , Triazoles/pharmacology
15.
Anal Biochem ; 460: 39-46, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24882269

ABSTRACT

Performing kinetic studies on protein ligand interactions provides important information on complex formation and dissociation. Beside kinetic parameters such as association rates and residence times, kinetic experiments also reveal insights into reaction mechanisms. Exploiting intrinsic tryptophan fluorescence a parallelized high-throughput Förster resonance energy transfer (FRET)-based reporter displacement assay with very low protein consumption was developed to enable the large-scale kinetic characterization of the binding of ligands to recombinant human histone deacetylases (HDACs) and a bacterial histone deacetylase-like amidohydrolase (HDAH) from Bordetella/Alcaligenes. For the binding of trichostatin A (TSA), suberoylanilide hydroxamic acid (SAHA), and two other SAHA derivatives to HDAH, two different modes of action, simple one-step binding and a two-step mechanism comprising initial binding and induced fit, were verified. In contrast to HDAH, all compounds bound to human HDAC1, HDAC6, and HDAC8 through a two-step mechanism. A quantitative view on the inhibitor-HDAC systems revealed two types of interaction, fast binding and slow dissociation. We provide arguments for the thesis that the relationship between quantitative kinetic and mechanistic information and chemical structures of compounds will serve as a valuable tool for drug optimization.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylases/metabolism , Alcaligenes/enzymology , Bordetella/enzymology , Fluorescent Dyes/metabolism , Humans , Kinetics , Ligands , Protein Binding
16.
J Mol Recognit ; 27(3): 160-72, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24446380

ABSTRACT

Thermodynamic studies on ligand-protein binding have become increasingly important in the process of drug design. In combination with structural data and molecular dynamics simulations, thermodynamic studies provide relevant information about the mode of interaction between compounds and their target proteins and therefore build a sound basis for further drug optimization. Using the example of histone deacetylases (HDACs), particularly the histone deacetylase like amidohydrolase (HDAH) from Bordetella/Alcaligenes, a novel sensitive competitive fluorescence resonance energy transfer-based binding assay was developed and the thermodynamics of interaction of both fluorescent ligands and inhibitors to histone deacetylase like amidohydrolase were investigated. The assay consumes only small amounts of valuable target proteins and is suitable for fast kinetic and mechanistic studies as well as high throughput screening applications. Binding affinity increased with increasing length of aliphatic spacers (n = 4-7) between the hydroxamate moiety and the dansyl head group of ligand probes. Van't Hoff plots revealed an optimum in enthalpy contribution to the free energy of binding for the dansyl-ligand with hexyl spacer. The selectivity in the series of dansyl-ligands against human class I HDAC1 but not class II HDACs 4 and 6 increased with the ratio of ΔH(0)/ΔG(0). The data clearly emphasize the importance of thermodynamic signatures as useful general guidance for the optimization of ligands or rational drug design.


Subject(s)
Alcaligenes/chemistry , Bacterial Proteins/chemistry , Bordetella/chemistry , Histone Deacetylase 1/chemistry , Alcaligenes/enzymology , Bacterial Proteins/metabolism , Binding Sites , Binding, Competitive , Biological Assay , Bordetella/enzymology , Dansyl Compounds/chemistry , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer , Histone Deacetylase 1/metabolism , Humans , Kinetics , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Probes/chemistry , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thermodynamics
17.
ACS Med Chem Lett ; 5(1): 23-28, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24436777

ABSTRACT

Modulation of protein-protein interactions (PPIs) with small molecules has been hampered by a lack of lucid methods capable of reliably identifying high-quality hits. In fragment screening, the low ligand efficiencies associated with PPI target sites pose significant challenges to fragment binding detection. Here, we investigate the requirements for ligand-based NMR techniques to detect rule-of-three compliant fragments that form part of known high-affinity inhibitors of the PPI between the von Hippel-Lindau protein and the alpha subunit of hypoxia-inducible factor 1 (pVHL:HIF-1α). Careful triaging allowed rescuing weak but specific binding of fragments that would otherwise escape detection at this PPI. Further structural information provided by saturation transfer difference (STD) group epitope mapping, protein-based NMR, competitive isothermal titration calorimetry (ITC), and X-ray crystallography confirmed the binding mode of the rescued fragments. Our findings have important implications for PPI druggability assessment by fragment screening as they reveal an accessible threshold for fragment detection and validation.

18.
Beilstein J Org Chem ; 9: 81-8, 2013.
Article in English | MEDLINE | ID: mdl-23400330

ABSTRACT

There has been significant interest in the bioactivity of the natural product psammaplin A, most recently as a potent and isoform selective HDAC inhibitor. Here we report our preliminary studies on thioester HDAC inhibitors derived from the active monomeric (thiol) form of psammaplin A, as a means to improve compound delivery into cells. We have discovered that such compounds exhibit both potent cytotoxicity and enzymatic inhibitory activity against recombinant HDAC1. The latter effect is surprising since previous SAR suggested that modification of the thiol functionality should detrimentally affect HDAC potency. We therefore also report our preliminary studies on the mechanism of action of this observed effect.

19.
ChemMedChem ; 8(1): 149-56, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23184734

ABSTRACT

Novel picolinamide-based histone deacetylase (HDAC) inhibitors were developed, drawing inspiration from the natural product psammaplin A. We found that the HDAC potency and isoform selectivity provided by the oxime unit of psammaplin A could be reproduced by using carefully chosen heterocyclic frameworks. The resulting (hetero)aromatic amide based compounds displayed very high potency and isoform selectivity among the HDAC family, in addition to excellent ligand efficiency relative to previously reported HDAC inhibitors. In particular, the high HDAC1 isoform selectivity provided by the chloropyridine motif represents a valuable design criterion for the development of new lead compounds and chemical probes that target HDAC1.


Subject(s)
Disulfides/chemistry , Disulfides/pharmacology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Picolinic Acids/chemistry , Picolinic Acids/pharmacology , Tyrosine/analogs & derivatives , Amides/chemistry , Amides/pharmacology , Histone Deacetylases/metabolism , Humans , Molecular Docking Simulation , Protein Isoforms/metabolism , Tyrosine/chemistry , Tyrosine/pharmacology
20.
J Med Chem ; 55(4): 1731-50, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22280363

ABSTRACT

Psammaplin A (11c) is a marine metabolite previously reported to be a potent inhibitor of two classes of epigenetic enzymes: histone deacetylases and DNA methyltransferases. The design and synthesis of a focused library based on the psammaplin A core has been carried out to probe the molecular features of this molecule responsible for its activity. By direct in vitro assay of the free thiol generated upon reduction of the dimeric psammaplin scaffold, we have unambiguously demonstrated that 11c functions as a natural prodrug, with the reduced form being highly potent against HDAC1 in vitro (IC(50) 0.9 nM). Furthermore, we have shown it to have high isoform selectivity, being 360-fold selective for HDAC1 over HDAC6 and more than 1000-fold less potent against HDAC7 and HDAC8. SAR around our focused library revealed a number of features, most notably the oxime functionality to be important to this selectivity. Many of the compounds show significant cytotoxicity in A549, MCF7, and W138 cells, with the SAR of cytotoxicity correlating to HDAC inhibition. Furthermore, compound treatment causes upregulation of histone acetylation but little effect on tubulin acetylation. Finally, we have found no evidence for 11c functioning as a DNMT inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Disulfides/pharmacology , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Prodrugs/pharmacology , Tyrosine/analogs & derivatives , Acetylation , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Crystallography, X-Ray , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Dimerization , Disulfides/chemical synthesis , Disulfides/chemistry , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histones/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Models, Molecular , Prodrugs/chemical synthesis , Prodrugs/chemistry , Structure-Activity Relationship , Tubulin/metabolism , Tyrosine/chemical synthesis , Tyrosine/chemistry , Tyrosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...