Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 559(7715): 498-506, 2018 07.
Article in English | MEDLINE | ID: mdl-30046073

ABSTRACT

Infectious tropical diseases have a huge effect in terms of mortality and morbidity, and impose a heavy economic burden on affected countries. These diseases predominantly affect the world's poorest people. Currently available drugs are inadequate for the majority of these diseases, and there is an urgent need for new treatments. This Review discusses some of the challenges involved in developing new drugs to treat these diseases and highlights recent progress. While there have been notable successes, there is still a long way to go.


Subject(s)
Drug Discovery/trends , Infections/drug therapy , Tropical Climate , Tropical Medicine/trends , Animals , Coinfection , Humans , Infections/microbiology , Infections/parasitology , Infections/virology
3.
Nature ; 560(7717): 192-197, 2018 08.
Article in English | MEDLINE | ID: mdl-30046105

ABSTRACT

Visceral leishmaniasis causes considerable mortality and morbidity in many parts of the world. There is an urgent need for the development of new, effective treatments for this disease. Here we describe the development of an anti-leishmanial drug-like chemical series based on a pyrazolopyrimidine scaffold. The leading compound from this series (7, DDD853651/GSK3186899) is efficacious in a mouse model of visceral leishmaniasis, has suitable physicochemical, pharmacokinetic and toxicological properties for further development, and has been declared a preclinical candidate. Detailed mode-of-action studies indicate that compounds from this series act principally by inhibiting the parasite cdc-2-related kinase 12 (CRK12), thus defining a druggable target for visceral leishmaniasis.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Leishmania donovani/drug effects , Leishmania donovani/enzymology , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/parasitology , Molecular Targeted Therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Humans , Mice , Molecular Docking Simulation , Proteome/drug effects , Proteomics , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Reproducibility of Results , Substrate Specificity
4.
Nucleic Acids Res ; 49(9): 5351-5368, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33885823

ABSTRACT

Tuberculosis, caused by Mycobacterium tuberculosis, responsible for ∼1.5 million fatalities in 2018, is the deadliest infectious disease. Global spread of multidrug resistant strains is a public health threat, requiring new treatments. Aminoacyl-tRNA synthetases are plausible candidates as potential drug targets, because they play an essential role in translating the DNA code into protein sequence by attaching a specific amino acid to their cognate tRNAs. We report structures of M. tuberculosis Phe-tRNA synthetase complexed with an unmodified tRNAPhe transcript and either L-Phe or a nonhydrolyzable phenylalanine adenylate analog. High-resolution models reveal details of two modes of tRNA interaction with the enzyme: an initial recognition via indirect readout of anticodon stem-loop and aminoacylation ready state involving interactions of the 3' end of tRNAPhe with the adenylate site. For the first time, we observe the protein gate controlling access to the active site and detailed geometry of the acyl donor and tRNA acceptor consistent with accepted mechanism. We biochemically validated the inhibitory potency of the adenylate analog and provide the most complete view of the Phe-tRNA synthetase/tRNAPhe system to date. The presented topography of amino adenylate-binding and editing sites at different stages of tRNA binding to the enzyme provide insights for the rational design of anti-tuberculosis drugs.


Subject(s)
Mycobacterium tuberculosis/enzymology , Phenylalanine-tRNA Ligase/chemistry , RNA, Transfer, Phe/chemistry , Transfer RNA Aminoacylation , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/metabolism , Humans , Ligands , Models, Molecular , Mycobacterium tuberculosis/genetics , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/metabolism , Phenylalanine-tRNA Ligase/metabolism , Protein Binding , RNA, Transfer, Phe/metabolism , Thermus thermophilus/enzymology
5.
Antimicrob Agents Chemother ; 66(6): e0023722, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35647647

ABSTRACT

Artemisinin-based combination therapies have been crucial in driving down the global burden of malaria, the world's largest parasitic killer. However, their efficacy is now threatened by the emergence of resistance in Southeast Asia and sub-Saharan Africa. Thus, there is a pressing need to develop new antimalarials with diverse mechanisms of action. One area of Plasmodium metabolism that has recently proven rich in exploitable antimalarial targets is protein synthesis, with a compound targeting elongation factor 2 now in clinical development and inhibitors of several aminoacyl-tRNA synthetases in lead optimization. Given the promise of these components of translation as viable drug targets, we rationalized that an assay containing all functional components of translation would be a valuable tool for antimalarial screening and drug discovery. Here, we report the development and validation of an assay platform that enables specific inhibitors of Plasmodium falciparum translation (PfIVT) to be identified. The primary assay in this platform monitors the translation of a luciferase reporter in a P. falciparum lysate-based expression system. Hits identified in this primary assay are assessed in a counterscreen assay that enables false positives that directly interfere with the luciferase to be triaged. The remaining hit compounds are then assessed in an equivalent human IVT assay. This platform of assays was used to screen MMV's Pandemic and Pathogen Box libraries, identifying several selective inhibitors of protein synthesis. We believe this new high-throughput screening platform has the potential to greatly expedite the discovery of antimalarials that act via this highly desirable mechanism of action.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria , Antimalarials/pharmacology , Antimalarials/therapeutic use , Drug Discovery , High-Throughput Screening Assays , Humans , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Plasmodium falciparum/genetics
6.
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
7.
Hum Reprod ; 37(3): 466-475, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35048946

ABSTRACT

STUDY QUESTION: Can a high-throughput screening (HTS) platform facilitate male fertility drug discovery? SUMMARY ANSWER: An HTS platform identified a large number of compounds that enhanced sperm motility. WHAT IS KNOWN ALREADY: Several efforts to find small molecules modulating sperm function have been performed but none have used high-throughput technology. STUDY DESIGN, SIZE, DURATION: Healthy donor semen samples were used and samples were pooled (3-5 donors per pool). Primary screening was performed singly; dose-response screening was performed in duplicate (using independent donor pools). PARTICIPANTS/MATERIALS, SETTING, METHODS: Spermatozoa isolated from healthy donors were prepared by density gradient centrifugation and incubated in 384-well plates with compounds (6.25 µM) to identify those compounds with enhancing effects on motility. Approximately 17 000 compounds from the libraries, ReFRAME, Prestwick, Tocris, LOPAC, CLOUD and MMV Pathogen Box, were screened. Dose-response experiments of screening hits were performed to confirm the enhancing effect on sperm motility. Experiments were performed in a university setting. MAIN RESULTS AND THE ROLE OF CHANCE: From our primary single concentration screening, 105 compounds elicited an enhancing effect on sperm motility compared to dimethylsulphoxide-treated wells. Confirmed enhancing compounds were grouped based on their annotated targets/target classes. A major target class, phosphodiesterase inhibitors, were identified, in particular PDE10A inhibitors as well as number of compounds not previously known to enhance human sperm motility, such as those related to GABA signalling. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Although this approach provides data about the activity of the compound, it is only a starting point. For example, further substantive experiments are necessary to provide a more comprehensive picture of each compound's activity, the effect on the kinetics of the cell populations and subpopulations, and their potential mechanisms of action. Compounds have been tested with prepared donor spermatozoa, incubated under non-capacitating conditions, and only incubated with compounds for a relatively short period of time. Therefore, the effect of compounds under different conditions, for example in whole semen, for longer incubation times, or using samples from patient groups, may be different and require further study. All experiments were performed in vitro. WIDER IMPLICATIONS OF THE FINDINGS: This phenotypic screening assay identified a large number of compounds that increased sperm motility. In addition to furthering our understanding of human sperm function, for example identifying new avenues for discovery, we highlight potential compounds as promising start-point for a medicinal chemistry programme for potential enhancement of male fertility. Moreover, with disclosure of the results of screening, we present a substantial resource to inform further work in the field. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the Bill and Melinda Gates Foundation, Scottish Funding Council and Scottish Universities Life Science Alliance. C.L.R.B. is Editor for RBMO. C.L.R.B. receives funding from Chief Scientists Office (Scotland), ESHRE and Genus PLC, consulting fees from Exscientia and lecture fees from Cooper Surgical and Ferring. S.M.d.S. is an Associate Editor of Human Reproduction, and an Associate Editor of Reproduction and Fertility. S.M.d.S. receives funding from Cooper Surgical and British Dietetic Society. No other authors declared a COI.


Subject(s)
Infertility, Male , Sperm Motility , Fertility , High-Throughput Screening Assays , Humans , Infertility, Male/drug therapy , Male , Phosphoric Diester Hydrolases/pharmacology , Phosphoric Diester Hydrolases/therapeutic use , Spermatozoa
8.
Proc Natl Acad Sci U S A ; 116(19): 9318-9323, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30962368

ABSTRACT

Visceral leishmaniasis (VL), caused by the protozoan parasites Leishmania donovani and Leishmania infantum, is one of the major parasitic diseases worldwide. There is an urgent need for new drugs to treat VL, because current therapies are unfit for purpose in a resource-poor setting. Here, we describe the development of a preclinical drug candidate, GSK3494245/DDD01305143/compound 8, with potential to treat this neglected tropical disease. The compound series was discovered by repurposing hits from a screen against the related parasite Trypanosoma cruzi Subsequent optimization of the chemical series resulted in the development of a potent cidal compound with activity against a range of clinically relevant L. donovani and L. infantum isolates. Compound 8 demonstrates promising pharmacokinetic properties and impressive in vivo efficacy in our mouse model of infection comparable with those of the current oral antileishmanial miltefosine. Detailed mode of action studies confirm that this compound acts principally by inhibition of the chymotrypsin-like activity catalyzed by the ß5 subunit of the L. donovani proteasome. High-resolution cryo-EM structures of apo and compound 8-bound Leishmania tarentolae 20S proteasome reveal a previously undiscovered inhibitor site that lies between the ß4 and ß5 proteasome subunits. This induced pocket exploits ß4 residues that are divergent between humans and kinetoplastid parasites and is consistent with all of our experimental and mutagenesis data. As a result of these comprehensive studies and due to a favorable developability and safety profile, compound 8 is being advanced toward human clinical trials.


Subject(s)
Antiprotozoal Agents/administration & dosage , Leishmania donovani/drug effects , Leishmania infantum/drug effects , Leishmaniasis, Visceral/diagnostic imaging , Proteasome Inhibitors/administration & dosage , Protozoan Proteins/antagonists & inhibitors , Animals , Antiprotozoal Agents/chemistry , Binding Sites , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Leishmania donovani/chemistry , Leishmania donovani/enzymology , Leishmania infantum/chemistry , Leishmania infantum/enzymology , Leishmaniasis, Visceral/parasitology , Male , Mice , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Protein Conformation , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
9.
Reproduction ; 163(1): R1-R9, 2021 12 27.
Article in English | MEDLINE | ID: mdl-34882578

ABSTRACT

Despite recent advances in male reproductive health research, there remain many elements of male infertility where our understanding is incomplete. Consequently, diagnostic tools and treatments for men with sperm dysfunction, other than medically assisted reproduction, are limited. On the other hand, the gaps in our knowledge of the mechanisms which underpin sperm function have hampered the development of male non-hormonal contraceptives. The study of mature spermatozoa is inherently difficult. They are a unique and highly specialised cell type which does not actively transcribe or translate proteins and cannot be cultured for long periods of time or matured in vitro. One large-scale approach to both increasing the understanding of sperm function and the discovery and development of compounds that can modulate sperm function is to directly observe responses to compounds with phenotypic screening techniques. These target agnostic approaches can be developed into high-throughput screening platforms with the potential to drastically increase advances in the field. Here, we discuss the rationale and development of high-throughput phenotypic screening platforms for mature human spermatozoa and the multiple potential applications these present, as well as the current limitations and leaps in our understanding and the capabilities needed to overcome them. Further development and use of these technologies could lead to the identification of compounds which positively or negatively affect sperm cell motility or function or novel platforms for toxicology or environmental chemical testing among other applications. Ultimately, each of these potential applications is also likely to increase the understanding within the field of sperm biology.


Subject(s)
High-Throughput Screening Assays , Infertility, Male , Humans , Infertility, Male/metabolism , Male , Sperm Motility , Spermatozoa/metabolism
10.
J Org Chem ; 86(17): 11333-11340, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34351743

ABSTRACT

A selection of 3,4-diaminoindoles were required for a recent drug discovery project. To this end, a 10-step synthesis was developed from 4-nitroindole. This synthesis was subsequently adapted and used to synthesize 3,5-; 3,6-; and 3,7-diaminoindoles from the corresponding 5-, 6-, or 7-nitroindole. These novel intermediates feature orthogonal protecting groups that allow them to be further diversified. This is the first reported synthesis of these types of compounds.


Subject(s)
Indoles
11.
J Comput Aided Mol Des ; 35(10): 1025-1036, 2021 10.
Article in English | MEDLINE | ID: mdl-34458939

ABSTRACT

Water molecules play a crucial role in protein-ligand binding, and many tools exist that aim to predict the position and relative energies of these important, but challenging participants of biomolecular recognition. The available tools are, in general, capable of predicting the location of water molecules. However, predicting the effects of their displacement is still very challenging. In this work, a linear-scaling quantum mechanics-based approach was used to assess water network energetics and the changes in network stability upon ligand structural modifications. This approach offers a valuable way to improve understanding of SAR data and help guide compound design.


Subject(s)
Proteins/metabolism , Thermodynamics , Water/chemistry , Binding Sites , Ligands , Models, Molecular , Protein Binding , Proteins/chemistry , Water/metabolism
12.
Proc Natl Acad Sci U S A ; 115(38): 9616-9621, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30185555

ABSTRACT

African trypanosomes cause lethal and neglected tropical diseases, known as sleeping sickness in humans and nagana in animals. Current therapies are limited, but fortunately, promising therapies are in advanced clinical and veterinary development, including acoziborole (AN5568 or SCYX-7158) and AN11736, respectively. These benzoxaboroles will likely be key to the World Health Organization's target of disease control by 2030. Their mode of action was previously unknown. We have developed a high-coverage overexpression library and use it here to explore drug mode of action in Trypanosoma brucei Initially, an inhibitor with a known target was used to select for drug resistance and to test massive parallel library screening and genome-wide mapping; this effectively identified the known target and validated the approach. Subsequently, the overexpression screening approach was used to identify the target of the benzoxaboroles, Cleavage and Polyadenylation Specificity Factor 3 (CPSF3, Tb927.4.1340). We validated the CPSF3 endonuclease as the target, using independent overexpression strains. Knockdown provided genetic validation of CPSF3 as essential, and GFP tagging confirmed the expected nuclear localization. Molecular docking and CRISPR-Cas9-based editing demonstrated how acoziborole can specifically block the active site and mRNA processing by parasite, but not host CPSF3. Thus, our findings provide both genetic and chemical validation for CPSF3 as an important drug target in trypanosomes and reveal inhibition of mRNA maturation as the mode of action of the trypanocidal benzoxaboroles. Understanding the mechanism of action of benzoxaborole-based therapies can assist development of improved therapies, as well as the prediction and monitoring of resistance, if or when it arises.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/physiology , Trypanosomiasis, African/prevention & control , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Boron Compounds/pharmacology , Boron Compounds/therapeutic use , CRISPR-Cas Systems , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Drug Resistance/drug effects , Drug Resistance/genetics , Gene Knockdown Techniques , Gene Library , High-Throughput Screening Assays/methods , Humans , Molecular Docking Simulation , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Processing, Post-Transcriptional/drug effects , RNA, Messenger/metabolism , RNA, Protozoan/metabolism , Trypanocidal Agents/therapeutic use , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/transmission , Trypanosomiasis, African/veterinary , Valine/analogs & derivatives , Valine/pharmacology , Valine/therapeutic use
13.
Bioorg Med Chem ; 27(10): 2066-2074, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30975501

ABSTRACT

Many drugs currently used are covalent inhibitors and irreversibly inhibit their targets. Most of these were discovered through serendipity. Covalent inhibitions can have many advantages from a pharmacokinetic perspective. However, until recently most organisations have shied away from covalent compound design due to fears of non-specific inhibition of off-target proteins leading to toxicity risks. However, there has been a renewed interest in covalent modifiers as potential drugs, as it possible to get highly selective compounds. It is therefore important to know how reactive a warhead is and to be able to select the least reactive warhead possible to avoid toxicity. A robust NMR based assay was developed and used to measure the reactivity of a variety of covalent warheads against serine and cysteine - the two most common targets for covalent drugs. A selection of these warheads also had their reactivity measured against threonine, tyrosine, lysine, histidine and arginine to better understand our ability to target non-traditional residues. The reactivity was also measured at various pHs to assess what effect the environment in the active site would have on these reactions. The reactivity of a covalent modifier was found to be very dependent on the amino acid residue.


Subject(s)
Amino Acids/chemistry , Pharmaceutical Preparations/chemistry , Amino Acids/metabolism , Cysteine/chemistry , Cysteine/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Pharmaceutical Preparations/chemical synthesis , Pharmaceutical Preparations/metabolism , Serine/chemistry , Serine/metabolism
14.
Tetrahedron Lett ; 60(18): 1243-1247, 2019 May 02.
Article in English | MEDLINE | ID: mdl-31057189

ABSTRACT

During the course of a research program aimed at identifying novel antileishmanial compounds, a multi-gram synthesis of N-(trans-4-((4-methoxy-3-((R)-3-methylmorpholino)-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)cyclohexyl)-2-methylpropane-1-sulfonamide (( R )-1) was required. This letter describes optimisation of the reaction conditions and protecting group strategy for a key Buchwald-Hartwig coupling, delivering the required quantities of ( R )-1, as well as further compounds in the series.

15.
Article in English | MEDLINE | ID: mdl-29844044

ABSTRACT

The lack of information regarding the mechanisms of action (MoA) or specific molecular targets of phenotypically active compounds can prove a barrier to their development as chemotherapeutic agents. Here, we report the results of our orthogonal genetic, molecular, and biochemical studies to determine the MoA of a novel 7-substituted 8-hydroxy-1,6-naphthyridine (8-HNT) series that displays promising activity against Trypanosoma brucei and Leishmania donovani High-throughput loss-of-function genetic screens in T. brucei highlighted two probable zinc transporters associated with resistance to these compounds. These transporters localized to the parasite Golgi apparatus. Directed by these findings, the role of zinc and other divalent cations in the MoA of these compounds was investigated. 8-HNT compounds were found to directly deplete intracellular levels of Zn2+, while the addition of exogenous Zn2+ and Fe2+ reduced the potency of compounds from this series. Detailed biochemical analyses confirmed that 8-HNT compounds bind directly to a number of divalent cations, predominantly Zn2+, Fe2+, and Cu2+, forming 2:1 complexes with one of these cations. Collectively, our studies demonstrate transition metal depletion, due to chelation, as the MoA of the 8-HNT series of compounds. Strategies to improve the selectivity of 8-HNT compounds are discussed.


Subject(s)
Antiprotozoal Agents/pharmacology , Cation Transport Proteins/genetics , Chelating Agents/pharmacology , Naphthyridines/pharmacology , Protozoan Proteins/genetics , Zinc/metabolism , Antiprotozoal Agents/chemical synthesis , Cation Transport Proteins/metabolism , Cations, Divalent , Chelating Agents/chemical synthesis , Copper/metabolism , Gene Expression , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Iron/metabolism , Leishmania donovani/drug effects , Leishmania donovani/genetics , Leishmania donovani/growth & development , Leishmania donovani/metabolism , Mutation , Naphthyridines/chemical synthesis , Parasitic Sensitivity Tests , Protozoan Proteins/metabolism , Structure-Activity Relationship , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei brucei/metabolism
16.
Bioorg Med Chem Lett ; 28(18): 3025-3030, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30104093

ABSTRACT

Chagas' disease, caused by the protozoan parasite Trypanosoma cruzi, affects 8-10 million people across the Latin American population and is responsible for around 12,500 deaths per annum. The current frontline treatments, benznidazole and nifurtimox, are associated with side effects and lack efficacy in the chronic stage of the disease, leading to an urgent need for new treatments. A high throughput screening campaign against the physiologically relevant intracellular form of the parasite identified a series of 2,4-diamino-6-methylpyrimidines. Demonstrating the series did not work through the anti-target TcCYP51, and was generally cytocidal, confirmed its suitability for further development. This study reports the optimisation of selectivity and metabolic stability of the series and identification of a suitable lead for further optimisation.


Subject(s)
Chagas Disease/drug therapy , Pyrimidines/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Structure , Parasitic Sensitivity Tests , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , Trypanosoma cruzi/drug effects
17.
Nature ; 492(7428): 215-20, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23235874

ABSTRACT

The clinical efficacy and safety of a drug is determined by its activity profile across many proteins in the proteome. However, designing drugs with a specific multi-target profile is both complex and difficult. Therefore methods to design drugs rationally a priori against profiles of several proteins would have immense value in drug discovery. Here we describe a new approach for the automated design of ligands against profiles of multiple drug targets. The method is demonstrated by the evolution of an approved acetylcholinesterase inhibitor drug into brain-penetrable ligands with either specific polypharmacology or exquisite selectivity profiles for G-protein-coupled receptors. Overall, 800 ligand-target predictions of prospectively designed ligands were tested experimentally, of which 75% were confirmed to be correct. We also demonstrate target engagement in vivo. The approach can be a useful source of drug leads when multi-target profiles are required to achieve either selectivity over other drug targets or a desired polypharmacology.


Subject(s)
Drug Design , Ligands , Animals , Automation , Drug Delivery Systems , Female , Male , Mice , Mice, Inbred C57BL , Models, Theoretical , Pharmacological Phenomena , Reproducibility of Results
18.
Malar J ; 16(1): 446, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29115999

ABSTRACT

BACKGROUND: Protein kinases have been shown to be key drug targets, especially in the area of oncology. It is of interest to explore the possibilities of protein kinases as a potential target class in Plasmodium spp., the causative agents of malaria. However, protein kinase biology in malaria is still being investigated. Therefore, rather than assaying against individual protein kinases, a library of 4731 compounds with protein kinase inhibitor-like scaffolds was screened against the causative parasite, Plasmodium falciparum. This approach is more holistic and considers the whole kinome, making it possible to identify compounds that inhibit more than one P. falciparum protein kinase, or indeed other malaria targets. RESULTS: As a result of this screen, 9 active compound series were identified; further validation was carried out on 4 of these series, with 3 being progressed into hits to lead chemistry. The detailed evaluation of one of these series is described. DISCUSSION: This screening approach proved to be an effective way to identify series for further optimisation against malaria. Compound optimisation was carried out in the absence of knowledge of the molecular target. Some of the series had to be halted for various reasons. Mode of action studies to find the molecular target may be useful when problems prevent further chemical optimisation. CONCLUSIONS: Progressible series were identified through phenotypic screening of a relatively small focused kinase scaffold chemical library.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Protein Kinase Inhibitors/pharmacology , Drug Evaluation, Preclinical
19.
Nature ; 464(7289): 728-32, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20360736

ABSTRACT

African sleeping sickness or human African trypanosomiasis, caused by Trypanosoma brucei spp., is responsible for approximately 30,000 deaths each year. Available treatments for this disease are poor, with unacceptable efficacy and safety profiles, particularly in the late stage of the disease when the parasite has infected the central nervous system. Here we report the validation of a molecular target and the discovery of associated lead compounds with the potential to address this lack of suitable treatments. Inhibition of this target-T. brucei N-myristoyltransferase-leads to rapid killing of trypanosomes both in vitro and in vivo and cures trypanosomiasis in mice. These high-affinity inhibitors bind into the peptide substrate pocket of the enzyme and inhibit protein N-myristoylation in trypanosomes. The compounds identified have promising pharmaceutical properties and represent an opportunity to develop oral drugs to treat this devastating disease. Our studies validate T. brucei N-myristoyltransferase as a promising therapeutic target for human African trypanosomiasis.


Subject(s)
Acyltransferases/antagonists & inhibitors , Antiparasitic Agents/pharmacology , Antiparasitic Agents/therapeutic use , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology , Acyltransferases/metabolism , Aminopyridines/chemistry , Aminopyridines/metabolism , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Antiparasitic Agents/chemistry , Antiparasitic Agents/metabolism , Enzyme Assays , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Mice , Molecular Structure , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Rats , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Time Factors , Trypanosoma brucei brucei/growth & development
20.
Antimicrob Agents Chemother ; 58(10): 5747-57, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25022590

ABSTRACT

This paper reports an evaluation of a melamino nitroheterocycle, a potential lead for further development as an agent against human African trypanosomiasis (HAT). Studies on its efficacy, physicochemical and biopharmaceutical properties, and potential for toxicity are described. The compound previously had been shown to possess exceptional activity against Trypanosoma brucei in in vitro assays comparable to that of melarsoprol. Here, we demonstrate that the compound also was curative in the stringent acute mouse model T. brucei rhodesiense STIB 900 when given intraperitoneally at 40 mg/kg of body weight. Nevertheless, activity was only moderate when the oral route was used, and no cure was obtained when the compound was tested in a stage 2 rodent model of infection. Genotoxic profiling revealed that the compound induces DNA damage by a mechanism apparently independent from nitroreduction and involving the introduction of base pair substitutions (Ames test), possibly caused by oxidative damage of the DNA (comet test). No significant genotoxicity was observed at the chromosome level (micronucleus assay). The lack of suitable properties for oral and central nervous system uptake and the genotoxic liabilities prevent the progression of this melamine nitroheterocycle as a drug candidate for HAT. Further modification of the compound is required to improve the pharmacokinetic properties of the molecule and to separate the trypanocidal activity from the toxic potential.


Subject(s)
Trypanocidal Agents/therapeutic use , Trypanosoma brucei rhodesiense/drug effects , Trypanosoma brucei rhodesiense/pathogenicity , Trypanosomiasis, African/drug therapy , Animals , Disease Models, Animal , Humans , Male , Mice , Parasitic Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL