Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 2158, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35444200

ABSTRACT

Drug resistance and a dire lack of transmission-blocking antimalarials hamper malaria elimination. Here, we present the pantothenamide MMV693183 as a first-in-class acetyl-CoA synthetase (AcAS) inhibitor to enter preclinical development. Our studies demonstrate attractive drug-like properties and in vivo efficacy in a humanized mouse model of Plasmodium falciparum infection. The compound shows single digit nanomolar in vitro activity against P. falciparum and P. vivax clinical isolates, and potently blocks P. falciparum transmission to Anopheles mosquitoes. Genetic and biochemical studies identify AcAS as the target of the MMV693183-derived antimetabolite, CoA-MMV693183. Pharmacokinetic-pharmacodynamic modelling predict that a single 30 mg oral dose is sufficient to cure a malaria infection in humans. Toxicology studies in rats indicate a > 30-fold safety margin in relation to the predicted human efficacious exposure. In conclusion, MMV693183 represents a promising candidate for further (pre)clinical development with a novel mode of action for treatment of malaria and blocking transmission.


Subject(s)
Antimalarials , Folic Acid Antagonists , Malaria, Falciparum , Malaria, Vivax , Malaria , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Malaria, Vivax/drug therapy , Mice , Pantothenic Acid/analogs & derivatives , Plasmodium falciparum/genetics , Rats
2.
Article in English | MEDLINE | ID: mdl-34501726

ABSTRACT

The early stages of the COVID-19 pandemic presented the characteristics of a traumatic event that could trigger post-traumatic stress disorder. Emergency Medical Services workers are already a high-risk group due to their professional development. The research project aimed to analyse the impact of the COVID-19 pandemic on EMS professionals in terms of their mental health. For this purpose, we present a descriptive crosssectional study with survey methodology. A total of 317 EMS workers (doctors, nurses, and emergency medical technicians) were recruited voluntarily. Psychological distress, post-traumatic stress disorder, and insomnia were assessed. The instruments were the General Health Questionnaire-12 (GHQ-12), the Davidson Trauma Scale (DTS-8), and the Athens Insomnia Scale (AIS-8). We found that 36% of respondents had psychological distress, 30.9% potentially had PTSD, and 60.9% experienced insomnia. Years of work experience were found to be positively correlated, albeit with low effect, with the PTSD score (r = 0.133). Finally, it can be stated that the COVID-19 pandemic has been a traumatic event for EMS workers. The number of professionals presenting psychological distress, possible PTSD, or insomnia increased dramatically during the early phases of the pandemic. This study highlights the need for mental health disorder prevention programmes for EMS workers in the face of a pandemic.


Subject(s)
COVID-19 , Stress Disorders, Post-Traumatic , Humans , Pandemics , SARS-CoV-2 , Spain/epidemiology , Stress Disorders, Post-Traumatic/epidemiology
3.
Bioorg Chem ; 115: 105244, 2021 10.
Article in English | MEDLINE | ID: mdl-34452759

ABSTRACT

Malaria is a devastating parasitic disease caused by parasites from the genus Plasmodium. Therapeutic resistance has been reported against all clinically available antimalarials, threatening our ability to control the disease and therefore there is an ongoing need for the development of novel antimalarials. Towards this goal, we identified the 2-(N-phenyl carboxamide) triazolopyrimidine class from a high throughput screen of the Janssen Jumpstarter library against the asexual stages of the P. falciparum parasite. Here we describe the structure activity relationship of the identified class and the optimisation of asexual stage activity while maintaining selectivity against the human HepG2 cell line. The most potent analogues from this study were shown to exhibit equipotent activity against P. falciparum multidrug resistant strains and P. knowlesi asexual parasites. Asexual stage phenotyping studies determined the triazolopyrimidine class arrests parasites at the trophozoite stage, but it is likely these parasites are still metabolically active until the second asexual cycle, and thus have a moderate to slow onset of action. Non-NADPH dependent degradation of the central carboxamide and low aqueous solubility was observed in in vitro ADME profiling. A significant challenge remains to correct these liabilities for further advancement of the 2-(N-phenyl carboxamide) triazolopyrimidine scaffold as a potential moderate to slow acting partner in a curative or prophylactic antimalarial treatment.


Subject(s)
Antimalarials/pharmacology , Erythrocytes/drug effects , Plasmodium falciparum/drug effects , Plasmodium knowlesi/drug effects , Purines/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Erythrocytes/parasitology , Humans , Molecular Structure , Parasitic Sensitivity Tests , Purines/chemical synthesis , Purines/chemistry , Structure-Activity Relationship
4.
J Med Chem ; 64(17): 12582-12602, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34437804

ABSTRACT

A phenotypic high-throughput screen allowed discovery of quinazolinone-2-carboxamide derivatives as a novel antimalarial scaffold. Structure-activity relationship studies led to identification of a potent inhibitor 19f, 95-fold more potent than the original hit compound, active against laboratory-resistant strains of malaria. Profiling of 19f suggested a fast in vitro killing profile. In vivo activity in a murine model of human malaria in a dose-dependent manner constitutes a concomitant benefit.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Quinazolinones/pharmacology , Administration, Oral , Animals , Humans , Mice , Molecular Structure , Plasmodium falciparum/drug effects , Quinazolinones/chemistry , Structure-Activity Relationship
5.
Sci Transl Med ; 13(603)2021 07 21.
Article in English | MEDLINE | ID: mdl-34290058

ABSTRACT

The emergence and spread of Plasmodium falciparum resistance to first-line antimalarials creates an imperative to identify and develop potent preclinical candidates with distinct modes of action. Here, we report the identification of MMV688533, an acylguanidine that was developed following a whole-cell screen with compounds known to hit high-value targets in human cells. MMV688533 displays fast parasite clearance in vitro and is not cross-resistant with known antimalarials. In a P. falciparum NSG mouse model, MMV688533 displays a long-lasting pharmacokinetic profile and excellent safety. Selection studies reveal a low propensity for resistance, with modest loss of potency mediated by point mutations in PfACG1 and PfEHD. These proteins are implicated in intracellular trafficking, lipid utilization, and endocytosis, suggesting interference with these pathways as a potential mode of action. This preclinical candidate may offer the potential for a single low-dose cure for malaria.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria , Parasites , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Endocytosis , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Plasmodium falciparum
6.
ACS Infect Dis ; 7(6): 1680-1689, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33929818

ABSTRACT

Prolyl-tRNA synthetase (PRS) is a clinically validated antimalarial target. Screening of a set of PRS ATP-site binders, initially designed for human indications, led to identification of 1-(pyridin-4-yl)pyrrolidin-2-one derivatives representing a novel antimalarial scaffold. Evidence designates cytoplasmic PRS as the drug target. The frontrunner 1 and its active enantiomer 1-S exhibited low-double-digit nanomolar activity against resistant Plasmodium falciparum (Pf) laboratory strains and development of liver schizonts. No cross-resistance with strains resistant to other known antimalarials was noted. In addition, a similar level of growth inhibition was observed against clinical field isolates of Pf and P. vivax. The slow killing profile and the relative high propensity to develop resistance in vitro (minimum inoculum resistance of 8 × 105 parasites at a selection pressure of 3 × IC50) constitute unfavorable features for treatment of malaria. However, potent blood stage and antischizontal activity are compelling for causal prophylaxis which does not require fast onset of action. Achieving sufficient on-target selectivity appears to be particularly challenging and should be the primary focus during the next steps of optimization of this chemical series. Encouraging preliminary off-target profile and oral efficacy in a humanized murine model of Pf malaria allowed us to conclude that 1-(pyridin-4-yl)pyrrolidin-2-one derivatives represent a promising starting point for the identification of novel antimalarial prophylactic agents that selectively target Plasmodium PRS.


Subject(s)
Amino Acyl-tRNA Synthetases , Antimalarials , Malaria, Falciparum , Malaria , Animals , Antimalarials/pharmacology , Humans , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Mice , Plasmodium falciparum
7.
ChemMedChem ; 14(14): 1329-1335, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31188540

ABSTRACT

Herein we describe the optimization of a phenotypic hit against Plasmodium falciparum based on an aminoacetamide scaffold. This led to N-(3-chloro-4-fluorophenyl)-2-methyl-2-{[4-methyl-3-(morpholinosulfonyl)phenyl]amino}propanamide (compound 28) with low-nanomolar activity against the intraerythrocytic stages of the malaria parasite, and which was found to be inactive in a mammalian cell counter-screen up to 25 µm. Inhibition of gametes in the dual gamete activation assay suggests that this family of compounds may also have transmission blocking capabilities. Whilst we were unable to optimize the aqueous solubility and microsomal stability to a point at which the aminoacetamides would be suitable for in vivo pharmacokinetic and efficacy studies, compound 28 displayed excellent antimalarial potency and selectivity; it could therefore serve as a suitable chemical tool for drug target identification.


Subject(s)
Acetamides/pharmacology , Antimalarials/pharmacology , Acetamides/chemical synthesis , Acetamides/pharmacokinetics , Animals , Antimalarials/chemical synthesis , Antimalarials/pharmacokinetics , Humans , Mice , Microsomes, Liver/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium berghei/drug effects , Plasmodium cynomolgi/drug effects , Plasmodium falciparum/drug effects , Structure-Activity Relationship
8.
Proc Natl Acad Sci U S A ; 116(14): 7015-7020, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30894487

ABSTRACT

Malaria and cryptosporidiosis, caused by apicomplexan parasites, remain major drivers of global child mortality. New drugs for the treatment of malaria and cryptosporidiosis, in particular, are of high priority; however, there are few chemically validated targets. The natural product cladosporin is active against blood- and liver-stage Plasmodium falciparum and Cryptosporidium parvum in cell-culture studies. Target deconvolution in P. falciparum has shown that cladosporin inhibits lysyl-tRNA synthetase (PfKRS1). Here, we report the identification of a series of selective inhibitors of apicomplexan KRSs. Following a biochemical screen, a small-molecule hit was identified and then optimized by using a structure-based approach, supported by structures of both PfKRS1 and C. parvum KRS (CpKRS). In vivo proof of concept was established in an SCID mouse model of malaria, after oral administration (ED90 = 1.5 mg/kg, once a day for 4 d). Furthermore, we successfully identified an opportunity for pathogen hopping based on the structural homology between PfKRS1 and CpKRS. This series of compounds inhibit CpKRS and C. parvum and Cryptosporidium hominis in culture, and our lead compound shows oral efficacy in two cryptosporidiosis mouse models. X-ray crystallography and molecular dynamics simulations have provided a model to rationalize the selectivity of our compounds for PfKRS1 and CpKRS vs. (human) HsKRS. Our work validates apicomplexan KRSs as promising targets for the development of drugs for malaria and cryptosporidiosis.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum/enzymology , Enzyme Inhibitors/pharmacology , Lysine-tRNA Ligase/antagonists & inhibitors , Malaria, Falciparum , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Animals , Cryptosporidiosis/drug therapy , Cryptosporidiosis/enzymology , Disease Models, Animal , Enzyme Inhibitors/chemistry , Humans , Lysine-tRNA Ligase/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/enzymology , Mice, SCID , Protozoan Proteins/metabolism
9.
ACS Med Chem Lett ; 10(1): 137-141, 2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30655961

ABSTRACT

Malaria is a major tropical disease where important needs are to mitigate symptoms and to prevent the establishment of infection. Cyclopeptides containing N-methyl amino acids with in vitro activity against erythrocytic forms as well as liver stage are presented. The synthesis, parasitological characterization, physicochemical properties, in vivo evaluation, and mice pharmacokinetics are described.

10.
Article in English | MEDLINE | ID: mdl-30559138

ABSTRACT

A series of 4-amino 2-anilinoquinazolines optimized for activity against the most lethal malaria parasite of humans, Plasmodium falciparum, was evaluated for activity against other human Plasmodium parasites and related apicomplexans that infect humans and animals. Four of the most promising compounds from the 4-amino 2-anilinoquinazoline series were equally as effective against the asexual blood stages of the zoonotic P. knowlesi, suggesting that they could also be effective against the closely related P. vivax, another important human pathogen. The 2-anilinoquinazoline compounds were also potent against an array of P. falciparum parasites resistant to clinically available antimalarial compounds, although slightly less so than against the drug-sensitive 3D7 parasite line. The apicomplexan parasites Toxoplasma gondii, Babesia bovis, and Cryptosporidium parvum were less sensitive to the 2-anilinoquinazoline series with a 50% effective concentration generally in the low micromolar range, suggesting that the yet to be discovered target of these compounds is absent or highly divergent in non-Plasmodium parasites. The 2-anilinoquinazoline compounds act as rapidly as chloroquine in vitro and when tested in rodents displayed a half-life that contributed to the compound's capacity to clear P. falciparum blood stages in a humanized mouse model. At a dose of 50 mg/kg of body weight, adverse effects to the humanized mice were noted, and evaluation against a panel of experimental high-risk off targets indicated some potential off-target activity. Further optimization of the 2-anilinoquinazoline antimalarial class will concentrate on improving in vivo efficacy and addressing adverse risk.


Subject(s)
Aniline Compounds/pharmacology , Antiparasitic Agents/pharmacology , Babesia bovis/drug effects , Cryptosporidium parvum/drug effects , Plasmodium falciparum/drug effects , Quinazolines/pharmacology , Toxoplasma/drug effects , Animals , Antimalarials/pharmacology , Cell Line , Chloroquine/pharmacology , Disease Models, Animal , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Parasitic Sensitivity Tests , Rats , Rats, Sprague-Dawley
11.
Nat Commun ; 8(1): 430, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874661

ABSTRACT

To combat drug resistance, new chemical entities are urgently required for use in next generation anti-malarial combinations. We report here the results of a medicinal chemistry programme focused on an imidazopyridine series targeting the Plasmodium falciparum cyclic GMP-dependent protein kinase (PfPKG). The most potent compound (ML10) has an IC50 of 160 pM in a PfPKG kinase assay and inhibits P. falciparum blood stage proliferation in vitro with an EC50 of 2.1 nM. Oral dosing renders blood stage parasitaemia undetectable in vivo using a P. falciparum SCID mouse model. The series targets both merozoite egress and erythrocyte invasion, but crucially, also blocks transmission of mature P. falciparum gametocytes to Anopheles stephensi mosquitoes. A co-crystal structure of PvPKG bound to ML10, reveals intimate molecular contacts that explain the high levels of potency and selectivity we have measured. The properties of this series warrant consideration for further development to produce an antimalarial drug.Protein kinases are promising drug targets for treatment of malaria. Here, starting with a medicinal chemistry approach, Baker et al. generate an imidazopyridine that selectively targets Plasmodium falciparum PKG, inhibits blood stage parasite growth in vitro and in mice and blocks transmission to mosquitoes.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Imidazoles/therapeutic use , Malaria/enzymology , Malaria/transmission , Pyridines/therapeutic use , Animals , Cell Line , Crystallography, X-Ray , Culicidae , Cyclic GMP-Dependent Protein Kinases/chemistry , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Female , Humans , Imidazoles/pharmacology , Life Cycle Stages/drug effects , Malaria/drug therapy , Mice, Inbred BALB C , Models, Molecular , Plasmodium chabaudi/drug effects , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Treatment Outcome
12.
J Med Chem ; 60(13): 5889-5908, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28635296

ABSTRACT

Carboxamide pyrazinyloxy benzoxaboroles were investigated with the goal to identify a molecule with satisfactory antimalarial activity, physicochemical properties, pharmacokinetic profile, in vivo efficacy, and safety profile. This optimization effort discovered 46, which met our target candidate profile. Compound 46 had excellent activity against cultured Plasmodium falciparum, and in vivo against P. falciparum and P. berghei in infected mice. It exhibited good PK properties in mice, rats, and dogs. It was highly active against the other 11 P. falciparum strains, which are mostly resistant to chloroquine and pyrimethamine. The rapid parasite in vitro reduction and in vivo parasite clearance profile of 46 were similar to those of artemisinin and chloroquine, two rapid-acting antimalarials. It was nongenotoxic in an Ames assay, an in vitro micronucleus assay, and an in vivo rat micronucleus assay when dosed orally up to 2000 mg/kg. The combined properties of this novel benzoxaborole support its progression to preclinical development.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Boron Compounds/chemistry , Boron Compounds/pharmacology , Malaria/drug therapy , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Amides/chemistry , Amides/pharmacokinetics , Amides/pharmacology , Amides/therapeutic use , Animals , Antimalarials/pharmacokinetics , Antimalarials/therapeutic use , Boron Compounds/pharmacokinetics , Boron Compounds/therapeutic use , Dogs , Female , Humans , Malaria, Falciparum/drug therapy , Male , Mice , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
13.
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
14.
Antimicrob Agents Chemother ; 60(10): 5949-56, 2016 10.
Article in English | MEDLINE | ID: mdl-27458216

ABSTRACT

The discovery and development of new antimalarial drugs are becoming imperative because of the spread of resistance to current clinical treatments. The lack of robustly validated antimalarial targets and the difficulties with the building in of whole-cell activity in screening hits are hampering target-based approaches. However, phenotypic screens of structurally diverse molecule libraries are offering new opportunities for the identification of novel antimalarials. Several methodologies can be used to determine the whole-cell in vitro potencies of antimalarial hits. The [(3)H]hypoxanthine incorporation assay is considered the "gold standard" assay for measurement of the activity of antimalarial compounds against intraerythrocytic forms of Plasmodium falciparum However, the method has important limitations, as the assay is not amenable for high-throughput screening since it remains associated with the 96-well plate format. We have overcome this drawback by adapting the [(3)H]hypoxanthine incorporation method to a 384-well high-density format by coupling a homogeneous scintillation proximity assay (SPA) and thus eliminating the limiting filtration step. This SPA has been validated using a diverse set of 1,000 molecules, including both a representative set from the Tres Cantos Antimalarial Set (TCAMS) of compounds and molecules inactive against whole cells. The results were compared with those from the P. falciparum lactate dehydrogenase whole-cell assay, another method that is well established as a surrogate for parasite growth and is amenable for high-throughput screening. The results obtained demonstrate that the SPA-based [(3)H]hypoxanthine incorporation assay is a suitable design that is adaptable to high-throughput antimalarial drug screening and that maintains the features, robustness, and reliability of the standard filtration hypoxanthine incorporation method.


Subject(s)
Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Plasmodium falciparum/growth & development , Hypoxanthine/analysis , Hypoxanthine/metabolism , Inhibitory Concentration 50 , Plasmodium falciparum/drug effects , Reproducibility of Results , Tritium
15.
J Med Chem ; 58(16): 6448-55, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26222445

ABSTRACT

Screening of the GSK corporate collection, some 1.9 million compounds, against Plasmodium falciparum (Pf), revealed almost 14000 active hits that are now known as the Tres Cantos Antimalarial Set (TCAMS). Followup work by Calderon et al. clustered and computationally filtered the TCAMS through a variety of criteria and reported 47 series containing a total of 522 compounds. From this enhanced set, we identified the carbamoyl triazole TCMDC-134379 (1), a known serine protease inhibitor, as an excellent starting point for SAR profiling. Lead optimization of 1 led to several molecules with improved antimalarial potency, metabolic stabilities in mouse and human liver microsomes, along with acceptable cytotoxicity profiles. Analogue 44 displayed potent in vitro activity (IC50 = 10 nM) and oral activity in a SCID mouse model of Pf infection with an ED50 of 100 and ED90 of between 100 and 150 mg kg(-1), respectively. The results presented encourage further investigations to identify the target of these highly active compounds.


Subject(s)
Antimalarials/chemical synthesis , Antimalarials/pharmacology , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacology , Triazoles/chemical synthesis , Triazoles/pharmacology , Animals , Antimalarials/metabolism , High-Throughput Screening Assays , Humans , In Vitro Techniques , Malaria/drug therapy , Malaria/psychology , Malaria, Falciparum/drug therapy , Mice , Mice, SCID , Microsomes, Liver/metabolism , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Serine Proteinase Inhibitors/metabolism , Structure-Activity Relationship , Triazoles/metabolism
16.
J Med Chem ; 58(13): 5344-54, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26067904

ABSTRACT

A series of 6-hetaryloxy benzoxaborole compounds was designed and synthesized for a structure-activity relationship (SAR) investigation to assess the changes in antimalarial activity which result from 6-aryloxy structural variation, substituent modification on the pyrazine ring, and optimization of the side chain ester group. This SAR study discovered highly potent 6-(2-(alkoxycarbonyl)pyrazinyl-5-oxy)-1,3-dihydro-1-hydroxy-2,1-benzoxaboroles (9, 27-34) with IC50s = 0.2-22 nM against cultured Plasmodium falciparum W2 and 3D7 strains. Compound 9 also demonstrated excellent in vivo efficacy against P. berghei in infected mice (ED90 = 7.0 mg/kg).


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Boron Compounds/chemistry , Boron Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Malaria, Falciparum/drug therapy , Microsomes, Liver/drug effects , Plasmodium falciparum/drug effects , Pyrazines/chemistry , Pyrazines/pharmacology , Animals , Cell Survival/drug effects , Female , Humans , Jurkat Cells , Malaria, Falciparum/parasitology , Mice , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship
17.
J Med Chem ; 58(11): 4573-80, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25906200

ABSTRACT

Malaria continues to be a major global health problem, being particularly devastating in the African population under the age of five. Artemisinin-based combination therapies (ACTs) are the first-line treatment recommended by the WHO to treat Plasmodium falciparum malaria, but clinical resistance against them has already been reported. As a consequence, novel chemotypes are urgently needed. Herein we report a novel, in vivo active, fast-acting antimalarial chemotype based on a benzimidazole core. This discovery is the result of a medicinal chemistry plan focused on improving the developability profile of an antichlamydial chemical class previously reported by our group.


Subject(s)
Antimalarials/chemical synthesis , Antimalarials/pharmacology , Benzamides/chemical synthesis , Benzamides/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Cell Proliferation/drug effects , Drug Design , Amides/chemical synthesis , Amides/pharmacokinetics , Amides/pharmacology , Animals , Antimalarials/pharmacokinetics , Benzamides/pharmacokinetics , Benzimidazoles/pharmacokinetics , Cells, Cultured , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Female , Humans , Malaria, Falciparum , Mice, Inbred NOD , Mice, SCID , Models, Molecular , Molecular Structure , Plasmodium falciparum , Structure-Activity Relationship , Tissue Distribution
18.
Antimicrob Agents Chemother ; 59(2): 950-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25421480

ABSTRACT

Current antimalarials are under continuous threat due to the relentless development of drug resistance by malaria parasites. We previously reported promising in vitro parasite-killing activity with the histone methyltransferase inhibitor BIX-01294 and its analogue TM2-115. Here, we further characterize these diaminoquinazolines for in vitro and in vivo efficacy and pharmacokinetic properties to prioritize and direct compound development. BIX-01294 and TM2-115 displayed potent in vitro activity, with 50% inhibitory concentrations (IC50s) of <50 nM against drug-sensitive laboratory strains and multidrug-resistant field isolates, including artemisinin-refractory Plasmodium falciparum isolates. Activities against ex vivo clinical isolates of both P. falciparum and Plasmodium vivax were similar, with potencies of 300 to 400 nM. Sexual-stage gametocyte inhibition occurs at micromolar levels; however, mature gametocyte progression to gamete formation is inhibited at submicromolar concentrations. Parasite reduction ratio analysis confirms a high asexual-stage rate of killing. Both compounds examined displayed oral efficacy in in vivo mouse models of Plasmodium berghei and P. falciparum infection. The discovery of a rapid and broadly acting antimalarial compound class targeting blood stage infection, including transmission stage parasites, and effective against multiple malaria-causing species reveals the diaminoquinazoline scaffold to be a very promising lead for development into greatly needed novel therapies to control malaria.


Subject(s)
Antimalarials/therapeutic use , Azepines/therapeutic use , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Malaria/drug therapy , Quinazolines/therapeutic use , Animals , Antimalarials/chemistry , Azepines/chemistry , Female , Hep G2 Cells , Histone Methyltransferases , Humans , Malaria, Falciparum/drug therapy , Mice , Mice, SCID , Plasmodium berghei/drug effects , Plasmodium berghei/pathogenicity , Plasmodium falciparum/drug effects , Plasmodium falciparum/pathogenicity , Quinazolines/chemistry
19.
Sci Transl Med ; 5(177): 177ra37, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23515079

ABSTRACT

The goal for developing new antimalarial drugs is to find a molecule that can target multiple stages of the parasite's life cycle, thus impacting prevention, treatment, and transmission of the disease. The 4(1H)-quinolone-3-diarylethers are selective potent inhibitors of the parasite's mitochondrial cytochrome bc1 complex. These compounds are highly active against the human malaria parasites Plasmodium falciparum and Plasmodium vivax. They target both the liver and blood stages of the parasite as well as the forms that are crucial for disease transmission, that is, the gametocytes, the zygote, the ookinete, and the oocyst. Selected as a preclinical candidate, ELQ-300 has good oral bioavailability at efficacious doses in mice, is metabolically stable, and is highly active in blocking transmission in rodent models of malaria. Given its predicted low dose in patients and its predicted long half-life, ELQ-300 has potential as a new drug for the treatment, prevention, and, ultimately, eradication of human malaria.


Subject(s)
Antimalarials/pharmacology , Quinolones/pharmacology , Animals , Antimalarials/chemistry , Atovaquone/chemistry , Atovaquone/pharmacology , Drug Resistance , Drug Synergism , Life Cycle Stages/drug effects , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Mice , Plasmodium falciparum/drug effects , Plasmodium vivax/drug effects , Proguanil/chemistry , Proguanil/pharmacology , Pyridones/chemistry , Pyridones/pharmacology , Quinolones/chemistry
20.
PLoS One ; 7(2): e30949, 2012.
Article in English | MEDLINE | ID: mdl-22383983

ABSTRACT

Chemotherapy is still the cornerstone for malaria control. Developing drugs against Plasmodium parasites and monitoring their efficacy requires methods to accurately determine the parasite killing rate in response to treatment. Commonly used techniques essentially measure metabolic activity as a proxy for parasite viability. However, these approaches are susceptible to artefacts, as viability and metabolism are two parameters that are coupled during the parasite life cycle but can be differentially affected in response to drug actions. Moreover, traditional techniques do not allow to measure the speed-of-action of compounds on parasite viability, which is an essential efficacy determinant. We present here a comprehensive methodology to measure in vitro the direct effect of antimalarial compounds over the parasite viability, which is based on limiting serial dilution of treated parasites and re-growth monitoring. This methodology allows to precisely determine the killing rate of antimalarial compounds, which can be quantified by the parasite reduction ratio and parasite clearance time, which are key mode-of-action parameters. Importantly, we demonstrate that this technique readily permits to determine compound killing activities that might be otherwise missed by traditional, metabolism-based techniques. The analysis of a large set of antimalarial drugs reveals that this viability-based assay allows to discriminate compounds based on their antimalarial mode-of-action. This approach has been adapted to perform medium throughput screening, facilitating the identification of fast-acting antimalarial compounds, which are crucially needed for the control and possibly the eradication of malaria.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Animals , Cell Culture Techniques/methods , Drug Evaluation, Preclinical , Humans , Hypoxanthine/chemistry , Inhibitory Concentration 50 , Reproducibility of Results , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...