Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Med Chem ; 19(5): 478-484, 2023.
Article in English | MEDLINE | ID: mdl-35993460

ABSTRACT

OBJECTIVE: A series of novel, substituted tetracyclic benzothiazepines were designed and prepared in an effort to optimize the potency of this chemical class against drug-resistant strains of the malaria parasite. METHODS: Tetracyclic benzothiazepines bearing structural modification at seven distinct positions within the structure were synthesized in Knoevenagel condensation reactions followed by sequential intermolecular thio-Michael and then intramolecular imine formation reactions. Following purification and chemical characterization, the novel compounds were tested for in vitro efficacy against blood-stage P. falciparum and liver-stage P. berghei and also for in vivo efficacy against P. berghei. RESULTS: Benzothiazepines bearing structural modification at the sulfur atom and at the three carbocycles within the molecule were successfully synthesized. The majority of analogs inhibited bloodstage P. falciparum with submicromolar IC50 values. The potency of an 8-methoxy-substituted analog 12 exceeded that of chloroquine in all three P. falciparum strains tested. The parent benzothiazepine 1 possessed liver-stage activity, inhibiting P. berghei sporozoites infecting HepG2 cells with an IC50 of 106.4 nM and an IC90 of 408.9 nM, but failed to enhance the longevity of P. berghei infected mice compared to the controls. Compounds displayed modest toxicity toward HepG2 cells and were tolerated by mice at the highest dose tested, 640 mg/kg/dose once daily for three days. CONCLUSION: The tetracyclic benzothiazepine described, which inhibits P. berghei infected hepatic cells with an IC50 of 106.4 nM, would appear to warrant further investigation. Optimization of ADME properties may be required since the most active analogs are probably excessively lipophilic.


Subject(s)
Antimalarials , Malaria , Animals , Mice , Plasmodium falciparum , Antimalarials/pharmacology , Malaria/drug therapy , Plasmodium berghei , Liver
2.
Am J Trop Med Hyg ; 88(6): 1138-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23530079

ABSTRACT

Anti-malarial 8-aminoquinolines drugs cause acute hemolytic anemia in individuals with glucose-6-phosphate dehydrogenase deficiency (G6PDD). Efforts to develop non-hemolytic 8-aminoquinolines have been severely limited caused by the lack of a predictive in vivo animal model of hemolytic potential that would allow screening of candidate compounds. This report describes a G6PDD mouse model with a phenotype closely resembling the G6PDD phenotype found in the African A-type G6PDD human. These G6PDD mice, given different doses of primaquine, which used as a reference hemolytic drug, display a full array of hemolytic anemia parameters, consistently and reproducibly. The hemolytic and therapeutic indexes were generated for evaluation of hemotoxicity of drugs. This model demonstrated a complete hemolytic toxicity response to another known hemolytic antimalarial drug, pamaquine, but no response to non-hemolytic drugs, chloroquine and mefloquine. These results suggest that this model is suitable for evaluation of selected 8-AQ type candidate antimalarial drugs for their hemolytic potential.


Subject(s)
Aminoquinolines/adverse effects , Anemia, Hemolytic/physiopathology , Antimalarials/adverse effects , Acute Disease , Aminoquinolines/administration & dosage , Anemia, Hemolytic/etiology , Animals , Antimalarials/administration & dosage , Chloroquine/administration & dosage , Chloroquine/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Genotype , Glucosephosphate Dehydrogenase Deficiency/genetics , Glucosephosphate Dehydrogenase Deficiency/metabolism , Glutathione/blood , Haptoglobins/analysis , Hemolytic Agents/administration & dosage , Hemolytic Agents/adverse effects , Male , Mefloquine/administration & dosage , Mefloquine/adverse effects , Mice , Phenotype , Primaquine/administration & dosage , Primaquine/adverse effects , Reticulocyte Count
3.
PLoS One ; 7(7): e40416, 2012.
Article in English | MEDLINE | ID: mdl-22792308

ABSTRACT

Mechanisms of drug resistance in Plasmodium vivax have been difficult to study partially because of the difficulties in culturing the parasite in vitro. This hampers monitoring drug resistance and research to develop or evaluate new drugs. There is an urgent need for a novel method to study mechanisms of P. vivax drug resistance. In this paper we report the development and application of the first Plasmodium falciparum expression system to stably express P. vivax dhfr-ts alleles. We used the piggyBac transposition system for the rapid integration of wild-type, single mutant (117N) and quadruple mutant (57L/58R/61M/117T) pvdhfr-ts alleles into the P. falciparum genome. The majority (81%) of the integrations occurred in non-coding regions of the genome; however, the levels of pvdhfr transcription driven by the P. falciparum dhfr promoter were not different between integrants of non-coding and coding regions. The integrated quadruple pvdhfr mutant allele was much less susceptible to antifolates than the wild-type and single mutant pvdhfr alleles. The resistance phenotype was stable without drug pressure. All the integrated clones were susceptible to the novel antifolate JPC-2067. Therefore, the piggyBac expression system provides a novel and important tool to investigate drug resistance mechanisms and gene functions in P. vivax.


Subject(s)
Plasmodium falciparum/genetics , Plasmodium vivax/enzymology , Protozoan Proteins/genetics , Tetrahydrofolate Dehydrogenase/genetics , Thymidylate Synthase/genetics , Amino Acid Substitution , Antimalarials/pharmacology , Cells, Cultured , Drug Resistance , Erythrocytes/parasitology , Folic Acid Antagonists/pharmacology , Gene Dosage , Humans , Inhibitory Concentration 50 , Mutagenesis, Insertional , Plasmodium falciparum/drug effects , Plasmodium vivax/genetics , Proguanil/pharmacology , Protozoan Proteins/biosynthesis , Pyrimethamine/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Tetrahydrofolate Dehydrogenase/biosynthesis , Thymidylate Synthase/biosynthesis , Transfection , Triazines/pharmacology
4.
J Med Chem ; 54(19): 6634-46, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21848332

ABSTRACT

A series of new guanidylimidazole derivatives was prepared and evaluated in mice and Rhesus monkeys infected with malarial sporozoites. The majority of the new compounds showed poor metabolic stability and weak in vitro activities in three clones of Plasmodium falciparum. Compounds 8a, 8h, 9a, 16a, and 16e cured the mice infected with sporozoites of P. berghei at 160 and 320 mg/kg/day × 3 po. Compounds 8a showed better causal prophylactic activity than primaquine, tafenoquine, and Malarone in the Rhesus test. In the radical curative test, 8a cured one monkey and delayed relapse of another for 74 days at 30 mg/kg/day × 7 by im. By oral dosing, 8a delayed relapse 81 days for one and 32 days for other vs 11-12 days for control monkeys treated with 10 mg/kg of chloroquine by po alone. Compound 8h, which showed superior activity to 8a in mouse test, delayed the relapse of treated monkeys for 21-26 days at 30 mg/kg/day × 7 by oral.


Subject(s)
Antimalarials/chemical synthesis , Guanidines/chemical synthesis , Imidazoles/chemical synthesis , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Crystallography, X-Ray , Guanidines/chemistry , Guanidines/pharmacology , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Imidazolines/chemical synthesis , Imidazolines/chemistry , Imidazolines/pharmacology , In Vitro Techniques , Macaca mulatta , Malaria/drug therapy , Mice , Microsomes, Liver/metabolism , Plasmodium berghei , Plasmodium falciparum/drug effects , Recurrence , Structure-Activity Relationship
5.
J Med Chem ; 54(13): 4523-35, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21627120

ABSTRACT

A series of 2-guanidino-4-oxoimidazoline (deoxo-IZ) derivatives was prepared and showed potent antimalarial activities in rodent and Rhesus models. Compound 8e, the most potent analogues of this series, is the first non-8-aminoqinoline antimalarial that demonstrated radical curative activity in non-human primate by oral route and showed causal prophylactic activity comparable to that of the commonly used clinical drugs in Rhesus monkeys infected with sporozoites of Plasmodium cynomolgi. The metabolic stability and metabolites profile indicated that the new deoxo-IZ derivatives (8) may act as prodrugs of the corresponding IZ (1 and 2) derivatives.


Subject(s)
Antimalarials/chemical synthesis , Guanidines/chemical synthesis , Imidazolidines/chemical synthesis , Imidazolines/chemical synthesis , Prodrugs/chemical synthesis , Administration, Oral , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Guanidines/chemistry , Guanidines/pharmacology , Imidazolidines/chemistry , Imidazolidines/pharmacology , Imidazolines/chemistry , Imidazolines/pharmacology , Macaca mulatta , Malaria/drug therapy , Malaria/prevention & control , Mice , Plasmodium berghei , Plasmodium cynomolgi , Plasmodium falciparum/drug effects , Prodrugs/chemistry , Prodrugs/pharmacology , Structure-Activity Relationship
6.
Mol Biochem Parasitol ; 177(1): 12-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21251932

ABSTRACT

Atovaquone is a hydroxy-naphthoquinone that is used to treat parasitic and fungal infections including Plasmodium falciparum (malaria), Pneumocystis jivorecii (pneumonia) and Toxoplasma gondii (toxoplasmosis). It blocks mitochondrial oxidation of ubiquinol in these organisms by binding to the ubiquinol oxidation site of the cytochrome bc(1) complex. Failure of atovaquone treatment has been linked to the appearance of mutations in the mitochondrially encoded gene for cytochrome b. In order to determine the optimal parameters required for inhibition of respiration in parasites and pathogenic fungi and overcome drug resistance, we have synthesized and tested the inhibitory activity of novel hydroxy-naphthoquinones against blood stage P. falciparum and liver stage P. berghei and against cytochrome bc(1) complexes isolated from yeast strains bearing mutations in cytochrome b associated with resistance in Plasmodium, Pneumocystis, and Toxoplasma. One of the new inhibitors is highly effective against an atovaquone resistant Plasmodium and illustrates the type of modification to the hydroxy-naphthoquinone ring of atovaquone that might mitigate drug resistance.


Subject(s)
Antifungal Agents/pharmacology , Antiprotozoal Agents/pharmacology , Drug Design , Drug Resistance , Naphthoquinones/pharmacology , Antifungal Agents/chemistry , Antiprotozoal Agents/chemistry , Cell Line , Humans , Malaria/parasitology , Molecular Structure , Naphthoquinones/chemistry , Plasmodium/drug effects , Plasmodium/genetics , Plasmodium/growth & development , Plasmodium/metabolism , Structure-Activity Relationship , Yeasts/drug effects , Yeasts/genetics , Yeasts/metabolism
7.
Antimicrob Agents Chemother ; 54(9): 3927-32, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20566761

ABSTRACT

Plasmodium vivax resistance to antifolates is prevalent throughout Australasia and is caused by point mutations within the parasite dihydrofolate reductase (DHFR)-thymidylate synthase. Several unique mutations have been reported in P. vivax DHFR, and their roles in resistance to classic and novel antifolates are not entirely clear due, in part, to the inability to culture P. vivax in vitro. In this study, we use a homologous system to episomally express both wild-type and various mutant P. vivax dhfr (pvdhfr) alleles in an antifolate-sensitive line of P. falciparum and to assess their influences on the susceptibility of the recipient P. falciparum line to commonly used and new antifolate drugs. Although the wild-type pvdhfr-transfected P. falciparum line was as susceptible to antifolate drugs as the P. falciparum parent line, the single (117N), double (57L/117T and 58R/117T), and quadruple (57L/58R/61M/117T) mutant pvdhfr alleles conferred a marked reduction in their susceptibilities to antifolates. The resistance index increased with the number of mutations in these alleles, indicating that these mutations contribute to antifolate resistance directly. In contrast, the triple mutant allele (58R/61M/117T) significantly reversed the resistance to all antifolates, indicating that 61M may be a compensatory mutation. These findings help elucidate the mechanism of antifolate resistance and the effect of existing mutations in the parasite population on the current and new generation of antifolate drugs. It also demonstrates that the episomal transfection system has the potential to provide a rapid screening system for drug development and for studying drug resistance mechanisms in P. vivax.


Subject(s)
Multienzyme Complexes/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium vivax/enzymology , Plasmodium vivax/genetics , Tetrahydrofolate Dehydrogenase/genetics , Thymidylate Synthase/genetics , Animals , Folic Acid Antagonists/pharmacology , Multienzyme Complexes/physiology , Mutation , Parasitic Sensitivity Tests , Plasmodium falciparum/genetics , Plasmodium vivax/drug effects , Tetrahydrofolate Dehydrogenase/physiology , Thymidylate Synthase/physiology , Transfection
8.
Mol Biochem Parasitol ; 172(1): 9-18, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20332005

ABSTRACT

Cyclin-dependent kinases (CDKs) have an established role in metazoans and yeast in DNA replication, transcription and cell cycle regulation. Several CDKs and their effectors have been identified in the malaria parasite Plasmodium falciparum and their biological functions are beginning to be investigated. Here we report results from the functional characterization of Pfmrk and its effector PfMAT1. We validated the interactions between Pfmrk and PfMAT1 and pinpointed their intracellular location. Co-immunoprecipitation studies demonstrated physical interaction between the two proteins and identified the C-terminal domain of PfMAT1 as the Pfmrk activator domain. Immunofluorescence analyses using GFP and RFP-tagged versions of Pfmrk and PfMAT1, respectively, demonstrated the co-localization of these two proteins to the parasite nucleus. Bacterial two-hybrid screen of a P. falciparum cDNA library using Pfmrk as the bait identified two plasmodial DNA replication proteins, PfRFC-5 and PfMCM6, as interactors with Pfmrk. We demonstrate that that these two proteins are substrates of Pfmrk-mediated phosphorylation and that PfMAT1 confers substrate specificity to the Pfmrk kinase complex. Collectively, these data suggest a role for Pfmrk in the nucleus of the parasite presumably in regulation of the DNA replication machinery.


Subject(s)
Cyclin-Dependent Kinases/metabolism , DNA Replication , Plasmodium falciparum/physiology , Protein Interaction Mapping , Protein Kinases/metabolism , Protozoan Proteins/metabolism , Cell Nucleus/chemistry , Immunoprecipitation , Phosphorylation , Plasmodium falciparum/chemistry , Protein Binding , Protein Structure, Tertiary , Substrate Specificity , Two-Hybrid System Techniques
9.
Malar J ; 7: 72, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-18442404

ABSTRACT

BACKGROUND: In order to maximize the useful therapeutic life of antimalarial drugs, it is crucial to understand the mechanisms by which parasites resistant to antimalarial drugs are selected and spread in natural populations. Recent work has demonstrated that pyrimethamine-resistance conferring mutations in Plasmodium falciparum dihydrofolate reductase (dhfr) have arisen rarely de novo, but spread widely in Asia and Africa. The origin and spread of mutations in Plasmodium vivax dhfr were assessed by constructing haplotypes based on sequencing dhfr and its flanking regions. METHODS: The P. vivax dhfr coding region, 792 bp upstream and 683 bp downstream were amplified and sequenced from 137 contemporary patient isolates from Colombia, India, Indonesia, Papua New Guinea, Sri Lanka, Thailand, and Vanuatu. A repeat motif located 2.6 kb upstream of dhfr was also sequenced from 75 of 137 patient isolates, and mutational relationships among the haplotypes were visualized using the programme Network. RESULTS: Synonymous and non-synonymous single nucleotide polymorphisms (SNPs) within the dhfr coding region were identified, as was the well-documented in-frame insertion/deletion (indel). SNPs were also identified upstream and downstream of dhfr, with an indel and a highly polymorphic repeat region identified upstream of dhfr. The regions flanking dhfr were highly variable. The double mutant (58R/117N) dhfr allele has evolved from several origins, because the 58R is encoded by at least 3 different codons. The triple (58R/61M/117T) and quadruple (57L/61M/117T/173F, 57I/58R/61M/117T and 57L/58R/61M/117T) mutant alleles had at least three independent origins in Thailand, Indonesia, and Papua New Guinea/Vanuatu. CONCLUSION: It was found that the P. vivax dhfr coding region and its flanking intergenic regions are highly polymorphic and that mutations in P. vivax dhfr that confer antifolate resistance have arisen several times in the Asian region. This contrasts sharply with the selective sweep of rare antifolate resistant alleles observed in the P. falciparum populations in Asia and Africa. The finding of multiple origins of resistance-conferring mutations has important implications for drug policy.


Subject(s)
Antimalarials/pharmacology , Drug Resistance/genetics , Plasmodium vivax/drug effects , Pyrimethamine/pharmacology , Tetrahydrofolate Dehydrogenase/genetics , Animals , Codon , DNA, Protozoan/genetics , DNA, Protozoan/isolation & purification , Malaria, Vivax/drug therapy , Malaria, Vivax/parasitology , Mutation , Plasmodium vivax/genetics , Polymorphism, Single Nucleotide , Pyrimethamine/therapeutic use
10.
J Antimicrob Chemother ; 60(4): 811-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17646199

ABSTRACT

OBJECTIVES: To assess the antimalarial pharmacodynamics and pharmacokinetics of the novel dihydrofolate reductase (DHFR) inhibitor, JPC2056 and its principal active metabolite JPC2067 in cynomolgus monkeys using an in vivo-in vitro model. METHODS: In a two-phase crossover design, five cynomolgus monkeys were administered a single dose (20 mg/kg) and multiple doses (20 mg/kg daily for 3 days) of JPC2056. Plasma samples collected from treated monkeys were assessed for in vitro antimalarial activity against Plasmodium falciparum lines having wild-type (D6), double-mutant (K1) and quadruple-mutant (TM90-C2A) DHFR-thymidylate synthase (TS) and a P. falciparum line transformed with a Plasmodium vivax dhfr-ts quadruple-mutant allele (D6-PvDHFR). Plasma JPC2056 and JPC2067 concentrations were measured by LC-mass spectrometry. RESULTS: The mean inhibitory dilution (ID(90)) of monkey plasma at 3 h after drug administration against D6, K1 and TM90-C2A was, respectively, 1253, 585 and 869 after the single-dose regimen and 1613, 1120 and 1396 following the multiple-dose regimen. Less activity was observed with the same monkey plasma samples against the D6-PvDHFR line, with a mean ID(90) of 53 after multiple dosing. Geometric mean plasma concentrations of JPC2056 and JPC2067 at 3 h after drug administration were, respectively, 113 and 12 ng/mL after the single dose and 150 and 17 ng/mL after multiple dosing. The mean elimination half-life of JPC2056 was shorter than its metabolite after both regimens (single dose, 7.3 versus 11.8 h; multiple doses, 6.6 versus 11.1 h). CONCLUSIONS: The high potency of JPC2056 against P. falciparum DHFR-TS quadruple-mutant lines provides optimism for the future development of JPC2056 for the treatment of malaria infections.


Subject(s)
Antimalarials/pharmacology , Antimalarials/pharmacokinetics , Animals , Antimalarials/administration & dosage , Antimalarials/blood , Attention , Chromatography, High Pressure Liquid , Drug Resistance, Microbial/genetics , Half-Life , Macaca fascicularis , Male , Mass Spectrometry , Parasitic Sensitivity Tests , Plasma/chemistry , Plasmodium falciparum/drug effects , Plasmodium vivax/drug effects
11.
J Infect Dis ; 196(3): 467-74, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17597462

ABSTRACT

With the emergence of drug-resistant vivax malaria, in vitro studies are urgently needed to examine resistance mechanisms and for drug development. Currently, Plasmodium vivax culturing is inadequate for addressing these needs; therefore, surrogate biological systems have been developed. Although these systems are informative, they do not address Plasmodium species-specific mechanisms, such as drug delivery through erythrocytes and parasite membranes. Here, we demonstrate that P. falciparum is an excellent biological system for expression of P. vivax dhfr-ts alleles to assess dihydrofolate reductase (DHFR)-thymidylate synthase interactions with antifolates. Our results show that the P. vivax dhfr-ts quadruple-mutant allele AMRU1, expressed in P. falciparum, provides significant protection against pyrimethamine, cycloguanil, and clocicguanil. Moreover, the PvDHFR quadruple mutant confers greater resistance to cycloguanil, clociguanil, and WR99210 than the PfDHFR quadruple mutant. Modeling of both P. vivax and P. falciparum DHFR quadruple mutants suggests that mutations unique to P. vivax DHFR are responsible for differences seen in parasite susceptibility to antifolates.


Subject(s)
Antimalarials/pharmacology , Drug Resistance , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Plasmodium vivax/enzymology , Tetrahydrofolate Dehydrogenase/genetics , Thymidylate Synthase/genetics , Animals , Animals, Genetically Modified , Binding Sites , Gene Expression Regulation , Mutation , Protein Conformation , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/metabolism
12.
Am J Trop Med Hyg ; 75(4): 617-21, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17038682

ABSTRACT

The increasing use of sulfadoxine-pyrimethamine (SP) for the treatment of chloroquine-resistant Plasmodium falciparum has resulted in increased reports of SP resistance of P. falciparum worldwide. Selection of SP-resistant Plasmodium vivax in areas where P. falciparum and P. vivax co-exist is not entirely clear. We examined the prevalence and extent of point mutations in pvdhfr and pvdhps in 70 P. vivax isolates from China, East Timor, Papua New Guinea (PNG), Philippines, Vanuatu, and Vietnam. Mutations in seven codon positions were found in pvdhfr, with the majority of isolates having double mutations (S58R/S117N). The greatest range of mutations was observed in the PNG and Vanuatu isolates, ranging from single to quadruple mutations (F57L/S58R/T61M/S117T). Single mutations in pvdhps were observed only in parasites with mutations in corresponding pvdhfr. Parasites with the S58R/S117N dhfr allelic type showed an MIC level for pyrimethamine and cycloguanil comparable to that previously reported, but were susceptible to WR99210.


Subject(s)
Antimalarials/pharmacology , Dihydropteroate Synthase/genetics , Folic Acid Antagonists/pharmacology , Plasmodium vivax/genetics , Point Mutation , Tetrahydrofolate Dehydrogenase/genetics , Amino Acid Sequence/genetics , Animals , Asia, Southeastern , China , Dihydropteroate Synthase/chemistry , Drug Combinations , Drug Resistance/genetics , Humans , Melanesia , Molecular Sequence Data , Plasmodium vivax/drug effects , Plasmodium vivax/enzymology , Proguanil , Pyrimethamine/pharmacology , Pyrimethamine/therapeutic use , Sulfadoxine/pharmacology , Sulfadoxine/therapeutic use , Tetrahydrofolate Dehydrogenase/chemistry , Triazines/pharmacology
13.
J Med Chem ; 48(8): 2805-13, 2005 Apr 21.
Article in English | MEDLINE | ID: mdl-15828818

ABSTRACT

Phenoxypropoxybiguanides, such as 1 (PS-15), are prodrugs analogous to the relationship of proguanil and its active metabolite cycloguanil. Unlike cycloguanil, however, 1a (WR99210), the active metabolite of 1, has retained in vitro potency against newly emerging antifolate-resistant malaria parasites. Unfortunately, manufacturing processes and gastrointestinal intolerance have prevented the clinical development of 1. In vitro antimalarial activity and in vitro metabolism studies have been performed on newly synthesized phenoxypropoxybiguanide analogues. All of the active dihydrotriazine metabolites exhibited potent antimalarial activity with in vitro IC(50) values less than 0.04 ng/mL. In vitro metabolism studies in human liver microsomes identified the production of not only the active dihydrotriazine metabolite, but also a desalkylation on the carbonyl chain, and multiple hydroxylated metabolites. The V(max) for production of the active metabolites ranged from 10.8 to 27.7 pmol/min/mg protein with the K(m) ranging from 44.8 to 221 microM. The results of these studies will be used to guide the selection of a lead candidate.


Subject(s)
Antimalarials/pharmacokinetics , Biguanides/pharmacokinetics , Microsomes, Liver/metabolism , Prodrugs/pharmacokinetics , Triazines/metabolism , Animals , Antimalarials/chemistry , Antimalarials/metabolism , Biguanides/chemistry , Biguanides/metabolism , Chromatography, Liquid , Drug Resistance , Folic Acid Antagonists/pharmacology , Humans , In Vitro Techniques , Mass Spectrometry , Plasmodium falciparum/drug effects , Prodrugs/chemistry , Prodrugs/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...