Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 175
Filtrer
1.
J Med Chem ; 2024 Oct 03.
Article de Anglais | MEDLINE | ID: mdl-39361938

RÉSUMÉ

Inhibition of the lactate transporter PfFNT is a valid novel mode of action against malaria parasites. Current pyridine-substituted pentafluoro-3-hydroxy-pent-2-en-1-ones act as substrate analogs with submicromolar EC50 in vitro, and >99.7% activity in mice. The recently solved structure of a PfFNT-inhibitor complex visualized the binding mode. Here, we extended the inhibitor layout by series of amine- and anilide-linked pyridine p-substituents to generate additional interactions in the cytoplasmic vestibule. Virtual docking indicated hydrogen bonding to Tyr31 and Ser102. Fluorescence cross-correlation spectroscopy yielded respectively enhanced target affinity. Strikingly, the in vitro activity increased by 1 order of magnitude to 14.8 nM at negligible cytotoxicity. While p-amine substitutions were rapidly metabolized, the more stable p-acetanilide cleared 99.7% of parasites at 4 × 50 mg kg-1 in a mouse malaria model. Future stabilization of the p-substitution against metabolism may translate the gain in in vitro potency to the in vivo situation.

2.
Chemistry ; : e202403408, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39348488

RÉSUMÉ

A total synthesis of the enantiopure syn,syn-tosyl-samroiyotmycin A, a C2-symmetric 20-membered antimalarial macrodiolide with syn,syn-configuration of the 8,24-dihydroxy-9,25-dimethyl units and it's enantiopure anti,anti-derivative is described. The synthesis was accomplished utilizing a linear approach in 7 steps and 3 % overall yield via a sequence of diastereoselective methylation of SuperQuat oxazolidinone auxiliary, cross metathesis and Yamaguchi macrolactonization of fully functionalized seco-acids. By a similar approach we gained access to several samroiyotmycin analogues and precursors. Antimalarial activity was tested on multi-resistant (K1) and sensitive (Nf54) P. falciparum strains providing insight into structure activity relationships. Both tosyl-oxazol unit as well as the syn-configuration of the two contiguous stereogenic centers turned out to be beneficial for antiplasmodial activity. For instance, syn,syn-tosyl-samroiyotmycin A showed 3.4 times higher activities than the "tosyl-free" natural product.

3.
Cell Chem Biol ; 31(8): 1503-1517.e19, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39084225

RÉSUMÉ

Malaria remains a global health concern as drug resistance threatens treatment programs. We identified a piperidine carboxamide (SW042) with anti-malarial activity by phenotypic screening. Selection of SW042-resistant Plasmodium falciparum (Pf) parasites revealed point mutations in the Pf_proteasome ß5 active-site (Pfß5). A potent analog (SW584) showed efficacy in a mouse model of human malaria after oral dosing. SW584 had a low propensity to generate resistance (minimum inoculum for resistance [MIR] >109) and was synergistic with dihydroartemisinin. Pf_proteasome purification was facilitated by His8-tag introduction onto ß7. Inhibition of Pfß5 correlated with parasite killing, without inhibiting human proteasome isoforms or showing cytotoxicity. The Pf_proteasome_SW584 cryoelectron microscopy (cryo-EM) structure showed that SW584 bound non-covalently distal from the catalytic threonine, in an unexplored pocket at the ß5/ß6/ß3 subunit interface that has species differences between Pf and human proteasomes. Identification of a reversible, species selective, orally active series with low resistance propensity provides a path for drugging this essential target.


Sujet(s)
Antipaludiques , Pipéridines , Plasmodium falciparum , Inhibiteurs du protéasome , Pipéridines/composition chimique , Pipéridines/pharmacologie , Plasmodium falciparum/effets des médicaments et des substances chimiques , Plasmodium falciparum/enzymologie , Animaux , Antipaludiques/pharmacologie , Antipaludiques/composition chimique , Humains , Souris , Inhibiteurs du protéasome/pharmacologie , Inhibiteurs du protéasome/composition chimique , Inhibiteurs du protéasome/synthèse chimique , Administration par voie orale , Proteasome endopeptidase complex/métabolisme , Paludisme/traitement médicamenteux , Paludisme/parasitologie , Amides/composition chimique , Amides/pharmacologie , Amides/synthèse chimique , Paludisme à Plasmodium falciparum/traitement médicamenteux , Femelle , Structure moléculaire
4.
Eur J Med Chem ; 276: 116677, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-39024967

RÉSUMÉ

Emerging resistance to current antimalarials is reducing their effectiveness and therefore there is a need to develop new antimalarial therapies. Toward this goal, high throughput screens against the P. falciparum asexual parasite identified the pyrazolopyridine 4-carboxamide scaffold. Structure-activity relationship analysis of this chemotype defined that the N1-tert-butyl group and aliphatic foliage in the 3- and 6-positions were necessary for activity, while the inclusion of a 7'-aza-benzomorpholine on the 4-carboxamide motif resulted in potent anti-parasitic activity and increased aqueous solubility. A previous report that resistance to the pyrazolopyridine class is associated with the ABCI3 transporter was confirmed, with pyrazolopyridine 4-carboxamides showing an increase in potency against parasites when the ABCI3 transporter was knocked down. The low metabolic stability intrinsic to the pyrazolopyridine scaffold and the slow rate by which the compounds kill asexual parasites resulted in poor performance in a P. berghei asexual blood stage mouse model. Lowering the risk of resistance and mitigating the metabolic stability and cytochrome P450 inhibition will be challenges in the future development of the pyrazolopyrimidine antimalarial class.


Sujet(s)
Antipaludiques , Plasmodium falciparum , Pyrazoles , Pyridines , Antipaludiques/pharmacologie , Antipaludiques/composition chimique , Antipaludiques/synthèse chimique , Plasmodium falciparum/effets des médicaments et des substances chimiques , Relation structure-activité , Pyrazoles/composition chimique , Pyrazoles/pharmacologie , Pyrazoles/synthèse chimique , Animaux , Pyridines/pharmacologie , Pyridines/composition chimique , Pyridines/synthèse chimique , Souris , Tests de sensibilité parasitaire , Structure moléculaire , Résistance aux substances/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Humains
5.
Elife ; 132024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38976500

RÉSUMÉ

New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum (PfA-M1) and Plasmodium vivax (PvA-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets PfA-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on PfA-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of PfA-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.


Sujet(s)
Antipaludiques , Plasmodium falciparum , Plasmodium vivax , Protéomique , Protéines de protozoaire , Antipaludiques/pharmacologie , Antipaludiques/composition chimique , Plasmodium falciparum/enzymologie , Plasmodium falciparum/effets des médicaments et des substances chimiques , Plasmodium vivax/enzymologie , Plasmodium vivax/effets des médicaments et des substances chimiques , Humains , Protéines de protozoaire/métabolisme , Protéines de protozoaire/antagonistes et inhibiteurs , Protéines de protozoaire/composition chimique , Protéomique/méthodes , Aminopeptidases/métabolisme , Aminopeptidases/antagonistes et inhibiteurs , Aminopeptidases/composition chimique
6.
J Med Chem ; 67(13): 11401-11420, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38918002

RÉSUMÉ

Structure-activity relationship studies of 2,8-disubstituted-1,5-naphthyridines, previously reported as potent inhibitors of Plasmodium falciparum (Pf) phosphatidylinositol-4-kinase ß (PI4K), identified 1,5-naphthyridines with basic groups at 8-position, which retained Plasmodium PI4K inhibitory activity but switched primary mode of action to the host hemoglobin degradation pathway through inhibition of hemozoin formation. These compounds showed minimal off-target inhibitory activity against the human phosphoinositide kinases and MINK1 and MAP4K kinases, which were associated with the teratogenicity and testicular toxicity observed in rats for the PfPI4K inhibitor clinical candidate MMV390048. A representative compound from the series retained activity against field isolates and lab-raised drug-resistant strains of Pf. It was efficacious in the humanized NSG mouse malaria infection model at a single oral dose of 32 mg/kg. This compound was nonteratogenic in the zebrafish embryo model of teratogenicity and has a low predicted human dose, indicating that this series has the potential to deliver a preclinical candidate for malaria.


Sujet(s)
1-Phosphatidylinositol 4-kinase , Antipaludiques , Hémoprotéines , Naphtyridines , Plasmodium falciparum , Danio zébré , Plasmodium falciparum/effets des médicaments et des substances chimiques , Animaux , Naphtyridines/pharmacologie , Naphtyridines/composition chimique , Naphtyridines/synthèse chimique , Naphtyridines/usage thérapeutique , Antipaludiques/pharmacologie , Antipaludiques/composition chimique , Antipaludiques/synthèse chimique , 1-Phosphatidylinositol 4-kinase/antagonistes et inhibiteurs , 1-Phosphatidylinositol 4-kinase/métabolisme , Humains , Relation structure-activité , Hémoprotéines/antagonistes et inhibiteurs , Hémoprotéines/métabolisme , Souris , Rats , Paludisme à Plasmodium falciparum/traitement médicamenteux , Mâle , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique
7.
mBio ; 15(6): e0096624, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38717141

RÉSUMÉ

To combat the global burden of malaria, development of new drugs to replace or complement current therapies is urgently required. Here, we show that the compound MMV1557817 is a selective, nanomolar inhibitor of both Plasmodium falciparum and Plasmodium vivax aminopeptidases M1 and M17, leading to inhibition of end-stage hemoglobin digestion in asexual parasites. MMV1557817 can kill sexual-stage P. falciparum, is active against murine malaria, and does not show any shift in activity against a panel of parasites resistant to other antimalarials. MMV1557817-resistant P. falciparum exhibited a slow growth rate that was quickly outcompeted by wild-type parasites and were sensitized to the current clinical drug, artemisinin. Overall, these results confirm MMV1557817 as a lead compound for further drug development and highlights the potential of dual inhibition of M1 and M17 as an effective multi-species drug-targeting strategy.IMPORTANCEEach year, malaria infects approximately 240 million people and causes over 600,000 deaths, mostly in children under 5 years of age. For the past decade, artemisinin-based combination therapies have been recommended by the World Health Organization as the standard malaria treatment worldwide. Their widespread use has led to the development of artemisinin resistance in the form of delayed parasite clearance, alongside the rise of partner drug resistance. There is an urgent need to develop and deploy new antimalarial agents with novel targets and mechanisms of action. Here, we report a new and potent antimalarial compound, known as MMV1557817, and show that it targets multiple stages of the malaria parasite lifecycle, is active in a preliminary mouse malaria model, and has a novel mechanism of action. Excitingly, resistance to MMV15578117 appears to be self-limiting, suggesting that development of the compound may provide a new class of antimalarial.


Sujet(s)
Aminopeptidases , Antipaludiques , Plasmodium falciparum , Plasmodium vivax , Antipaludiques/pharmacologie , Plasmodium falciparum/effets des médicaments et des substances chimiques , Plasmodium falciparum/enzymologie , Animaux , Souris , Plasmodium vivax/effets des médicaments et des substances chimiques , Plasmodium vivax/enzymologie , Aminopeptidases/antagonistes et inhibiteurs , Aminopeptidases/métabolisme , Résistance aux substances , Humains , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/parasitologie , Protéines de protozoaire/antagonistes et inhibiteurs , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique , Femelle
8.
Res Sq ; 2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38746424

RÉSUMÉ

New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum ( Pf A-M1) and Plasmodium vivax ( Pv A-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets Pf A-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on Pf A-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of Pf A-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.

9.
Phytomedicine ; 129: 155644, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38761524

RÉSUMÉ

BACKGROUND: A global death toll of 608,000 in 2022 and emerging parasite resistance to artemisinin, the mainstay of antimalarial chemotherapy derived from the Chinese herb Artemisia annua, urge the development of novel antimalarials. A clinical trial has found high antimalarial potency for aqueous extracts of A. annua as well as its African counterpart Artemisia afra, which contains only trace amounts of artemisinin. The artemisinin-independent antimalarial activity of A. afra points to the existence of other antimalarials present in the plant. However, the publication was retracted due to ethical and methodological concerns in the trial, so the only evidence for antimalarial activity of A. afra is built on in vitro studies reporting efficacy only in the microgram per milliliter range. HYPOTHESIS: Our study aims to shed more light on the controversy around the antimalarial activity of A. afra by assessing its efficacy in mice. In particular, we are testing the hypothesis that A. afra contains a pro-drug that is inactive in vitro but active in vivo after metabolization by the mammalian host. METHODS: Plasmodium berghei-infected mice were treated once or thrice (on three consecutive days) with various doses of A. afra, A. annua, or pure artemisinin. RESULTS: Aqueous powder suspensions of A. annua but not A. afra showed antimalarial activity in mice. CONCLUSION: Our experiments conducted in mice do not support the pro-drug hypothesis.


Sujet(s)
Antipaludiques , Artemisia , Artémisinines , Paludisme , Extraits de plantes , Plasmodium berghei , Poudres , Antipaludiques/pharmacologie , Animaux , Artemisia/composition chimique , Paludisme/traitement médicamenteux , Plasmodium berghei/effets des médicaments et des substances chimiques , Artémisinines/pharmacologie , Souris , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Artemisia annua/composition chimique , Suspensions , Mâle
10.
Front Cell Infect Microbiol ; 14: 1396786, 2024.
Article de Anglais | MEDLINE | ID: mdl-38746786

RÉSUMÉ

Antimalarial resistance to the first-line partner drug piperaquine (PPQ) threatens the effectiveness of artemisinin-based combination therapy. In vitro piperaquine resistance is characterized by incomplete growth inhibition, i.e. increased parasite growth at higher drug concentrations. However, the 50% inhibitory concentrations (IC50) remain relatively stable across parasite lines. Measuring parasite viability of a drug-resistant Cambodian Plasmodium falciparum isolate in a parasite reduction ratio (PRR) assay helped to better understand the resistance phenotype towards PPQ. In this parasite isolate, incomplete growth inhibition translated to only a 2.5-fold increase in IC50 but a dramatic decrease of parasite killing in the PRR assay. Hence, this pilot study reveals the potential of in vitro parasite viability assays as an important, additional tool when it comes to guiding decision-making in preclinical drug development and post approval. To the best of our knowledge, this is the first time that a compound was tested against a drug-resistant parasite in the in vitro PRR assay.


Sujet(s)
Antipaludiques , Résistance aux substances , Concentration inhibitrice 50 , Paludisme à Plasmodium falciparum , Plasmodium falciparum , Quinoléines , Plasmodium falciparum/effets des médicaments et des substances chimiques , Plasmodium falciparum/croissance et développement , Quinoléines/pharmacologie , Antipaludiques/pharmacologie , Humains , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/parasitologie , Tests de sensibilité parasitaire , Projets pilotes , Artémisinines/pharmacologie
11.
ACS Infect Dis ; 10(4): 1286-1297, 2024 04 12.
Article de Anglais | MEDLINE | ID: mdl-38556981

RÉSUMÉ

Malaria is caused by parasites of the Plasmodium genus and remains one of the most pressing human health problems. The spread of parasites resistant to or partially resistant to single or multiple drugs, including frontline antimalarial artemisinin and its derivatives, poses a serious threat to current and future malaria control efforts. In vitro drug assays are important for identifying new antimalarial compounds and monitoring drug resistance. Due to its robustness and ease of use, the [3H]-hypoxanthine incorporation assay is still considered a gold standard and is widely applied, despite limited sensitivity and the dependence on radioactive material. Here, we present a first-of-its-kind chemiluminescence-based antimalarial drug screening assay. The effect of compounds on P. falciparum is monitored by using a dioxetane-based substrate (AquaSpark ß-D-galactoside) that emits high-intensity luminescence upon removal of a protective group (ß-D-galactoside) by a transgenic ß-galactosidase reporter enzyme. This biosensor enables highly sensitive, robust, and cost-effective detection of asexual, intraerythrocytic P. falciparum parasites without the need for parasite enrichment, washing, or purification steps. We are convinced that the ultralow detection limit of less than 100 parasites of the presented biosensor system will become instrumental in malaria research, including but not limited to drug screening.


Sujet(s)
Antipaludiques , Antifoliques , Paludisme à Plasmodium falciparum , Paludisme , Humains , Antipaludiques/pharmacologie , Plasmodium falciparum , Paludisme/traitement médicamenteux , Paludisme à Plasmodium falciparum/parasitologie , Antifoliques/pharmacologie , Galactoside/pharmacologie , Galactoside/usage thérapeutique
12.
ACS Med Chem Lett ; 15(4): 463-469, 2024 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-38628794

RÉSUMÉ

Toward addressing the cardiotoxicity liability associated with the antimalarial drug astemizole (AST, hERG IC50 = 0.0042 µM) and its derivatives, we designed and synthesized analogues based on compound 1 (Pf NF54 IC50 = 0.012 µM; hERG IC50 = 0.63 µM), our previously identified 3-trifluoromethyl-1,2,4-oxadiazole AST analogue. Compound 11 retained in vitro multistage antiplasmodium activity (ABS PfNF54 IC50 = 0.017 µM; gametocytes PfiGc/PfLGc IC50 = 1.24/1.39 µM, and liver-stage PbHepG2 IC50 = 2.30 µM), good microsomal metabolic stability (MLM CLint < 11 µL·min-1·mg-1, EH < 0.33), and solubility (150 µM). It shows a ∼6-fold and >6000-fold higher selectivity against human ether-á-go-go-related gene higher selectively potential over hERG relative to 1 and AST, respectively. Despite the excellent in vitro antiplasmodium activity profile, in vivo efficacy in the Plasmodium berghei mouse infection model was diminished, attributable to suboptimal oral bioavailability (F = 14.9%) at 10 mg·kg-1 resulting from poor permeability (log D7.4 = -0.82). No cross-resistance was observed against 44 common Pf mutant lines, suggesting activity via a novel mechanism of action.

13.
Eur J Med Chem ; 270: 116354, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38554474

RÉSUMÉ

Malaria is a devastating disease that causes significant morbidity worldwide. The development of new antimalarial chemotypes is urgently needed because of the emergence of resistance to frontline therapies. Independent phenotypic screening campaigns against the Plasmodium asexual parasite, including our own, identified the aryl amino acetamide hit scaffold. In a prior study, we identified the STAR-related lipid transfer protein (PfSTART1) as the molecular target of this antimalarial chemotype. In this study, we combined structural elements from the different aryl acetamide hit subtypes and explored the structure-activity relationship. It was shown that the inclusion of an endocyclic nitrogen, to generate the tool compound WJM-715, improved aqueous solubility and modestly improved metabolic stability in rat hepatocytes. Metabolic stability in human liver microsomes remains a challenge for future development of the aryl acetamide class, which was underscored by modest systemic exposure and a short half-life in mice. The optimized aryl acetamide analogs were cross resistant to parasites with mutations in PfSTART1, but not to other drug-resistant mutations, and showed potent binding to recombinant PfSTART1 by biophysical analysis, further supporting PfSTART1 as the likely molecular target. The optimized aryl acetamide analogue, WJM-715 will be a useful tool for further investigating the druggability of PfSTART1 across the lifecycle of the malaria parasite.


Sujet(s)
Antipaludiques , Protéines de transport , Paludisme à Plasmodium falciparum , Paludisme , Rats , Souris , Humains , Animaux , Antipaludiques/composition chimique , Plasmodium falciparum , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme/traitement médicamenteux , Acétamides/pharmacologie , Lipides
14.
Chimia (Aarau) ; 77(9): 593-602, 2023 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-38047835

RÉSUMÉ

Thanks to its expertise in clinical research, epidemiology, infectious diseases, microbiology, parasitology, public health, translational research and tropical medicine, coupled with deeply rooted partnerships with institutions in low- and middle-income countries (LMICs), the Swiss Tropical and Public Health Institute (Swiss TPH) has been a key contributor in many drug research and development consortia involving academia, pharma and product development partnerships. Our know-how of the maintenance of parasites and their life-cycles in the laboratory, plus our strong ties to research centres and disease control programme managers in LMICs with access to field sites and laboratories, have enabled systems for drug efficacy testing in vitro and in vivo, clinical research, and modelling to support the experimental approaches. Thus, Swiss TPH has made fundamental contributions towards the development of new drugs - and the better use of old drugs - for neglected tropical diseases and infectious diseases of poverty, such as Buruli ulcer, Chagas disease, food-borne trematodiasis (e.g. clonorchiasis, fascioliasis and opisthorchiasis), human African trypanosomiasis, leishmaniasis, malaria, schistosomiasis, soil-transmitted helminthiasis and tuberculosis. In this article, we show case the success stories of molecules to which Swiss TPH has made a substantial contribution regarding their use as anti-infective compounds with the ultimate aim to improve people's health and well-being.


Sujet(s)
Ulcère de Buruli , Maladies transmissibles , Médecine tropicale , Humains , Santé publique , Suisse , Maladies transmissibles/traitement médicamenteux
15.
ACS Infect Dis ; 9(10): 1964-1980, 2023 Oct 13.
Article de Anglais | MEDLINE | ID: mdl-37695781

RÉSUMÉ

We discovered dibenzannulated medium-ring keto lactams (11,12-dihydro-5H-dibenzo[b,g]azonine-6,13-diones) as a new antimalarial chemotype. Most of these had chromatographic LogD7.4 values ranging from <0 to 3 and good kinetic solubilities (12.5 to >100 µg/mL at pH 6.5). The more polar compounds in the series (LogD7.4 values of <2) had the best metabolic stability (CLint values of <50 µL/min/mg protein in human liver microsomes). Most of the compounds had relatively low cytotoxicity, with IC50 values >30 µM, and there was no correlation between antiplasmodial activity and cytotoxicity. The four most potent compounds had Plasmodium falciparum IC50 values of 4.2 to 9.4 nM and in vitro selectivity indices of 670 to >12,000. They were more than 4 orders-of-magnitude less potent against three other protozoal pathogens (Trypanosoma brucei rhodesiense, Trypanosoma cruzi, and Leishmania donovani) but did have relatively high potency against Toxoplasma gondii, with IC50 values ranging from 80 to 200 nM. These keto lactams are converted into their poorly soluble 4(1H)-quinolone transannular condensation products in vitro in culture medium and in vivo in mouse blood. The similar antiplasmodial potencies of three keto lactam-quinolone pairs suggest that the quinolones likely contribute to the antimalarial activity of the lactams.


Sujet(s)
Antipaludiques , Quinolinone , Trypanosoma cruzi , Souris , Animaux , Humains , Antipaludiques/pharmacologie , Antipaludiques/composition chimique , Lactames , Trypanosoma brucei rhodesiense
16.
ACS Infect Dis ; 9(9): 1695-1710, 2023 09 08.
Article de Anglais | MEDLINE | ID: mdl-37639221

RÉSUMÉ

With the resistance increasing to current antimalarial medicines, there is an urgent need to discover new drug targets and to develop new medicines against these targets. We therefore screened the Open Global Health Library of Merck KGaA, Darmstadt, Germany, of 250 compounds against the asexual blood stage of the deadliest malarial parasite Plasmodium falciparum, from which eight inhibitors with low micromolar potency were found. Due to its combined potencies against parasite growth and inhibition of red blood cell invasion, the pyridyl-furan compound OGHL250 was prioritized for further optimization. The potency of the series lead compound (WEHI-518) was improved 250-fold to low nanomolar levels against parasite blood-stage growth. Parasites selected for resistance to a related compound, MMV396797, were also resistant to WEHI-518 as well as KDU731, an inhibitor of the phosphatidylinositol kinase PfPI4KIIIB, suggesting that this kinase is the target of the pyridyl-furan series. Inhibition of PfPI4KIIIB blocks multiple stages of the parasite's life cycle and other potent inhibitors are currently under preclinical development. MMV396797-resistant parasites possess an E1316D mutation in PfPKI4IIIB that clusters with known resistance mutations of other inhibitors of the kinase. Building upon earlier studies that showed that PfPI4KIIIB inhibitors block the development of the invasive merozoite parasite stage, we show that members of the pyridyl-furan series also block invasion and/or the conversion of merozoites into ring-stage intracellular parasites through inhibition of protein secretion and export into red blood cells.


Sujet(s)
Parasites , Animaux , Plasmodium falciparum/génétique , Santé mondiale , Érythrocytes , Transport des protéines , Furanes
17.
Antimicrob Agents Chemother ; 67(8): e0035623, 2023 08 17.
Article de Anglais | MEDLINE | ID: mdl-37428074

RÉSUMÉ

Malaria parasites in the blood stage express a single transmembrane transport protein for the release of the glycolytic end product l-lactate/H+ from the cell. This transporter is a member of the strictly microbial formate-nitrite transporter (FNT) family and a novel putative drug target. Small, drug-like FNT inhibitors potently block lactate transport and kill Plasmodium falciparum parasites in culture. The protein structure of Plasmodium falciparum FNT (PfFNT) in complex with the inhibitor has been resolved and confirms its previously predicted binding site and its mode of action as a substrate analog. Here, we investigated the mutational plasticity and essentiality of the PfFNT target on a genetic level, and established its in vivo druggability using mouse malaria models. We found that, besides a previously identified PfFNT G107S resistance mutation, selection of parasites at 3 × IC50 (50% inhibitory concentration) gave rise to two new point mutations affecting inhibitor binding: G21E and V196L. Conditional knockout and mutation of the PfFNT gene showed essentiality in the blood stage, whereas no phenotypic defects in sexual development were observed. PfFNT inhibitors mainly targeted the trophozoite stage and exhibited high potency in P. berghei- and P. falciparum-infected mice. Their in vivo activity profiles were comparable to that of artesunate, demonstrating strong potential for the further development of PfFNT inhibitors as novel antimalarials.


Sujet(s)
Antipaludiques , Paludisme à Plasmodium falciparum , Parasites , Animaux , Souris , Transporteurs d'acides monocarboxyliques/composition chimique , Transporteurs d'acides monocarboxyliques/génétique , Plasmodium falciparum/génétique , Plasmodium falciparum/métabolisme , Paludisme à Plasmodium falciparum/parasitologie , Antipaludiques/pharmacologie , Antipaludiques/composition chimique , Parasites/métabolisme , Lactates/métabolisme , Plasmodium berghei/génétique , Plasmodium berghei/métabolisme , Protéines de protozoaire/métabolisme
18.
Pharmaceuticals (Basel) ; 16(2)2023 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-37009844

RÉSUMÉ

With artemisinin-resistant Plasmodium falciparum parasites emerging in Africa, the need for new antimalarial chemotypes is persistently high. The ideal pharmacodynamic parameters of a candidate drug are a rapid onset of action and a fast rate of parasite killing or clearance. To determine these parameters, it is essential to discriminate viable from nonviable parasites, which is complicated by the fact that viable parasites can be metabolically inactive, whilst dying parasites can still be metabolically active and morphologically unaffected. Standard growth inhibition assays, read out via microscopy or [3H] hypoxanthine incorporation, cannot reliably discriminate between viable and nonviable parasites. Conversely, the in vitro parasite reduction ratio (PRR) assay is able to measure viable parasites with high sensitivity. It provides valuable pharmacodynamic parameters, such as PRR, 99.9% parasite clearance time (PCT99.9%) and lag phase. Here we report the development of the PRR assay version 2 (V2), which comes with a shorter assay duration, optimized quality controls and an objective, automated analysis pipeline that systematically estimates PRR, PCT99.9% and lag time and returns meaningful secondary parameters such as the maximal killing rate of a drug (Emax) at the assayed concentration. These parameters can be fed directly into pharmacokinetic/pharmacodynamic models, hence aiding and standardizing lead selection, optimization, and dose prediction.

19.
ACS Infect Dis ; 9(3): 668-691, 2023 03 10.
Article de Anglais | MEDLINE | ID: mdl-36853190

RÉSUMÉ

The development of new antimalarials is required because of the threat of resistance to current antimalarial therapies. To discover new antimalarial chemotypes, we screened the Janssen Jumpstarter library against the P. falciparum asexual parasite and identified the 7-N-substituted-3-oxadiazole quinolone hit class. We established the structure-activity relationship and optimized the antimalarial potency. The optimized analog WJM228 (17) showed robust metabolic stability in vitro, although the aqueous solubility was limited. Forward genetic resistance studies uncovered that WJM228 targets the Qo site of cytochrome b (cyt b), an important component of the mitochondrial electron transport chain (ETC) that is essential for pyrimidine biosynthesis and an established antimalarial target. Profiling against drug-resistant parasites confirmed that WJM228 confers resistance to the Qo site but not Qi site mutations, and in a biosensor assay, it was shown to impact the ETC via inhibition of cyt b. Consistent with other cyt b targeted antimalarials, WJM228 prevented pre-erythrocytic parasite and male gamete development and reduced asexual parasitemia in a P. berghei mouse model of malaria. Correcting the limited aqueous solubility and the high susceptibility to cyt b Qo site resistant parasites found in the clinic will be major obstacles in the future development of the 3-oxadiazole quinolone antimalarial class.


Sujet(s)
Antipaludiques , Antifoliques , Paludisme à Plasmodium falciparum , Quinolinone , Animaux , Souris , Antipaludiques/pharmacologie , Cytochromes b , Antifoliques/métabolisme , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/parasitologie , Plasmodium falciparum , Quinolinone/pharmacologie
20.
ACS Infect Dis ; 9(3): 527-539, 2023 03 10.
Article de Anglais | MEDLINE | ID: mdl-36763526

RÉSUMÉ

Current malaria treatments are threatened by drug resistance, and new drugs are urgently needed. In a phenotypic screen for new antimalarials, we identified (S)-SW228703 ((S)-SW703), a tyrosine amide with asexual blood and liver stage activity and a fast-killing profile. Resistance to (S)-SW703 is associated with mutations in the Plasmodium falciparum cyclic amine resistance locus (PfCARL) and P. falciparum acetyl CoA transporter (PfACT), similarly to several other compounds that share features such as fast activity and liver-stage activity. Compounds with these resistance mechanisms are thought to act in the ER, though their targets are unknown. The tyramine of (S)-SW703 is shared with some reported PfCARL-associated compounds; however, we observed that strict S-stereochemistry was required for the activity of (S)-SW703, suggesting differences in the mechanism of action or binding mode. (S)-SW703 provides a new chemical series with broad activity for multiple life-cycle stages and a fast-killing mechanism of action, available for lead optimization to generate new treatments for malaria.


Sujet(s)
Antipaludiques , Paludisme à Plasmodium falciparum , Paludisme , Humains , Antipaludiques/pharmacologie , Antipaludiques/composition chimique , Plasmodium falciparum/génétique , Plasmodium falciparum/métabolisme , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme/traitement médicamenteux , Foie , Amines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE