Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Elife ; 122023 Sep 22.
Article in English | MEDLINE | ID: mdl-37737220

ABSTRACT

Drug resistance remains a major obstacle to malaria control and eradication efforts, necessitating the development of novel therapeutic strategies to treat this disease. Drug combinations based on collateral sensitivity, wherein resistance to one drug causes increased sensitivity to the partner drug, have been proposed as an evolutionary strategy to suppress the emergence of resistance in pathogen populations. In this study, we explore collateral sensitivity between compounds targeting the Plasmodium dihydroorotate dehydrogenase (DHODH). We profiled the cross-resistance and collateral sensitivity phenotypes of several DHODH mutant lines to a diverse panel of DHODH inhibitors. We focus on one compound, TCMDC-125334, which was active against all mutant lines tested, including the DHODH C276Y line, which arose in selections with the clinical candidate DSM265. In six selections with TCMDC-125334, the most common mechanism of resistance to this compound was copy number variation of the dhodh locus, although we did identify one mutation, DHODH I263S, which conferred resistance to TCMDC-125334 but not DSM265. We found that selection of the DHODH C276Y mutant with TCMDC-125334 yielded additional genetic changes in the dhodh locus. These double mutant parasites exhibited decreased sensitivity to TCMDC-125334 and were highly resistant to DSM265. Finally, we tested whether collateral sensitivity could be exploited to suppress the emergence of resistance in the context of combination treatment by exposing wildtype parasites to both DSM265 and TCMDC-125334 simultaneously. This selected for parasites with a DHODH V532A mutation which were cross-resistant to both compounds and were as fit as the wildtype parent in vitro. The emergence of these cross-resistant, evolutionarily fit parasites highlights the mutational flexibility of the DHODH enzyme.


Malaria affects around 240 million people around the world every year. The microscopic parasite responsible for the disease are carried by certain mosquitoes and gets transmitted to humans through bites. These parasites are increasingly acquiring genetic mutations that make anti-malaria medication less effective, creating an urgent need for alternative treatment approaches. Several new malaria drugs being explored in preclinical research work by binding to an enzyme known as DHODH and preventing it from performing its usual role in the parasite. Previous work found that, in some cases, malaria parasites that evolved resistance to one type of DHODH inhibitor (by acquiring mutations in their DHODH enzyme) then became more vulnerable to another kind. It may be possible to leverage this 'collateral sensitivity' by designing treatments which combine two DHODH inhibitors and therefore make it harder for the parasites to evolve resistance. To investigate this possibility, Mandt et al. first tested several DHODH inhibitors to find the one that was most potent against drug-resistant parasites. In subsequent experiments, they combined TCMDC-125334, the best candidate that emerged from these tests, with a DHODH inhibitor that works well against vulnerable parasites. However, the parasites still rapidly evolved resistance. Further work identified a new DHODH mutation that allowed the parasites to evade both drugs simultaneously. Together, these findings suggest that the DHODH enzyme may not be the best target for new malaria drugs because many it can acquire many possible mutations that confer resistance. Such results may inform other studies that aim to harness collateral sensitivity to fight against a range of harmful agents.


Subject(s)
Antimalarials , Malaria, Falciparum , Oxidoreductases Acting on CH-CH Group Donors , Parasites , Animals , Humans , Dihydroorotate Dehydrogenase , Malaria, Falciparum/parasitology , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Plasmodium falciparum , Antimalarials/pharmacology , Antimalarials/therapeutic use , DNA Copy Number Variations , Drug Collateral Sensitivity , Parasites/metabolism
2.
PLoS One ; 18(3): e0279144, 2023.
Article in English | MEDLINE | ID: mdl-36928885

ABSTRACT

Early Plasmodium falciparum and P. vivax infection requires parasite replication within host hepatocytes, referred to as liver stage (LS). However, limited understanding of infection dynamics in human LS exists due to species-specificity challenges. Reported here is a reproducible, easy-to-manipulate, and moderate-cost in vivo model to study human Plasmodium LS in mice; the ectopic huLiver model. Ectopic huLiver tumors were generated through subcutaneous injection of the HC-04 cell line and shown to be infectible by both freshly dissected sporozoites and through the bite of infected mosquitoes. Evidence for complete LS development was supported by the transition to blood-stage infection in mice engrafted with human erythrocytes. Additionally, this model was successfully evaluated for its utility in testing antimalarial therapeutics, as supported by primaquine acting as a causal prophylactic against P. falciparum. Presented here is a new platform for the study of human Plasmodium infection with the potential to aid in drug discovery.


Subject(s)
Communicable Diseases , Liver Diseases , Malaria, Falciparum , Malaria, Vivax , Malaria , Plasmodium , Mice , Animals , Humans , Liver/parasitology , Malaria/drug therapy , Malaria, Falciparum/parasitology , Hepatocytes/parasitology , Plasmodium falciparum , Sporozoites
3.
Sci Transl Med ; 14(667): eabo7219, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36260689

ABSTRACT

Compounds acting on multiple targets are critical to combating antimalarial drug resistance. Here, we report that the human "mammalian target of rapamycin" (mTOR) inhibitor sapanisertib has potent prophylactic liver stage activity, in vitro and in vivo asexual blood stage (ABS) activity, and transmission-blocking activity against the protozoan parasite Plasmodium spp. Chemoproteomics studies revealed multiple potential Plasmodium kinase targets, and potent inhibition of Plasmodium phosphatidylinositol 4-kinase type III beta (PI4Kß) and cyclic guanosine monophosphate-dependent protein kinase (PKG) was confirmed in vitro. Conditional knockdown of PI4Kß in ABS cultures modulated parasite sensitivity to sapanisertib, and laboratory-generated P. falciparum sapanisertib resistance was mediated by mutations in PI4Kß. Parasite metabolomic perturbation profiles associated with sapanisertib and other known PI4Kß and/or PKG inhibitors revealed similarities and differences between chemotypes, potentially caused by sapanisertib targeting multiple parasite kinases. The multistage activity of sapanisertib and its in vivo antimalarial efficacy, coupled with potent inhibition of at least two promising drug targets, provides an opportunity to reposition this pyrazolopyrimidine for malaria.


Subject(s)
Antimalarials , Plasmodium , Animals , Humans , Antimalarials/pharmacology , Antimalarials/therapeutic use , Plasmodium falciparum , MTOR Inhibitors , 1-Phosphatidylinositol 4-Kinase , Guanosine Monophosphate , Life Cycle Stages , TOR Serine-Threonine Kinases , Sirolimus , Mammals
4.
Angew Chem Int Ed Engl ; 60(17): 9279-9283, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33433953

ABSTRACT

Plasmodium falciparum proteasome (Pf20S) inhibitors are active against Plasmodium at multiple stages-erythrocytic, gametocyte, liver, and gamete activation stages-indicating that selective Pf20S inhibitors possess the potential to be therapeutic, prophylactic, and transmission-blocking antimalarials. Starting from a reported compound, we developed a noncovalent, macrocyclic peptide inhibitor of the malarial proteasome with high species selectivity and improved pharmacokinetic properties. The compound demonstrates specific, time-dependent inhibition of the ß5 subunit of the Pf20S, kills artemisinin-sensitive and artemisinin-resistant P. falciparum isolates in vitro and reduces parasitemia in humanized, P. falciparum-infected mice.


Subject(s)
Antimalarials/pharmacology , Drug Development , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Malaria, Falciparum/metabolism , Mice , Models, Molecular , Molecular Conformation , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry
5.
ACS Infect Dis ; 6(5): 1058-1075, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32272012

ABSTRACT

Little is known about the role of the three Jumonji C (JmjC) enzymes in Plasmodium falciparum (Pf). Here, we show that JIB-04 and other established inhibitors of mammalian JmjC histone demethylases kill asexual blood stage parasites and are even more potent at blocking gametocyte development and gamete formation. In late stage parasites, JIB-04 increased levels of trimethylated lysine residues on histones, suggesting the inhibition of P. falciparum Jumonji demethylase activity. These epigenetic defects coincide with deregulation of invasion, cell motor, and sexual development gene programs, including gene targets coregulated by the PfAP2-I transcription factor and chromatin-binding factor, PfBDP1. Mechanistically, we demonstrate that PfJmj3 converts 2-oxoglutarate to succinate in an iron-dependent manner consistent with mammalian Jumonji enzymes, and this catalytic activity is inhibited by JIB-04 and other Jumonji inhibitors. Our pharmacological studies of Jumonji activity in the malaria parasite provide evidence that inhibition of these enzymatic activities is detrimental to the parasite.


Subject(s)
Aminopyridines/pharmacology , Hydrazones/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Plasmodium falciparum/drug effects , Animals , Enzyme Inhibitors/pharmacology , Histones , Life Cycle Stages , Lysine
6.
J Med Chem ; 63(9): 4929-4956, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32248693

ABSTRACT

Malaria puts at risk nearly half the world's population and causes high mortality in sub-Saharan Africa, while drug resistance threatens current therapies. The pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase (DHODH) is a validated target for malaria treatment based on our finding that triazolopyrimidine DSM265 (1) showed efficacy in clinical studies. Herein, we describe optimization of a pyrrole-based series identified using a target-based DHODH screen. Compounds with nanomolar potency versus Plasmodium DHODH and Plasmodium parasites were identified with good pharmacological properties. X-ray studies showed that the pyrroles bind an alternative enzyme conformation from 1 leading to improved species selectivity versus mammalian enzymes and equivalent activity on Plasmodium falciparum and Plasmodium vivax DHODH. The best lead DSM502 (37) showed in vivo efficacy at similar levels of blood exposure to 1, although metabolic stability was reduced. Overall, the pyrrole-based DHODH inhibitors provide an attractive alternative scaffold for the development of new antimalarial compounds.


Subject(s)
Antimalarials/therapeutic use , Enzyme Inhibitors/therapeutic use , Malaria, Falciparum/drug therapy , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Pyrroles/therapeutic use , Animals , Antimalarials/chemical synthesis , Antimalarials/metabolism , Antimalarials/pharmacokinetics , Cell Line, Tumor , Crystallography, X-Ray , Dihydroorotate Dehydrogenase , Dogs , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Male , Mice, SCID , Microsomes, Liver/metabolism , Molecular Structure , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Parasitic Sensitivity Tests , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium vivax/drug effects , Plasmodium vivax/enzymology , Protein Binding , Pyrroles/chemical synthesis , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Rats , Structure-Activity Relationship
7.
ACS Infect Dis ; 6(1): 3-13, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31808676

ABSTRACT

In May 2019, the Wellcome Centre for Anti-Infectives Research (WCAIR) at the University of Dundee, UK, held an international conference with the aim of discussing some key questions around discovering new medicines for infectious diseases and a particular focus on diseases affecting Low and Middle Income Countries. There is an urgent need for new drugs to treat most infectious diseases. We were keen to see if there were lessons that we could learn across different disease areas and between the preclinical and clinical phases with the aim of exploring how we can improve and speed up the drug discovery, translational, and clinical development processes. We started with an introductory session on the current situation and then worked backward from clinical development to combination therapy, pharmacokinetic/pharmacodynamic (PK/PD) studies, drug discovery pathways, and new starting points and targets. This Viewpoint aims to capture some of the learnings.


Subject(s)
Communicable Disease Control , Communicable Diseases/drug therapy , Congresses as Topic , Combined Modality Therapy , Communicable Diseases/epidemiology , Drug Discovery , Drug Evaluation, Preclinical , HIV Infections/drug therapy , Humans , Poverty , United Kingdom
8.
Sci Transl Med ; 11(521)2019 12 04.
Article in English | MEDLINE | ID: mdl-31801884

ABSTRACT

Resistance has developed in Plasmodium malaria parasites to every antimalarial drug in clinical use, prompting the need to characterize the pathways mediating resistance. Here, we report a framework for assessing development of resistance of Plasmodium falciparum to new antimalarial therapeutics. We investigated development of resistance by P. falciparum to the dihydroorotate dehydrogenase (DHODH) inhibitors DSM265 and DSM267 in tissue culture and in a mouse model of P. falciparum infection. We found that resistance to these drugs arose rapidly both in vitro and in vivo. We identified 13 point mutations mediating resistance in the parasite DHODH in vitro that overlapped with the DHODH mutations that arose in the mouse infection model. Mutations in DHODH conferred increased resistance (ranging from 2- to ~400-fold) to DHODH inhibitors in P. falciparum in vitro and in vivo. We further demonstrated that the drug-resistant parasites carrying the C276Y mutation had mitochondrial energetics comparable to the wild-type parasite and also retained their fitness in competitive growth experiments. Our data suggest that in vitro selection of drug-resistant P. falciparum can predict development of resistance in a mouse model of malaria infection.


Subject(s)
Enzyme Inhibitors/therapeutic use , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Parasites/enzymology , Animals , Dihydroorotate Dehydrogenase , Disease Models, Animal , Drug Resistance/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Mice, SCID , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Parasites/drug effects , Phenotype , Plasmodium falciparum , Point Mutation/genetics , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/therapeutic use
9.
Science ; 365(6456)2019 08 30.
Article in English | MEDLINE | ID: mdl-31467193

ABSTRACT

The requirement for next-generation antimalarials to be both curative and transmission-blocking necessitates the identification of previously undiscovered druggable molecular pathways. We identified a selective inhibitor of the Plasmodium falciparum protein kinase PfCLK3, which we used in combination with chemogenetics to validate PfCLK3 as a drug target acting at multiple parasite life stages. Consistent with a role for PfCLK3 in RNA splicing, inhibition resulted in the down-regulation of more than 400 essential parasite genes. Inhibition of PfCLK3 mediated rapid killing of asexual liver- and blood-stage P. falciparum and blockade of gametocyte development, thereby preventing transmission, and also showed parasiticidal activity against P. berghei and P. knowlesi Hence, our data establish PfCLK3 as a target for drugs, with the potential to offer a cure-to be prophylactic and transmission blocking in malaria.


Subject(s)
Antimalarials/pharmacology , Molecular Targeted Therapy , Plasmodium falciparum/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Animals , Antimalarials/chemistry , Antimalarials/isolation & purification , Antimalarials/therapeutic use , Gametogenesis/drug effects , High-Throughput Screening Assays , Mice , Mice, Inbred BALB C , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Protozoan Proteins/genetics , RNA Splicing/genetics , Small Molecule Libraries/pharmacology
10.
ACS Omega ; 3(8): 9227-9240, 2018 Aug 31.
Article in English | MEDLINE | ID: mdl-30197997

ABSTRACT

Malaria kills nearly 0.5 million people yearly and impacts the lives of those living in over 90 countries where it is endemic. The current treatment programs are threatened by increasing drug resistance. Dihydroorotate dehydrogenase (DHODH) is now clinically validated as a target for antimalarial drug discovery as a triazolopyrimidine class inhibitor (DSM265) is currently undergoing clinical development. We discovered a related isoxazolopyrimidine series in a phenotypic screen, later determining that it targeted DHODH. To determine if the isoxazolopyrimidines could yield a drug candidate, we initiated hit-to-lead medicinal chemistry. Several potent analogues were identified, including a compound that showed in vivo antimalarial activity. The isoxazolopyrimidines were more rapidly metabolized than their triazolopyrimidine counterparts, and the pharmacokinetic data were not consistent with the goal of a single-dose treatment for malaria.

11.
J Med Chem ; 61(9): 4213-4227, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29665687

ABSTRACT

Optimization of a chemical series originating from whole-cell phenotypic screening against the human malaria parasite, Plasmodium falciparum, led to the identification of two promising 2,6-disubstituted imidazopyridine compounds, 43 and 74. These compounds exhibited potent activity against asexual blood stage parasites that, together with their in vitro absorption, distribution, metabolism, and excretion (ADME) properties, translated to in vivo efficacy with clearance of parasites in the PfSCID mouse model for malaria within 48 h of treatment.


Subject(s)
Drug Discovery , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Malaria/drug therapy , Plasmodium falciparum/physiology , Pyridines/chemistry , Pyridines/pharmacokinetics , Animals , Disease Models, Animal , Drug Stability , ERG1 Potassium Channel/metabolism , Humans , Imidazoles/metabolism , Imidazoles/therapeutic use , Malaria/genetics , Malaria/metabolism , Mice , Pyridines/metabolism , Pyridines/therapeutic use , Solubility , Structure-Activity Relationship , Tissue Distribution , Water/chemistry
12.
ACS Infect Dis ; 4(4): 508-515, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29336544

ABSTRACT

Drug resistance has been reported for every antimalarial in use highlighting the need for new strategies to protect the efficacy of therapeutics in development. We have previously shown that resistance can be suppressed with a population biology trap: by identifying situations where resistance to one compound confers hypersensitivity to another (collateral sensitivity), we can design combination therapies that not only kill the parasite but also guide its evolution away from resistance. We applied this concept to the Plasmodium falciparum dihydroorotate dehydrogenase ( PfDHODH) enzyme, a well validated antimalarial target with inhibitors in the development pipeline. Here, we report a high-throughput screen to identify compounds specifically active against PfDHODH resistant mutants. We additionally perform extensive cross-resistance profiling allowing us to identify compound pairs demonstrating the potential for mutually incompatible resistance. These combinations represent promising starting points for exploiting collateral sensitivity to extend the useful lifespan of new antimalarial therapeutics.


Subject(s)
Antimalarials/isolation & purification , Antimalarials/pharmacology , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Dihydroorotate Dehydrogenase , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods
13.
J Med Chem ; 60(24): 10118-10134, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29148755

ABSTRACT

A BioFocus DPI SoftFocus library of ∼35 000 compounds was screened against Mycobacterium tuberculosis (Mtb) in order to identify novel hits with antitubercular activity. The hits were evaluated in biology triage assays to exclude compounds suggested to function via frequently encountered promiscuous mechanisms of action including inhibition of the QcrB subunit of the cytochrome bc1 complex, disruption of cell-wall homeostasis, and DNA damage. Among the hits that passed this screening cascade, a 6-dialkylaminopyrimidine carboxamide series was prioritized for hit to lead optimization. Compounds from this series were active against clinical Mtb strains, while no cross-resistance to conventional antituberculosis drugs was observed. This suggested a novel mechanism of action, which was confirmed by chemoproteomic analysis leading to the identification of BCG_3193 and BCG_3827 as putative targets of the series with unknown function. Initial structure-activity relationship studies have resulted in compounds with moderate to potent antitubercular activity and improved physicochemical properties.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Structure-Activity Relationship , Administration, Oral , Animals , Antitubercular Agents/chemical synthesis , Blood Proteins/metabolism , Drug Stability , High-Throughput Screening Assays , Humans , Male , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Mycobacterium tuberculosis/isolation & purification , Proteomics/methods , Pyrimidines/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
15.
J Med Chem ; 60(16): 6880-6896, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28806082

ABSTRACT

Since the appearance of resistance to the current front-line antimalarial treatments, ACTs (artemisinin combination therapies), the discovery of novel chemical entities to treat the disease is recognized as a major global health priority. From the GSK antimalarial set, we identified an aminoxadiazole with an antiparasitic profile comparable with artemisinin (1), with no cross-resistance in a resistant strains panel and a potential new mode of action. A medicinal chemistry program allowed delivery of compounds such as 19 with high solubility in aqueous media, an acceptable toxicological profile, and oral efficacy. Further evaluation of the lead compounds showed that in vivo genotoxic degradants might be generated. The compounds generated during this medicinal chemistry program and others from the GSK collection were used to build a pharmacophore model which could be used in the virtual screening of compound collections and potentially identify new chemotypes that could deliver the same antiparasitic profile.


Subject(s)
2,2'-Dipyridyl/analogs & derivatives , Antimalarials/pharmacology , Oxadiazoles/pharmacology , 2,2'-Dipyridyl/administration & dosage , 2,2'-Dipyridyl/chemical synthesis , 2,2'-Dipyridyl/pharmacology , 2,2'-Dipyridyl/toxicity , Animals , Antimalarials/administration & dosage , Antimalarials/chemical synthesis , Antimalarials/toxicity , Atovaquone/pharmacology , Chloroquine/pharmacology , Drug Design , Female , Humans , Hydrazines/metabolism , Mice , Mutagenicity Tests , Mutagens/metabolism , Oxadiazoles/administration & dosage , Oxadiazoles/chemical synthesis , Oxadiazoles/toxicity , Parasitemia/drug therapy , Plasmodium falciparum/drug effects , Pyrimethamine/pharmacology , Structure-Activity Relationship
16.
Sci Transl Med ; 9(387)2017 04 26.
Article in English | MEDLINE | ID: mdl-28446690

ABSTRACT

As part of the global effort toward malaria eradication, phenotypic whole-cell screening revealed the 2-aminopyridine class of small molecules as a good starting point to develop new antimalarial drugs. Stemming from this series, we found that the derivative, MMV390048, lacked cross-resistance with current drugs used to treat malaria. This compound was efficacious against all Plasmodium life cycle stages, apart from late hypnozoites in the liver. Efficacy was shown in the humanized Plasmodium falciparum mouse model, and modest reductions in mouse-to-mouse transmission were achieved in the Plasmodium berghei mouse model. Experiments in monkeys revealed the ability of MMV390048 to be used for full chemoprotection. Although MMV390048 was not able to eliminate liver hypnozoites, it delayed relapse in a Plasmodium cynomolgi monkey model. Both genomic and chemoproteomic studies identified a kinase of the Plasmodium parasite, phosphatidylinositol 4-kinase, as the molecular target of MMV390048. The ability of MMV390048 to block all life cycle stages of the malaria parasite suggests that this compound should be further developed and may contribute to malaria control and eradication as part of a single-dose combination treatment.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Aminopyridines/therapeutic use , Antimalarials/therapeutic use , Sulfones/therapeutic use , Aminopyridines/pharmacology , Animals , Antimalarials/pharmacology , Female , Malaria/drug therapy , Malaria/enzymology , Male , Mice , Mice, SCID , Parasitic Sensitivity Tests , Plasmodium/drug effects , Plasmodium/pathogenicity , Sulfones/pharmacology
17.
Nat Commun ; 8: 14574, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262680

ABSTRACT

Benzoxaboroles are effective against bacterial, fungal and protozoan pathogens. We report potent activity of the benzoxaborole AN3661 against Plasmodium falciparum laboratory-adapted strains (mean IC50 32 nM), Ugandan field isolates (mean ex vivo IC50 64 nM), and murine P. berghei and P. falciparum infections (day 4 ED90 0.34 and 0.57 mg kg-1, respectively). Multiple P. falciparum lines selected in vitro for resistance to AN3661 harboured point mutations in pfcpsf3, which encodes a homologue of mammalian cleavage and polyadenylation specificity factor subunit 3 (CPSF-73 or CPSF3). CRISPR-Cas9-mediated introduction of pfcpsf3 mutations into parental lines recapitulated AN3661 resistance. PfCPSF3 homology models placed these mutations in the active site, where AN3661 is predicted to bind. Transcripts for three trophozoite-expressed genes were lost in AN3661-treated trophozoites, which was not observed in parasites selected or engineered for AN3661 resistance. Our results identify the pre-mRNA processing factor PfCPSF3 as a promising antimalarial drug target.


Subject(s)
Antimalarials/pharmacology , Boron Compounds/pharmacology , Cleavage And Polyadenylation Specificity Factor/chemistry , Plasmodium falciparum/drug effects , Protozoan Proteins/chemistry , RNA, Messenger/genetics , Amino Acid Sequence , Animals , Antimalarials/chemical synthesis , Boron Compounds/chemical synthesis , CRISPR-Cas Systems , Catalytic Domain , Cleavage And Polyadenylation Specificity Factor/antagonists & inhibitors , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Drug Resistance/genetics , Erythrocytes/drug effects , Erythrocytes/parasitology , Gene Editing/methods , Humans , Malaria/drug therapy , Malaria/parasitology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Mice , Molecular Docking Simulation , Mutation , Plasmodium berghei/drug effects , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA, Messenger/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Trophozoites/drug effects , Trophozoites/genetics , Trophozoites/growth & development , Trophozoites/metabolism
18.
ACS Infect Dis ; 3(1): 18-33, 2017 01 13.
Article in English | MEDLINE | ID: mdl-27704782

ABSTRACT

A potent, noncytotoxic indazole sulfonamide was identified by high-throughput screening of >100,000 synthetic compounds for activity against Mycobacterium tuberculosis (Mtb). This noncytotoxic compound did not directly inhibit cell wall biogenesis but triggered a slow lysis of Mtb cells as measured by release of intracellular green fluorescent protein (GFP). Isolation of resistant mutants followed by whole-genome sequencing showed an unusual gene amplification of a 40 gene region spanning from Rv3371 to Rv3411c and in one case a potential promoter mutation upstream of guaB2 (Rv3411c) encoding inosine monophosphate dehydrogenase (IMPDH). Subsequent biochemical validation confirmed direct inhibition of IMPDH by an uncompetitive mode of inhibition, and growth inhibition could be rescued by supplementation with guanine, a bypass mechanism for the IMPDH pathway. Beads containing immobilized indazole sulfonamides specifically interacted with IMPDH in cell lysates. X-ray crystallography of the IMPDH-IMP-inhibitor complex revealed that the primary interactions of these compounds with IMPDH were direct pi-pi interactions with the IMP substrate. Advanced lead compounds in this series with acceptable pharmacokinetic properties failed to show efficacy in acute or chronic murine models of tuberculosis (TB). Time-kill experiments in vitro suggest that sustained exposure to drug concentrations above the minimum inhibitory concentration (MIC) for 24 h were required for a cidal effect, levels that have been difficult to achieve in vivo. Direct measurement of guanine levels in resected lung tissue from tuberculosis-infected animals and patients revealed 0.5-2 mM concentrations in caseum and normal lung tissue. The high lesional levels of guanine and the slow lytic, growth-rate-dependent effect of IMPDH inhibition pose challenges to developing drugs against this target for use in treating TB.


Subject(s)
Antitubercular Agents/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Sulfonamides/pharmacology , Animals , Drug Design , Drug Discovery , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Mutation , Protein Conformation , Rabbits , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Tuberculosis/drug therapy
19.
ACS Cent Sci ; 2(10): 687-701, 2016 Oct 26.
Article in English | MEDLINE | ID: mdl-27800551

ABSTRACT

The development of new antimalarial compounds remains a pivotal part of the strategy for malaria elimination. Recent large-scale phenotypic screens have provided a wealth of potential starting points for hit-to-lead campaigns. One such public set is explored, employing an open source research mechanism in which all data and ideas were shared in real time, anyone was able to participate, and patents were not sought. One chemical subseries was found to exhibit oral activity but contained a labile ester that could not be replaced without loss of activity, and the original hit exhibited remarkable sensitivity to minor structural change. A second subseries displayed high potency, including activity within gametocyte and liver stage assays, but at the cost of low solubility. As an open source research project, unexplored avenues are clearly identified and may be explored further by the community; new findings may be cumulatively added to the present work.

20.
ACS Infect Dis ; 2(12): 945-957, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27641613

ABSTRACT

The emergence of drug-resistant malaria parasites continues to hamper efforts to control this lethal disease. Dihydroorotate dehydrogenase has recently been validated as a new target for the treatment of malaria, and a selective inhibitor (DSM265) of the Plasmodium enzyme is currently in clinical development. With the goal of identifying a backup compound to DSM265, we explored replacement of the SF5-aniline moiety of DSM265 with a series of CF3-pyridinyls while maintaining the core triazolopyrimidine scaffold. This effort led to the identification of DSM421, which has improved solubility, lower intrinsic clearance, and increased plasma exposure after oral dosing compared to DSM265, while maintaining a long predicted human half-life. Its improved physical and chemical properties will allow it to be formulated more readily than DSM265. DSM421 showed excellent efficacy in the SCID mouse model of P. falciparum malaria that supports the prediction of a low human dose (<200 mg). Importantly DSM421 showed equal activity against both P. falciparum and P. vivax field isolates, while DSM265 was more active on P. falciparum. DSM421 has the potential to be developed as a single-dose cure or once-weekly chemopreventative for both P. falciparum and P. vivax malaria, leading to its advancement as a preclinical development candidate.


Subject(s)
Enzyme Inhibitors/administration & dosage , Malaria, Falciparum/prevention & control , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Antimalarials/chemistry , Antimalarials/pharmacokinetics , Dihydroorotate Dehydrogenase , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Mice , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Pyrimidines/chemistry , Rats , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...