Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(24): 16695-16715, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36507890

ABSTRACT

Iterative medicinal chemistry optimization of an ester-containing astemizole (AST) analogue 1 with an associated metabolic instability liability led to the identification of a highly potent 3-trifluoromethyl-1,2,4-oxadiazole analogue 23 (PfNF54 IC50 = 0.012 µM; PfK1 IC50 = 0.040 µM) displaying high microsomal metabolic stability (HLM CLint < 11.6 µL·min-1·mg-1) and > 1000-fold higher selectivity over hERG compared to AST. In addition to asexual blood stage activity, the compound also shows activity against liver and gametocyte life cycle stages and demonstrates in vivo efficacy in Plasmodium berghei-infected mice at 4 × 50 mg·kg-1 oral dose. Preliminary interrogation of the mode of action using live-cell microscopy and cellular heme speciation revealed that 23 could be affecting multiple processes in the parasitic digestive vacuole, with the possibility of a novel target at play in the organelles associated with it.


Subject(s)
Antimalarials , Malaria , Mice , Animals , Plasmodium berghei , Antimalarials/pharmacology , Antimalarials/therapeutic use , Astemizole/pharmacology , Astemizole/therapeutic use , Plasmodium falciparum/metabolism , Malaria/drug therapy , Malaria/parasitology , Disease Models, Animal
2.
J Med Chem ; 64(17): 12790-12807, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34414766

ABSTRACT

Phenotypic whole cell high-throughput screening of a ∼150,000 diverse set of compounds against Mycobacterium tuberculosis (Mtb) in cholesterol-containing media identified 1,3-diarylpyrazolyl-acylsulfonamide 1 as a moderately active hit. Structure-activity relationship (SAR) studies demonstrated a clear scope to improve whole cell potency to MIC values of <0.5 µM, and a plausible pharmacophore model was developed to describe the chemical space of active compounds. Compounds are bactericidal in vitro against replicating Mtb and retained activity against multidrug-resistant clinical isolates. Initial biology triage assays indicated cell wall biosynthesis as a plausible mode-of-action for the series. However, no cross-resistance with known cell wall targets such as MmpL3, DprE1, InhA, and EthA was detected, suggesting a potentially novel mode-of-action or inhibition. The in vitro and in vivo drug metabolism and pharmacokinetics profiles of several active compounds from the series were established leading to the identification of a compound for in vivo efficacy proof-of-concept studies.


Subject(s)
Antitubercular Agents/pharmacology , Cell Wall/metabolism , Mycobacterium tuberculosis/drug effects , Sulfonamides/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Drug Discovery , Hep G2 Cells , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry
3.
J Med Chem ; 64(13): 9444-9457, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34138573

ABSTRACT

Screening of a library of small polar molecules against Mycobacterium tuberculosis (Mtb) led to the identification of a potent benzoheterocyclic oxime carbamate hit series. This series was subjected to medicinal chemistry progression underpinned by structure-activity relationship studies toward identifying a compound for proof-of-concept studies and defining a lead optimization strategy. Carbamate and free oxime frontrunner compounds with good stability in liver microsomes and no hERG channel inhibition liability were identified and evaluated in vivo for pharmacokinetic properties. Mtb-mediated permeation and metabolism studies revealed that the carbamates were acting as prodrugs. Toward mechanism of action elucidation, selected compounds were tested in biology triage assays to assess their activity against known promiscuous targets. Taken together, these data suggest a novel yet unknown mode of action for these antitubercular hits.


Subject(s)
Antitubercular Agents/pharmacology , Carbamates/pharmacology , Heterocyclic Compounds/pharmacology , Mycobacterium tuberculosis/drug effects , Oximes/pharmacology , Antitubercular Agents/chemistry , Antitubercular Agents/metabolism , Carbamates/chemistry , Carbamates/metabolism , Dose-Response Relationship, Drug , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/metabolism , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium tuberculosis/metabolism , Oximes/chemistry , Oximes/metabolism , Structure-Activity Relationship
4.
J Med Chem ; 64(8): 5198-5215, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33844521

ABSTRACT

A novel series of antimalarial benzimidazole derivatives incorporating phenolic Mannich base side chains at the C2 position, which possess dual asexual blood and sexual stage activities, is presented. Structure-activity relationship studies revealed that the 1-benzylbenzimidazole analogues possessed submicromolar asexual blood and sexual stage activities in contrast to the 1H-benzimidazole analogues, which were only active against asexual blood stage (ABS) parasites. Further, the former demonstrated microtubule inhibitory activity in ABS parasites but more significantly in stage II/III gametocytes. In addition to being bona fide inhibitors of hemozoin formation, the 1H-benzimidazole analogues also showed inhibitory effects on microtubules. In vivo efficacy studies in Plasmodium berghei-infected mice revealed that the frontrunner compound 41 exhibited high efficacy (98% reduction in parasitemia) when dosed orally at 4 × 50 mg/kg. Generally, the compounds were noncytotoxic to mammalian cells.


Subject(s)
Antimalarials/chemistry , Benzimidazoles/chemistry , Hemeproteins/metabolism , Mannich Bases/chemistry , Microtubules/metabolism , Administration, Oral , Animals , Antimalarials/metabolism , Antimalarials/pharmacology , Antimalarials/therapeutic use , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Disease Models, Animal , Drug Design , Drug Resistance/drug effects , Drug Stability , Half-Life , Hemeproteins/drug effects , Life Cycle Stages/drug effects , Malaria/drug therapy , Malaria/parasitology , Male , Mice , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Microtubules/drug effects , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Structure-Activity Relationship
5.
J Med Chem ; 64(4): 2291-2309, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33573376

ABSTRACT

A novel diazaspiro[3.4]octane series was identified from a Plasmodium falciparum whole-cell high-throughput screening campaign. Hits displayed activity against multiple stages of the parasite lifecycle, which together with a novel sp3-rich scaffold provided an attractive starting point for a hit-to-lead medicinal chemistry optimization and biological profiling program. Structure-activity-relationship studies led to the identification of compounds that showed low nanomolar asexual blood-stage activity (<50 nM) together with strong gametocyte sterilizing properties that translated to transmission-blocking activity in the standard membrane feeding assay. Mechanistic studies through resistance selection with one of the analogues followed by whole-genome sequencing implicated the P. falciparum cyclic amine resistance locus in the mode of resistance.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Spiro Compounds/pharmacology , Animals , Anopheles/drug effects , Antimalarials/chemical synthesis , Antimalarials/metabolism , Female , Germ Cells/drug effects , High-Throughput Screening Assays , Humans , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Rats , Spiro Compounds/chemical synthesis , Spiro Compounds/metabolism , Structure-Activity Relationship
7.
J Med Chem ; 64(1): 719-740, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33395287

ABSTRACT

Phenotypic screening of a Medicines for Malaria Venture compound library against Mycobacterium tuberculosis (Mtb) identified a cluster of pan-active 2-pyrazolylpyrimidinones. The biology triage of these actives using various tool strains of Mtb suggested a novel mechanism of action. The compounds were bactericidal against replicating Mtb and retained potency against clinical isolates of Mtb. Although selected MmpL3 mutant strains of Mtb showed resistance to these compounds, there was no shift in the minimum inhibitory concentration (MIC) against a mmpL3 hypomorph, suggesting mutations in MmpL3 as a possible resistance mechanism for the compounds but not necessarily as the target. RNA transcriptional profiling and the checkerboard board 2D-MIC assay in the presence of varying concentrations of ferrous salt indicated perturbation of the Fe-homeostasis by the compounds. Structure-activity relationship studies identified potent compounds with good physicochemical properties and in vitro microsomal metabolic stability with moderate selectivity over cytotoxicity against mammalian cell lines.


Subject(s)
Antitubercular Agents/chemistry , Pyrimidinones/chemistry , Animals , Antitubercular Agents/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Half-Life , Humans , Iron/metabolism , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microsomes/metabolism , Mutation , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/isolation & purification , Pyrazoles/chemistry , Pyrimidinones/metabolism , Pyrimidinones/pharmacology , Rats , Structure-Activity Relationship
8.
ACS Infect Dis ; 7(1): 34-46, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33319990

ABSTRACT

Recent studies on 3,6-diphenylated imidazopyridazines have demonstrated impressive in vitro activity and in vivo efficacy in mouse models of malaria infection. Herein, we report the synthesis and antiplasmodium evaluation of a new series of amidated analogues and demonstrate that these compounds potently inhibit Plasmodium phosphatidylinositol-4-kinase (PI4K) type IIIß while moderately inhibiting cyclic guanidine monophosphate (cGMP)-dependent protein kinase (PKG) activity in vitro. Using in silico docking, we predict key binding interactions for these analogues within the adenosine triphosphate (ATP)-binding site of PI4K and PKG, paving the way for structure-based optimization of imidazopyridazines targeting both Plasmodium PI4K and PKG. While several derivatives showed low nanomolar antiplasmodium activity (IC50 < 100 nM), some compounds, including piperazine analogue 28, resulted in strong dual PI4K and PKG inhibition. The compounds also demonstrated transmission-blocking potential, evident from their potent inhibition of early- and late-stage gametocytes. Finally, the current compounds generally showed improved aqueous solubility and reduced hERG (human ether-a-go-go-related gene) channel inhibition.


Subject(s)
1-Phosphatidylinositol 4-Kinase , Plasmodium , Cyclic GMP-Dependent Protein Kinases , Guanidine , Phosphatidylinositols , Plasmodium falciparum , Protein Kinases
9.
J Med Chem ; 63(21): 13013-13030, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33103428

ABSTRACT

A series of 2,4-disubstituted imidazopyridines, originating from a SoftFocus Kinase library, was identified from a high throughput phenotypic screen against the human malaria parasite Plasmodium falciparum. Hit compounds showed moderate asexual blood stage activity. During lead optimization, several issues were flagged such as cross-resistance against the multidrug-resistant K1 strain, in vitro cytotoxicity, and cardiotoxicity and were addressed through structure-activity and structure-property relationship studies. Pharmacokinetic properties were assessed in mice for compounds showing desirable in vitro activity, a selectivity window over cytotoxicity, and microsomal metabolic stability. Frontrunner compound 37 showed good exposure in mice combined with good in vitro activity against the malaria parasite, which translated into in vivo efficacy in the P. falciparum NOD-scid IL-2Rγnull (NSG) mouse model. Preliminary mechanistic studies suggest inhibition of hemozoin formation as a contributing mode of action.


Subject(s)
Antimalarials/chemistry , Hemeproteins/antagonists & inhibitors , Imidazoles/chemistry , Plasmodium falciparum/physiology , Protozoan Proteins/antagonists & inhibitors , Pyridines/chemistry , Animals , Antimalarials/metabolism , Antimalarials/pharmacology , Antimalarials/therapeutic use , Disease Models, Animal , Half-Life , Hemeproteins/metabolism , Imidazoles/metabolism , Imidazoles/pharmacology , Imidazoles/therapeutic use , Life Cycle Stages/drug effects , Malaria/drug therapy , Malaria/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Microsomes, Liver/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Pyridines/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Structure-Activity Relationship
10.
ACS Infect Dis ; 6(7): 1951-1964, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32470286

ABSTRACT

Phenotypic whole-cell screening against Mycobacterium tuberculosis (Mtb) in glycerol-alanine-salts supplemented with Tween 80 and iron (GASTE-Fe) media led to the identification of a 2-aminoquinazolinone hit compound, sulfone 1 which was optimized for solubility by replacing the sulfone moiety with a sulfoxide 2. The synthesis and structure-activity relationship (SAR) studies identified several compounds with potent antimycobacterial activity, which were metabolically stable and noncytotoxic. Compound 2 displayed favorable in vitro properties and was therefore selected for in vivo pharmacokinetic (PK) studies where it was found to be extensively metabolized to the sulfone 1. Both derivatives exhibited promising PK parameters; however, when 2 was evaluated for in vivo efficacy in an acute TB infection mouse model, it was found to be inactive. In order to understand the in vitro and in vivo discrepancy, compound 2 was subsequently retested in vitro using different Mtb strains cultured in different media. This revealed that activity was only observed in media containing glycerol and led to the hypothesis that glycerol was not used as a primary carbon source by Mtb in the mouse lungs, as has previously been observed. Support for this hypothesis was provided by spontaneous-resistant mutant generation and whole genome sequencing studies, which revealed mutations mapping to glycerol metabolizing genes indicating that the 2-aminoquinazolinones kill Mtb in vitro via a glycerol-dependent mechanism of action.


Subject(s)
Antitubercular Agents , Mycobacterium tuberculosis , Animals , Antitubercular Agents/pharmacology , Drug Design , Mice , Structure-Activity Relationship
11.
ACS Omega ; 5(12): 6967-6982, 2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32258933

ABSTRACT

A phenotypic whole cell high-throughput screen against the asexual blood and liver stages of the malaria parasite identified a benzimidazole chemical series. Among the hits were the antiemetic benzimidazole drug Lerisetron 1 (IC50 NF54 = 0.81 µM) and its methyl-substituted analogue 2 (IC50 NF54 = 0.098 µM). A medicinal chemistry hit to lead effort led to the identification of chloro-substituted analogue 3 with high potency against the drug-sensitive NF54 (IC50 NF54 = 0.062 µM) and multidrug-resistant K1 (IC50 K1 = 0.054 µM) strains of the human malaria parasite Plasmodium falciparum. Compounds 2 and 3 gratifyingly showed in vivo efficacy in both Plasmodium berghei and P. falciparum mouse models of malaria. Cardiotoxicity risk as expressed in strong inhibition of the human ether-a-go-go-related gene (hERG) potassium channel was identified as a major liability to address. This led to the synthesis and biological assessment of around 60 analogues from which several compounds with improved antiplasmodial potency, relative to the lead compound 3, were identified.

12.
ACS Infect Dis ; 5(2): 303-315, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30525439

ABSTRACT

A drug repositioning approach was leveraged to derivatize astemizole (AST), an antihistamine drug whose antimalarial activity was previously identified in a high-throughput screen. The multistage activity potential against the Plasmodium parasite's life cycle of the subsequent analogues was examined by evaluating against the parasite asexual blood, liver, and sexual gametocytic stages. In addition, the previously reported contribution of heme detoxification to the compound's mode of action was interrogated. Ten of the 17 derivatives showed half-maximal inhibitory concentrations (IC50s) of <0.1 µM against the chloroquine (CQ)-sensitive Plasmodium falciparum NF54 ( PfNF54) strain while maintaining submicromolar potency against the multidrug-resistant strain, PfK1, with most showing low likelihood of cross-resistance with CQ. Selected analogues ( PfNF54-IC50 < 0.1 µM) were tested for cytotoxicity on Chinese hamster ovarian (CHO) cells and found to be highly selective (selectivity index > 100). Screening of AST and its analogues against gametocytes revealed their moderate activity (IC50: 1-5 µM) against late stage P. falciparum gametocytes, while the evaluation of activity against P. berghei liver stages identified one compound (3) with 3-fold greater activity than the parent AST compound. Mechanistic studies showed a strong correlation between in vitro inhibition of ß-hematin formation by the AST derivatives and their antiplasmodium IC50s. Analyses of intracellular inhibition of hemozoin formation within the parasite further yielded signatures attributable to a possible perturbation of the heme detoxification machinery.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Astemizole/analogs & derivatives , Hemeproteins/antagonists & inhibitors , Plasmodium falciparum/drug effects , Animals , CHO Cells , Chloroquine/pharmacology , Cricetulus , Drug Repositioning , Drug Resistance, Multiple , Inhibitory Concentration 50 , Life Cycle Stages
13.
Medchemcomm ; 9(10): 1733-1745, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30429978

ABSTRACT

3,6-Diarylated imidazopyridazines have recently been shown to possess good in vitro antiplasmodial and in vivo antimalarial activity. However, frontrunner compounds have been associated with poor solubility and a hERG (human ether-a-go-go-related gene) inhibition liability raising concerns for potential cardiotoxicity risks. Herein, we report the synthesis and structure-activity relationship studies of new imidazopyridazines aimed at improving aqueous solubility and countering hERG inhibition while maintaining antiplasmodial potency. While we identified new analogues with potent antiplasmodial activity (IC50 = 0.031 µM against the NF54 drug-sensitive strain, and IC50 = 0.0246 µM against the K1 multidrug resistant strain), hERG inhibition remained an issue. Excitingly, on the other hand, new analogues with a substantially improved hERG inhibition profile (IC50 = 7.83-32.3 µM) with sub-micromolar antiplasmodial activity (NF54, IC50 = 0.151-0.922 µM) were identified. Similarly, the introduced molecular features also resulted in analogues with moderate to high solubility (60-200 µM) while also displaying sub-micromolar antiplasmodial potency (NF54, IC50 = 0.136-0.99 µM).

15.
J Med Chem ; 61(20): 9371-9385, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30256636

ABSTRACT

A lead-optimization program around a 2,6-imidazopyridine scaffold was initiated based on the two early lead compounds, 1 and 2, that were shown to be efficacious in an in vivo humanized Plasmodium falciparum NODscidIL2Rγnull mouse malaria infection model. The observation of atypical dose-response curves when some compounds were tested against multidrug resistant malaria parasite strains guided the optimization process to define a chemical space that led to typical sigmoidal dose-response and complete kill of multidrug resistant parasites. After a structure and property analysis identified such a chemical space, compounds were prepared that displayed suitable activity, ADME, and safety profiles with respect to cytotoxicity and hERG inhibition.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Drug Resistance, Multiple/drug effects , Imidazoles/chemistry , Imidazoles/pharmacology , Plasmodium falciparum/drug effects , Pyridines/chemistry , Pyridines/pharmacology , Absorption, Physicochemical , Animals , Antimalarials/metabolism , Antimalarials/pharmacokinetics , Dose-Response Relationship, Drug , Imidazoles/metabolism , Imidazoles/pharmacokinetics , Mice , Pyridines/metabolism , Pyridines/pharmacokinetics , Structure-Activity Relationship , Tissue Distribution
16.
Article in English | MEDLINE | ID: mdl-30249687

ABSTRACT

The in vivo antimalarial efficacies of two phosphatidylinositol 4-kinase (PI4K) inhibitors, a 3,5-diaryl-2-aminopyrazine sulfoxide and its corresponding sulfone metabolite, were evaluated in the NOD-scid IL2Rγnull (NSG) murine malaria disease model of Plasmodium falciparum infection. We hypothesized that the sulfoxide would serve as a more soluble prodrug for the sulfone, which would lead to improved drug exposure with oral dosing. Both compounds had similar efficacy (90% effective dose [ED90], 0.1 mg kg-1 of body weight) across a quadruple-dose regimen. Pharmacokinetic profiling revealed rapid sulfoxide clearance via conversion to sulfone, with sulfone identified as the major active metabolite. When the sulfoxide was dosed, the exposure of the sulfone achieved was as much as 2.9-fold higher than when the sulfone was directly dosed, thereby demonstrating that the sulfoxide served as an effective prodrug for the treatment of malaria.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Parasitemia/drug therapy , Prodrugs/pharmacology , Pyrazines/pharmacology , Sulfones/pharmacology , Sulfoxides/pharmacology , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/genetics , 1-Phosphatidylinositol 4-Kinase/metabolism , Animals , Antimalarials/blood , Antimalarials/chemical synthesis , Antimalarials/pharmacokinetics , Biotransformation , Disease Models, Animal , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/parasitology , Gene Expression , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Parasitemia/pathology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Pyrazines/blood , Pyrazines/chemical synthesis , Pyrazines/pharmacokinetics , Sulfones/blood , Sulfones/chemical synthesis , Sulfones/pharmacokinetics , Sulfoxides/blood , Sulfoxides/chemical synthesis , Sulfoxides/pharmacokinetics , Treatment Outcome
17.
J Am Chem Soc ; 140(36): 11424-11437, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30107725

ABSTRACT

The Plasmodium proteasome is an emerging antimalarial target due to its essential role in all the major life cycle stages of the parasite and its contribution to the establishment of resistance to artemisinin (ART)-based therapies. However, because of a similarly essential role for the host proteasome, the key property of any antiproteasome therapeutic is selectivity. Several parasite-specific proteasome inhibitors have recently been reported, however, their selectivity must be improved to enable clinical development. Here we describe screening of diverse libraries of non-natural synthetic fluorogenic substrates to identify determinants at multiple positions on the substrate that produce enhanced selectivity. We find that selection of an optimal electrophilic "warhead" is essential to enable high selectivity that is driven by the peptide binding elements on the inhibitor. We also find that host cell toxicity is dictated by the extent of coinhibition of the human ß2 and ß5 subunits. Using this information, we identify compounds with over 3 orders of magnitude selectivity for the parasite enzyme. Optimization of the pharmacological properties resulted in molecules that retained high potency and selectivity, were soluble, sufficiently metabolically stable and orally bioavailable. These molecules are highly synergistic with ART and can clear parasites in a mouse model of infection, making them promising leads as antimalarial drugs.


Subject(s)
Artemisinins/pharmacology , Plasmodium falciparum/enzymology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Artemisinins/chemistry , Dose-Response Relationship, Drug , Molecular Structure , Proteasome Inhibitors/chemistry , Structure-Activity Relationship
18.
Article in English | MEDLINE | ID: mdl-29941635

ABSTRACT

The 2-aminopyridine MMV048 was the first drug candidate inhibiting Plasmodium phosphatidylinositol 4-kinase (PI4K), a novel drug target for malaria, to enter clinical development. In an effort to identify the next generation of PI4K inhibitors, the series was optimized to improve properties such as solubility and antiplasmodial potency across the parasite life cycle, leading to the 2-aminopyrazine UCT943. The compound displayed higher asexual blood stage, transmission-blocking, and liver stage activities than MMV048 and was more potent against resistant Plasmodium falciparum and Plasmodium vivax clinical isolates. Excellent in vitro antiplasmodial activity translated into high efficacy in Plasmodium berghei and humanized P. falciparum NOD-scid IL-2Rγ null mouse models. The high passive permeability and high aqueous solubility of UCT943, combined with low to moderate in vivo intrinsic clearance, resulted in sustained exposure and high bioavailability in preclinical species. In addition, the predicted human dose for a curative single administration using monkey and dog pharmacokinetics was low, ranging from 50 to 80 mg. As a next-generation Plasmodium PI4K inhibitor, UCT943, based on the combined preclinical data, has the potential to form part of a single-exposure radical cure and prophylaxis (SERCaP) to treat, prevent, and block the transmission of malaria.

19.
J Med Chem ; 61(13): 5692-5703, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29889526

ABSTRACT

A novel 2,8-disubstituted-1,5-naphthyridine hit compound stemming from the open access Medicines for Malaria Venture Pathogen Box formed a basis for a hit-to-lead medicinal chemistry program. Structure-activity relationship investigations resulted in compounds with potent antiplasmodial activity against both chloroquine sensitive (NF54) and multidrug resistant (K1) strains of the human malaria parasite Plasmodium falciparum. In the humanized P. falciparum mouse efficacy model, one of the frontrunner compounds showed in vivo efficacy at an oral dose of 4 × 50 mg·kg-1. In vitro mode-of-action studies revealed Plasmodium falciparum phosphatidylinositol-4-kinase as the target.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Malaria/drug therapy , Naphthyridines/chemistry , Naphthyridines/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , 1-Phosphatidylinositol 4-Kinase/chemistry , Animals , Antimalarials/chemistry , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Antimalarials/therapeutic use , Disease Models, Animal , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Mice , Models, Molecular , Naphthyridines/pharmacokinetics , Naphthyridines/therapeutic use , Plasmodium falciparum/physiology , Protein Conformation , Structure-Activity Relationship , Tissue Distribution
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...