Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
ACS Infect Dis ; 8(9): 1962-1974, 2022 09 09.
Article in English | MEDLINE | ID: mdl-36037410

ABSTRACT

There is a pressing need for new medicines to prevent and treat malaria. Most antimalarial drug discovery is reliant upon phenotypic screening. However, with the development of improved target validation strategies, target-focused approaches are now being utilized. Here, we describe the development of a toolkit to support the therapeutic exploitation of a promising target, lysyl tRNA synthetase (PfKRS). The toolkit includes resistant mutants to probe resistance mechanisms and on-target engagement for specific chemotypes; a hybrid KRS protein capable of producing crystals suitable for ligand soaking, thus providing high-resolution structural information to guide compound optimization; chemical probes to facilitate pulldown studies aimed at revealing the full range of specifically interacting proteins and thermal proteome profiling (TPP); as well as streamlined isothermal TPP methods to provide unbiased confirmation of on-target engagement within a biologically relevant milieu. This combination of tools and methodologies acts as a template for the development of future target-enabling packages.


Subject(s)
Antimalarials , Lysine-tRNA Ligase , Malaria , Antimalarials/chemistry , Antimalarials/pharmacology , Drug Discovery , Humans , Lysine-tRNA Ligase/chemistry , Lysine-tRNA Ligase/genetics , Lysine-tRNA Ligase/metabolism , Plasmodium falciparum/metabolism
2.
Antimicrob Agents Chemother ; 66(1): e0153521, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34606338

ABSTRACT

Phenotypic screening identified an arylsulfonamide compound with activity against Trypanosoma cruzi, the causative agent of Chagas' disease. Comprehensive mode of action studies revealed that this compound primarily targets the T. cruzi proteasome, binding at the interface between ß4 and ß5 subunits that catalyze chymotrypsin-like activity. A mutation in the ß5 subunit of the proteasome was associated with resistance to compound 1, while overexpression of this mutated subunit also reduced susceptibility to compound 1. Further genetically engineered and in vitro-selected clones resistant to proteasome inhibitors known to bind at the ß4/ß5 interface were cross-resistant to compound 1. Ubiquitinated proteins were additionally found to accumulate in compound 1-treated epimastigotes. Finally, thermal proteome profiling identified malic enzyme as a secondary target of compound 1, although malic enzyme inhibition was not found to drive potency. These studies identify a novel pharmacophore capable of inhibiting the T. cruzi proteasome that may be exploitable for anti-chagasic drug discovery.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Chagas Disease/drug therapy , Drug Discovery , Humans , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Trypanosoma cruzi/chemistry
3.
ACS Infect Dis ; 6(5): 1044-1057, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32275825

ABSTRACT

Methionyl-tRNA synthetase (MetRS) is a chemically validated drug target in kinetoplastid parasites Trypanosoma brucei and Leishmania donovani. To date, all kinetoplastid MetRS inhibitors described bind in a similar way to an expanded methionine pocket and an adjacent, auxiliary pocket. In the current study, we have identified a structurally novel class of inhibitors containing a 4,6-diamino-substituted pyrazolopyrimidine core (the MetRS02 series). Crystallographic studies revealed that MetRS02 compounds bind to an allosteric pocket in L. major MetRS not previously described, and enzymatic studies demonstrated a noncompetitive mode of inhibition. Homology modeling of the Trypanosoma cruzi MetRS enzyme revealed key differences in the allosteric pocket between the T. cruzi and Leishmania enzymes. These provide a likely explanation for the lower MetRS02 potencies that we observed for the T. cruzi enzyme compared to the Leishmania enzyme. The identification of a new series of MetRS inhibitors and the discovery of a new binding site in kinetoplastid MetRS enzymes provide a novel strategy in the search for new therapeutics for kinetoplastid diseases.


Subject(s)
Allosteric Site , Methionine-tRNA Ligase/chemistry , Protozoan Proteins/chemistry , Trypanosoma brucei brucei/enzymology , Methionine
4.
Article in English | MEDLINE | ID: mdl-31405854

ABSTRACT

Chagas' disease, which is caused by the Trypanosoma cruzi parasite, has become a global health problem that is currently treated with poorly tolerated drugs that require prolonged dosing. Therefore, there is a clinical need for new therapeutic agents that can mitigate these issues. The phosphomannomutase (PMM) and GDP-mannose pyrophosphorylase (GDP-MP) enzymes form part of the de novo biosynthetic pathway to the nucleotide sugar GDP-mannose. This nucleotide sugar is used either directly, or indirectly via the formation of dolichol-phosphomannose, for the assembly of all mannose-containing glycoconjugates. In T. cruzi, mannose-containing glycoconjugates include the cell-surface glycoinositol-phospholipids and the glycosylphosphatidylinositol-anchored mucin-like glycoproteins that dominate the cell surface architectures of all life cycle stages. This makes PMM and GDP-MP potentially attractive targets for a drug discovery program against Chagas' disease. To assess the ligandability of these enzymes in T. cruzi, we have screened 18,117 structurally diverse compounds exploring drug-like chemical space and 16,845 small polar fragment compounds using an assay interrogating the activities of both PMM and GDP-MP enzymes simultaneously. This resulted in 48 small fragment hits, and on retesting 20 were found to be active against the enzymes. Deconvolution revealed that these were all inhibitors of T. cruzi GDP-MP, with compounds 2 and 3 acting as uncompetitive and competitive inhibitors, respectively. Based on these findings, the T. cruzi PMM and GDP-MP enzymes were deemed not ligandable and poorly ligandable, respectively, using small molecules from conventional drug discovery chemical space. This presents a significant hurdle to exploiting these enzymes as therapeutic targets for Chagas' disease.


Subject(s)
Antiprotozoal Agents/pharmacology , Mannose/metabolism , Nucleotidyltransferases/metabolism , Phosphotransferases (Phosphomutases)/metabolism , Trypanosoma cruzi/enzymology , Chagas Disease/parasitology , Drug Discovery/methods , Mannosephosphates/metabolism , Nucleotidyltransferases/genetics , Phosphotransferases (Phosphomutases)/genetics
5.
Article in English | MEDLINE | ID: mdl-31307977

ABSTRACT

Chagas' disease, caused by the protozoan parasite Trypanosoma cruzi, is a potentially life-threatening condition that has become a global issue. Current treatment is limited to two medicines that require prolonged dosing and are associated with multiple side effects, which often lead to treatment discontinuation and failure. One way to address these shortcomings is through target-based drug discovery on validated T. cruzi protein targets. One such target is the proteasome, which plays a crucial role in protein degradation and turnover through chymotrypsin-, trypsin-, and caspase-like catalytic activities. In order to initiate a proteasome drug discovery program, we isolated proteasomes from T. cruzi epimastigotes and characterized their activity using a commercially available glow-like luminescence-based assay. We developed a high-throughput biochemical assay for the chymotrypsin-like activity of the T. cruzi proteasome, which was found to be sensitive, specific, and robust but prone to luminescence technology interference. To mitigate this, we also developed a counterscreen assay that identifies potential interferers at the levels of both the luciferase enzyme reporter and the mechanism responsible for a glow-like response. Interestingly, we also found that the peptide substrate for chymotrypsin-like proteasome activity was not specific and was likely partially turned over by other catalytic sites of the protein. Finally, we utilized these biochemical tools to screen 18,098 compounds, exploring diverse drug-like chemical space, which allowed us to identify 39 hits that were active in the primary screening assay and inactive in the counterscreen assay.


Subject(s)
High-Throughput Screening Assays/methods , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Cell-Free System , Luminescence , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , Reproducibility of Results , Trypanosoma cruzi/chemistry
6.
FEBS J ; 286(22): 4509-4524, 2019 11.
Article in English | MEDLINE | ID: mdl-31260169

ABSTRACT

Burkholderia pseudomallei is a serious, difficult to treat Gram-negative pathogen and an increase in the occurrence of drug-resistant strains has been detected. We have directed efforts to identify and to evaluate potential drug targets relevant to treatment of infection by B. pseudomallei. We have selected and characterised the essential enzyme d-alanine-d-alanine ligase (BpDdl), required for the ATP-assisted biosynthesis of a peptidoglycan precursor. A recombinant supply of protein supported high-resolution crystallographic and biophysical studies with ligands (AMP and AMP+d-Ala-d-Ala), and comparisons with orthologues enzymes suggest a ligand-induced conformational change occurring that might be relevant to the catalytic cycle. The detailed biochemical characterisation of the enzyme, development and optimisation of ligand binding assays supported the search for novel inhibitors by screening of selected compound libraries. In a similar manner to that observed previously in other studies, we note a paucity of hits that are worth follow-up and then in combination with a computational analysis of the active site, we conclude that this ligase represents a difficult target for drug discovery. Nevertheless, our reagents, protocols and data can underpin future efforts exploiting more diverse chemical libraries and structure-based approaches.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Burkholderia pseudomallei/enzymology , Enzyme Inhibitors/pharmacology , Peptide Synthases/chemistry , Adenosine Monophosphate/metabolism , Alanine/metabolism , Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/metabolism , Protein Binding , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
7.
PLoS One ; 14(5): e0217828, 2019.
Article in English | MEDLINE | ID: mdl-31150492

ABSTRACT

The discovery of 20 unconventional kinetochore proteins in Trypanosoma brucei has opened a new and interesting area of evolutionary research to study a biological process previously thought to be highly conserved in all eukaryotes. In addition, the discovery of novel proteins involved in a critical cellular process provides an opportunity to exploit differences between kinetoplastid and human kinetochore proteins to develop therapeutics for diseases caused by kinetoplastid parasites. Consequently, we identified two of the unconventional kinetochore proteins as key targets (the highly related kinases KKT10 and KKT19). Recombinant T. brucei KKT19 (TbKKT19) protein was produced, a peptide substrate phosphorylated by TbKKT19 identified (KKLRRTLSVA), Michaelis constants for KKLRRTLSVA and ATP were determined (179 µM and 102 µM respectively) and a robust high-throughput compatible biochemical assay developed. This biochemical assay was validated pharmacologically with inhibition by staurosporine and hypothemycin (IC50 values of 288 nM and 65 nM respectively). Surprisingly, a subsequent high-throughput screen of a kinase-relevant compound library (6,624 compounds) yielded few hits (8 hits; final hit rate 0.12%). The low hit rate observed was unusual for a kinase target, particularly when screened against a compound library enriched with kinase hinge binding scaffolds. In an attempt to understand the low hit rate a TbKKT19 homology model, based on human cdc2-like kinase 1 (CLK1), was generated. Analysis of the TbKKT19 sequence and structure revealed no obvious features that could explain the low hit rates. Further work will therefore be necessary to explore this unique kinetochore kinase as well as to assess whether the few hits identified can be developed into tool molecules or new drugs.


Subject(s)
Peptides/antagonists & inhibitors , Phosphotransferases/antagonists & inhibitors , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/diet therapy , Animals , Drug Discovery , High-Throughput Screening Assays , Humans , Kinetochores/drug effects , Kinetochores/enzymology , Peptides/chemistry , Phosphotransferases/chemistry , Phosphotransferases/genetics , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Staurosporine/pharmacology , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/parasitology , Zearalenone/analogs & derivatives , Zearalenone/pharmacology
8.
ACS Infect Dis ; 5(1): 111-122, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30380837

ABSTRACT

Visceral leishmaniasis (VL), caused by the protozoan parasites Leishmania donovani and L. infantum, is responsible for ∼30 000 deaths annually. Available treatments are inadequate, and there is a pressing need for new therapeutics. N-Myristoyltransferase (NMT) remains one of the few genetically validated drug targets in these parasites. Here, we sought to pharmacologically validate this enzyme in Leishmania. A focused set of 1600 pyrazolyl sulfonamide compounds was screened against L. major NMT in a robust high-throughput biochemical assay. Several potent inhibitors were identified with marginal selectivity over the human enzyme. There was little correlation between the enzyme potency of these inhibitors and their cellular activity against L. donovani axenic amastigotes, and this discrepancy could be due to poor cellular uptake due to the basicity of these compounds. Thus, a series of analogues were synthesized with less basic centers. Although most of these compounds continued to suffer from relatively poor antileishmanial activity, our most potent inhibitor of LmNMT (DDD100097, K i of 0.34 nM) showed modest activity against L. donovani intracellular amastigotes (EC50 of 2.4 µM) and maintained a modest therapeutic window over the human enzyme. Two unbiased approaches, namely, screening against our cosmid-based overexpression library and thermal proteome profiling (TPP), confirm that DDD100097 (compound 2) acts on-target within parasites. Oral dosing with compound 2 resulted in a 52% reduction in parasite burden in our mouse model of VL. Thus, NMT is now a pharmacologically validated target in Leishmania. The challenge in finding drug candidates remains to identify alternative strategies to address the drop-off in activity between enzyme inhibition and in vitro activity while maintaining sufficient selectivity over the human enzyme, both issues that continue to plague studies in this area.


Subject(s)
Acyltransferases/antagonists & inhibitors , Antiprotozoal Agents/pharmacology , Drug Discovery , Leishmania donovani/drug effects , Pyrazoles/chemistry , Pyrazoles/pharmacology , Animals , Cosmids , Female , High-Throughput Screening Assays , Humans , Leishmaniasis, Visceral/drug therapy , Mice , Mice, Inbred BALB C , Parasite Load , Proteome/analysis , Proteomics
9.
Nat Rev Microbiol ; 16(11): 714, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30206344

ABSTRACT

The structures of nifurtimox in Table 1 were incorrect and have been updated in the pdf and online. The authors apologize for any confusion caused.

10.
J Med Chem ; 61(18): 8374-8389, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30207721

ABSTRACT

Crystallography has guided the hybridization of two series of Trypanosoma brucei N-myristoyltransferase (NMT) inhibitors, leading to a novel highly selective series. The effect of combining the selectivity enhancing elements from two pharmacophores is shown to be additive and has led to compounds that have greater than 1000-fold selectivity for TbNMT vs HsNMT. Further optimization of the hybrid series has identified compounds with significant trypanocidal activity capable of crossing the blood-brain barrier. By using CF-1 mdr1a deficient mice, we were able to demonstrate full cures in vivo in a mouse model of stage 2 African sleeping sickness. This and previous work provides very strong validation for NMT as a drug target for human African trypanosomiasis in both the peripheral and central nervous system stages of disease.


Subject(s)
Acyltransferases/antagonists & inhibitors , Blood-Brain Barrier/drug effects , Drug Design , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , Animals , Cell Survival , Female , Humans , Mice , Mice, Inbred Strains , Models, Molecular , Molecular Structure , Protein Conformation , Structure-Activity Relationship , Trypanosomiasis, African/microbiology
11.
J Med Chem ; 60(23): 9790-9806, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29125744

ABSTRACT

N-Myristoyltransferase (NMT) represents a promising drug target within the parasitic protozoa Trypanosoma brucei (T. brucei), the causative agent for human African trypanosomiasis (HAT) or sleeping sickness. We have previously validated T. brucei NMT as a promising druggable target for the treatment of HAT in both stages 1 and 2 of the disease. We report on the use of the previously reported DDD85646 (1) as a starting point for the design of a class of potent, brain penetrant inhibitors of T. brucei NMT.


Subject(s)
Acyltransferases/antagonists & inhibitors , Aminopyridines/chemistry , Aminopyridines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , Acyltransferases/metabolism , Aminopyridines/chemical synthesis , Aminopyridines/pharmacokinetics , Animals , Brain/metabolism , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Mice , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/pharmacokinetics , Trypanosomiasis, African/metabolism
12.
ACS Infect Dis ; 3(10): 718-727, 2017 10 13.
Article in English | MEDLINE | ID: mdl-28967262

ABSTRACT

Methionyl-tRNA synthetase (MetRS) has been chemically validated as a drug target in the kinetoplastid parasite Trypanosoma brucei. In the present study, we investigate the validity of this target in the related trypanosomatid Leishmania donovani. Following development of a robust high-throughput compatible biochemical assay, a compound screen identified DDD806905 as a highly potent inhibitor of LdMetRS (Ki of 18 nM). Crystallography revealed this compound binds to the methionine pocket of MetRS with enzymatic studies confirming DDD806905 displays competitive inhibition with respect to methionine and mixed inhibition with respect to ATP binding. DDD806905 showed activity, albeit with different levels of potency, in various Leishmania cell-based viability assays, with on-target activity observed in both Leishmania promastigote cell assays and a Leishmania tarentolae in vitro translation assay. Unfortunately, this compound failed to show efficacy in an animal model of leishmaniasis. We investigated the potential causes for the discrepancies in activity observed in different Leishmania cell assays and the lack of efficacy in the animal model and found that high protein binding as well as sequestration of this dibasic compound into acidic compartments may play a role. Despite medicinal chemistry efforts to address the dibasic nature of DDD806905 and analogues, no progress could be achieved with the current chemical series. Although DDD806905 is not a developable antileishmanial compound, MetRS remains an attractive antileishmanial drug target.


Subject(s)
Antiprotozoal Agents/pharmacology , Enzyme Inhibitors/pharmacology , Leishmania donovani/enzymology , Methionine-tRNA Ligase/antagonists & inhibitors , Methionine-tRNA Ligase/metabolism , Drug Discovery , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Leishmania donovani/drug effects , Molecular Structure
14.
Nat Rev Microbiol ; 15(4): 217-231, 2017 Feb 27.
Article in English | MEDLINE | ID: mdl-28239154

ABSTRACT

The WHO recognizes human African trypanosomiasis, Chagas disease and the leishmaniases as neglected tropical diseases. These diseases are caused by parasitic trypanosomatids and range in severity from mild and self-curing to near invariably fatal. Public health advances have substantially decreased the effect of these diseases in recent decades but alone will not eliminate them. In this Review, we discuss why new drugs against trypanosomatids are required, approaches that are under investigation to develop new drugs and why the drug discovery pipeline remains essentially unfilled. In addition, we consider the important challenges to drug discovery strategies and the new technologies that can address them. The combination of new drugs, new technologies and public health initiatives is essential for the management, and hopefully eventual elimination, of trypanosomatid diseases from the human population.

15.
ChemMedChem ; 10(11): 1821-36, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26395087

ABSTRACT

The enzyme N-myristoyltransferase (NMT) from Trypanosoma brucei has been validated both chemically and biologically as a potential drug target for human African trypanosomiasis. We previously reported the development of some very potent compounds based around a pyrazole sulfonamide series, derived from a high-throughput screen. Herein we describe work around thiazolidinone and benzomorpholine scaffolds that were also identified in the screen. An X-ray crystal structure of the thiazolidinone hit in Leishmania major NMT showed the compound bound in the previously reported active site, utilising a novel binding mode. This provides potential for further optimisation. The benzomorpholinone was also found to bind in a similar region. Using an X-ray crystallography/structure-based design approach, the benzomorpholinone series was further optimised, increasing activity against T. brucei NMT by >1000-fold. A series of trypanocidal compounds were identified with suitable in vitro DMPK properties, including CNS exposure for further development. Further work is required to increase selectivity over the human NMT isoform and activity against T. brucei.


Subject(s)
Acyltransferases/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Trypanosoma brucei brucei/enzymology , Acyltransferases/metabolism , Binding Sites/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Trypanosoma brucei brucei/drug effects
16.
ACS Chem Biol ; 10(6): 1425-34, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25706802

ABSTRACT

Treatment of filamentous fungal infections relies on a limited repertoire of antifungal agents. Compounds possessing novel modes of action are urgently required. N-myristoylation is a ubiquitous modification of eukaryotic proteins. The enzyme N-myristoyltransferase (NMT) has been considered a potential therapeutic target in protozoa and yeasts. Here, we show that the filamentous fungal pathogen Aspergillus fumigatus possesses an active NMT enzyme that is essential for survival. Surprisingly, partial repression of the gene revealed downstream effects of N-myristoylation on cell wall morphology. Screening a library of inhibitors led to the discovery of a pyrazole sulphonamide compound that inhibits the enzyme and is fungicidal under partially repressive nmt conditions. Together with a crystallographic complex showing the inhibitor binding in the peptide substrate pocket, we provide evidence of NMT being a potential drug target in A. fumigatus.


Subject(s)
Acyltransferases/antagonists & inhibitors , Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Cell Wall/drug effects , Fungal Proteins/antagonists & inhibitors , Protein Processing, Post-Translational , Acyltransferases/chemistry , Acyltransferases/metabolism , Aminopyridines/chemistry , Aminopyridines/pharmacology , Antifungal Agents/chemistry , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/genetics , Aspergillus fumigatus/growth & development , Catalytic Domain , Cell Wall/chemistry , Cell Wall/enzymology , Crystallography, X-Ray , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Kinetics , Microbial Sensitivity Tests , Protein Binding , Protein Structure, Secondary , Pyrazoles/chemistry , Pyrazoles/pharmacology , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology
17.
J Med Chem ; 57(23): 9855-69, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25412409

ABSTRACT

Trypanosoma brucei N-myristoyltransferase (TbNMT) is an attractive therapeutic target for the treatment of human African trypanosomiasis (HAT). From previous studies, we identified pyrazole sulfonamide, DDD85646 (1), a potent inhibitor of TbNMT. Although this compound represents an excellent lead, poor central nervous system (CNS) exposure restricts its use to the hemolymphatic form (stage 1) of the disease. With a clear clinical need for new drug treatments for HAT that address both the hemolymphatic and CNS stages of the disease, a chemistry campaign was initiated to address the shortfalls of this series. This paper describes modifications to the pyrazole sulfonamides which markedly improved blood-brain barrier permeability, achieved by reducing polar surface area and capping the sulfonamide. Moreover, replacing the core aromatic with a flexible linker significantly improved selectivity. This led to the discovery of DDD100097 (40) which demonstrated partial efficacy in a stage 2 (CNS) mouse model of HAT.


Subject(s)
Acyltransferases/antagonists & inhibitors , Pyrazoles/chemical synthesis , Sulfonamides/chemical synthesis , Trypanocidal Agents/chemical synthesis , Trypanosomiasis, African/drug therapy , Aminopyridines/chemistry , Animals , Blood-Brain Barrier/drug effects , Central Nervous System/drug effects , Female , Humans , Inhibitory Concentration 50 , Mice , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects
18.
Biochem J ; 459(2): 323-32, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24444291

ABSTRACT

Co- and post-translational N-myristoylation is known to play a role in the correct subcellular localization of specific proteins in eukaryotes. The enzyme that catalyses this reaction, NMT (N-myristoyltransferase), has been pharmacologically validated as a drug target in the African trypanosome, Trypanosoma brucei. In the present study, we evaluate NMT as a potential drug target in Trypanosoma cruzi, the causative agent of Chagas' disease, using chemical and genetic approaches. Replacement of both allelic copies of TcNMT (T. cruzi NMT) was only possible in the presence of a constitutively expressed ectopic copy of the gene, indicating that this gene is essential for survival of T. cruzi epimastigotes. The pyrazole sulphonamide NMT inhibitor DDD85646 is 13-23-fold less potent against recombinant TcNMT than TbNMT (T. brucei NMT), with Ki values of 12.7 and 22.8 nM respectively, by scintillation proximity or coupled assay methods. DDD85646 also inhibits growth of T. cruzi epimastigotes (EC50=6.9 µM), but is ~1000-fold less potent than that reported for T. brucei. On-target activity is demonstrated by shifts in cell potency in lines that over- and under-express NMT and by inhibition of intracellular N-myristoylation of several proteins in a dose-dependent manner. Collectively, our findings suggest that N-myristoylation is an essential and druggable target in T. cruzi.


Subject(s)
Acyltransferases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Trypanosoma cruzi/enzymology , Acyltransferases/genetics , Aminopyridines , Animals , Chlorocebus aethiops , Cloning, Molecular , Gene Deletion , Kinetics , Organisms, Genetically Modified , Recombinant Proteins , Sulfonamides , Vero Cells
19.
PLoS Biol ; 11(6): e1001593, 2013.
Article in English | MEDLINE | ID: mdl-23824517

ABSTRACT

The drug molecule PTC124 (Ataluren) has been described as a read-through agent, capable of suppressing premature termination codons (PTCs) and restoring functional protein production from genes disrupted by nonsense mutations. Following the discovery of PTC124 there was some controversy regarding its mechanism of action with two reports attributing its activity to an off-target effect on the Firefly luciferase (FLuc) reporter used in the development of the molecule. Despite questions remaining as to its mechanism of action, development of PTC124 continued into the clinic and it is being actively pursued as a potential nonsense mutation therapy. To thoroughly test the ability of PTC124 to read through nonsense mutations, we conducted a detailed assessment comparing the efficacy of PTC124 with the classical aminoglycoside antibiotic read-through agent geneticin (G418) across a diverse range of in vitro reporter assays. We can confirm the off-target FLuc activity of PTC124 but found that, while G418 exhibits varying activity in every read-through assay, there is no evidence of activity for PTC124.


Subject(s)
Biological Assay , Codon, Nonsense/genetics , Genes, Reporter , Oxadiazoles/pharmacology , Animals , Cell Line , Collagen Type VII/metabolism , Gentamicins/pharmacology , Humans , Luciferases, Firefly/metabolism , Transfection , beta-Galactosidase/metabolism
20.
ACS Chem Biol ; 8(2): 387-96, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23138692

ABSTRACT

Glucose-1-phosphate thymidylyltransferase (RmlA) catalyzes the condensation of glucose-1-phosphate (G1P) with deoxy-thymidine triphosphate (dTTP) to yield dTDP-d-glucose and pyrophosphate. This is the first step in the l-rhamnose biosynthetic pathway. l-Rhamnose is an important component of the cell wall of many microorganisms, including Mycobacterium tuberculosis and Pseudomonas aeruginosa. Here we describe the first nanomolar inhibitors of P. aeruginosa RmlA. These thymine analogues were identified by high-throughput screening and subsequently optimized by a combination of protein crystallography, in silico screening, and synthetic chemistry. Some of the inhibitors show inhibitory activity against M. tuberculosis. The inhibitors do not bind at the active site of RmlA but bind at a second site remote from the active site. Despite this, the compounds act as competitive inhibitors of G1P but with high cooperativity. This novel behavior was probed by structural analysis, which suggests that the inhibitors work by preventing RmlA from undergoing the conformational change key to its ordered bi-bi mechanism.


Subject(s)
Enzyme Inhibitors/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Pseudomonas aeruginosa/enzymology , Thymine/pharmacology , Allosteric Site/drug effects , Binding, Competitive/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Models, Molecular , Molecular Structure , Nucleotidyltransferases/metabolism , Structure-Activity Relationship , Thymine/analogs & derivatives , Thymine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...