Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Molecules ; 27(19)2022 Oct 05.
Article in English | MEDLINE | ID: mdl-36235141

ABSTRACT

Three unique 5,6-seco-hexahydrodibenzopyrans (seco-HHDBP) machaeridiols A−C, reported previously from Machaerium Pers., have displayed potent activities against methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus faecium, and E. faecalis (VRE). In order to enrich the pipeline of natural product-derived antimicrobial compounds, a series of novel machaeridiol-based analogs (1−17) were prepared by coupling stemofuran, pinosylvin, and resveratrol legends with monoterpene units R-(−)-α-phellandrene, (−)-p-mentha-2,8-diene-1-ol, and geraniol, and their inhibitory activities were profiled against MRSA ATCC 1708, VRE ATCC 700221, and cancer signaling pathways. Compounds 5 and 11 showed strong in vitro activities with MIC values of 2.5 µg/mL and 1.25 µg/mL against MRSA, respectively, and 2.50 µg/mL against VRE, while geranyl analog 14 was found to be moderately active (MIC 5 µg/mL). The reduction of the double bonds of the monoterpene unit of compound 5 resulted in 17, which had the same antibacterial potency (MIC 1.25 µg/mL and 2.50 µg/mL) as its parent, 5. Furthermore, a combination study between seco-HHDBP 17 and HHDBP machaeriol C displayed a synergistic effect with a fractional inhibitory concentrations (FIC) value of 0.5 against MRSA, showing a four-fold decrease in the MIC values of both 17 and machaeriol C, while no such effect was observed between vancomycin and 17. Compounds 11 and 17 were further tested in vivo against nosocomial MRSA at a single intranasal dose of 30 mg/kg in a murine model, and both compounds were not efficacious under these conditions. Finally, compounds 1−17 were profiled against a panel of luciferase genes that assessed the activity of complex cancer-related signaling pathways (i.e., transcription factors) using T98G glioblastoma multiforme cells. Among the compounds tested, the geranyl-substituted analog 14 exhibited strong inhibition against several signaling pathways, notably Smad, Myc, and Notch, with IC50 values of 2.17 µM, 1.86 µM, and 2.15 µM, respectively. In contrast, the anti-MRSA actives 5 and 17 were found to be inactive (IC50 > 20 µM) across the panel of these cancer-signaling pathways.


Subject(s)
Anti-Infective Agents , Biological Products , Methicillin-Resistant Staphylococcus aureus , Neoplasms , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Biological Products/pharmacology , Luciferases , Mice , Microbial Sensitivity Tests , Monoterpenes/pharmacology , Resveratrol/pharmacology , Signal Transduction , Transcription Factors , Vancomycin/pharmacology
2.
3.
Molecules ; 26(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374444

ABSTRACT

The metabolic pathways in the apicoplast organelle of Plasmodium parasites are similar to those in plastids in plant cells and are suitable targets for malaria drug discovery. Some phytotoxins released by plant pathogenic fungi have been known to target metabolic pathways of the plastid; thus, they may also serve as potential antimalarial drug leads. An EtOAc extract of the broth of the endophyte Botryosphaeria dothidea isolated from a seed collected from a Torreya taxifolia plant with disease symptoms, showed in vitro antimalarial and phytotoxic activities. Bioactivity-guided fractionation of the extract afforded a mixture of two known isomeric phytotoxins, FRT-A and flavipucine (or their enantiomers, sapinopyridione and (-)-flavipucine), and two new unstable γ-lactam alkaloids dothilactaenes A and B. The isomeric mixture of phytotoxins displayed strong phytotoxicity against both a dicot and a monocot and moderate cytotoxicity against a panel of cell lines. Dothilactaene A showed no activity. Dothilactaene B was isolated from the active fraction, which showed moderate in vitro antiplasmodial activity with high selectivity index. In spite of this activity, its instability and various other biological activities shown by related compounds would preclude it from being a viable antimalarial lead.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Ascomycota/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Toxins, Biological/chemistry , Toxins, Biological/pharmacology , Antimalarials/isolation & purification , Molecular Structure , Plant Extracts/isolation & purification , Plasmodium/drug effects , Seeds/chemistry , Spectrum Analysis , Taxaceae/microbiology , Toxins, Biological/isolation & purification
4.
Malar J ; 18(1): 30, 2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30700282

ABSTRACT

BACKGROUND: The activity and haemolytic toxicity associated with primaquine has been linked to its reactive metabolites. The reactive metabolites are thought to be primarily formed through the action of cytochrome P450-mediated pathways. Human erythrocytes generally are not considered a significant contributor to drug biotransformation. As erythrocytes are the target of primaquine toxicity, the ability of erythrocytes to mediate the formation of reactive oxidative primaquine metabolites in the absence of hepatic enzymes, was evaluated. METHODS: Primaquine and its enantiomers were incubated separately with human red blood cells and haemoglobin. Post-incubation analysis was performed with UPLC-MS/MS to identify products of biotransformation. RESULTS: The major metabolite detected was identified as primaquine-5,6-orthoquinone, reflecting the pathway yielding putative active and haematotoxic metabolites of primaquine, which was formed by oxidative demethylation of 5-hydroxyprimaquine. Incubation of primaquine with haemoglobin in a cell-free system yielded similar results. It appears that the observed biotransformation is due to non-enzymatic processes, perhaps due to reactive oxygen species (ROS) present in erythrocytes or in the haemoglobin incubates. CONCLUSION: This study presents new evidence that primaquine-5,6-orthoquinone, the metabolite of primaquine reflecting the oxidative biotransformation pathway, is generated in erythrocytes, probably by non-enzymatic means, and may not require transport from the liver or other tissues.


Subject(s)
Antimalarials/metabolism , Erythrocytes/metabolism , Primaquine/metabolism , Quinones/metabolism , Reactive Oxygen Species/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Humans , In Vitro Techniques , Tandem Mass Spectrometry
5.
Molecules ; 24(4)2019 Feb 21.
Article in English | MEDLINE | ID: mdl-30795572

ABSTRACT

Bioassay-guided fractionation of an EtOAc extract of the broth of the endophytic fungus Nemania sp. UM10M (Xylariaceae) isolated from a diseased Torreya taxifolia leaf afforded three known cytochalasins, 19,20-epoxycytochalasins C (1) and D (2), and 18-deoxy-19,20-epoxy-cytochalasin C (3). All three compounds showed potent in vitro antiplasmodial activity and phytotoxicity with no cytotoxicity to Vero cells. These compounds exhibited moderate to weak cytotoxicity to some of the cell lines of a panel of solid tumor (SK-MEL, KB, BT-549, and SK-OV-3) and kidney epithelial cells (LLC-PK11). Evaluation of in vivo antimalarial activity of 19,20-epoxycytochalasin C (1) in a mouse model at 100 mg/kg dose showed that this compound had weak suppressive antiplasmodial activity and was toxic to animals.


Subject(s)
Antineoplastic Agents/pharmacology , Antiprotozoal Agents/pharmacology , Cytochalasins/pharmacology , Malaria/drug therapy , Taxaceae/microbiology , Xylariales/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/isolation & purification , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , Cytochalasins/chemistry , Cytochalasins/isolation & purification , Endophytes/chemistry , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Malaria/mortality , Malaria/parasitology , Male , Mice , Plant Leaves/microbiology , Plasmodium berghei/drug effects , Plasmodium berghei/growth & development , Survival Analysis , Vero Cells
6.
New Phytol ; 218(2): 616-629, 2018 04.
Article in English | MEDLINE | ID: mdl-29461628

ABSTRACT

Sorgoleone, a major component of the hydrophobic root exudates of Sorghum spp., is probably responsible for many of the allelopathic properties attributed to members of this genus. Much of the biosynthetic pathway for this compound has been elucidated, with the exception of the enzyme responsible for the catalysis of the addition of two hydroxyl groups to the resorcinol ring. A library prepared from isolated Sorghum bicolor root hair cells was first mined for P450-like sequences, which were then analyzed by quantitative reverse transcription-polymerase chain reaction (RT-qPCR) to identify those preferentially expressed in root hairs. Full-length open reading frames for each candidate were generated, and then analyzed biochemically using both a yeast expression system and transient expression in Nicotiana benthamiana leaves. RNA interference (RNAi)-mediated repression in transgenic S. bicolor was used to confirm the roles of these candidates in the biosynthesis of sorgoleone in planta. A P450 enzyme, designated CYP71AM1, was found to be capable of catalyzing the formation of dihydrosorgoleone using 5-pentadecatrienyl resorcinol-3-methyl ether as substrate, as determined by gas chromatography-mass spectroscopy (GC-MS). RNAi-mediated repression of CYP71AM1 in S. bicolor resulted in decreased sorgoleone contents in multiple independent transformant events. Our results strongly suggest that CYP71AM1 participates in the biosynthetic pathway of the allelochemical sorgoleone.


Subject(s)
Biosynthetic Pathways , Cytochrome P-450 Enzyme System/metabolism , Lipids/biosynthesis , Pheromones/biosynthesis , Plant Proteins/metabolism , Plant Roots/cytology , Sorghum/enzymology , Amino Acid Sequence , Benzoquinones , Cytochrome P-450 Enzyme System/chemistry , Gene Expression Regulation, Plant , Molecular Docking Simulation , Phylogeny , Plant Proteins/chemistry , RNA Interference , Saccharomyces cerevisiae/metabolism , Substrate Specificity , Nicotiana
7.
Chem Res Toxicol ; 29(7): 1132-41, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27223244

ABSTRACT

The administration of primaquine (PQ), an essential drug for the treatment and radical cure of malaria, can lead to methemoglobin formation and life-threatening hemolysis for glucose-6-phosphate dehydrogenase deficient patients. The ionization potential (IP, a quantitative measure of the ability to lose an electron) of the metabolites generated by antimalarial 8-aminoquinoline (8-AQ) drugs like PQ has been believed to be correlated in part to this methemoglobinemia hemotoxicity: the lower the IP of an 8-AQ derivative, the higher the concentration of methemoglobin generated. In this work, demethoxylated primaquine (AQ02) was employed as a model, by intensive computation at the B3LYP-SCRF(PCM)/6-311++G**//B3LYP/6-31G** level in water, to study the effects of hydroxylation at various positions on the ionization potential. Compared to the parent AQ02, the IPs of AQ02's metabolites hydroxylated at N1', C5, and C7 were lower by 61, 30, and 19 kJ/mol, respectively, while differences in the IP relative to PQ were small for hydroxylation at all other positions. The C6 position, at which the IP of the hydroxylated metabolite was greater than that of AQ02, by 2 kJ/mol, was found to be unique. Several literature and proposed 8-AQ analogues were studied to evaluate substituent effects on their potential to generate methemoglobin, with the finding that hydroxylations at N1' and C5 contribute the most to the potential hemotoxicity of PQ-based antimalarials, whereas hydroxylation at C7 has little effect. Phenoxylation at C5 in PQ-based 8-AQs can block the hydroxylation at C5 and reduce the potential for methemoglobin generation, while -CF3 and chlorines attached to the phenolic ring can further reduce the risk. The H-shift at N1' during the cationization of hydroxylated metabolites of 8-AQs sharply decreased their IPs, but this effect can be significantly reduced by the introduction of an electron-withdrawing group to the quinoline core. The results and this approach may be utilized for the design of safer antimalarial 8-AQ analogues.


Subject(s)
Aminoquinolines/toxicity , Antimalarials/toxicity , Methemoglobinemia/chemically induced , Electrochemistry , Hydroxylation
8.
Malar J ; 15: 466, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27618912

ABSTRACT

BACKGROUND: There has been some evidence to suggest that the addition of chloroquine (CQ) or quinine (QN) to 8-aminoquinoline (8-AQ) treatment regimens may increase the therapeutic efficacy of the 8-AQ and simultaneously mitigate against its haemolytic toxicity. However, both CQ and QN are considered effective, although perhaps moderate inhibitors of CYP2D6, an enzyme now regarded as necessary for primaquine (PQ) pharmacologic activity. An understanding of the influence of CQ and QN on the metabolism of PQ may shed light on the potential mechanisms of the beneficial interaction. METHODS: Differential metabolism of PQ enantiomers by recombinant human CYP2D6, monoamine oxidase A (MAO), and cryopreserved human hepatocytes in the presence/absence of CQ and QN. RESULTS: Both CQ and QN significantly inhibited the activity of CYP2D6. PQ depletion by MAO and human hepatocytes was not affected significantly by the presence of CQ and QN. CYP2D6-mediated hydroxylation was largely suppressed by both CQ and QN. The formation of the primary deaminated metabolites, including carboxyprimaquine (CPQ) and cyclized side chain derivative from the aldehyde (m/z 241), was not sensitive to the presence of CQ and QN. However, the appearance of the glucuronides of CPQ and PQ alcohol were significantly suppressed. CQ and QN also inhibited the appearance of the m/z 257 metabolite with a similar pattern, suggesting that it may be derived from the CPQ conjugate. The apparent quinone-imine of CPQ (m/z 289) was only partially suppressed by both QN and CQ, but with a differential pattern of inhibition for the two drugs. The m/z 274 (quinone-imine of a ring-hydroxylated PQ metabolite) and m/z 422 (an apparent glucose conjugate of PQ) metabolites in hepatocytes were strongly suppressed by both QN and CQ, perhaps a reflection of the 2D6 inhibition by these drugs. The formation of the carbamoyl glucuronide of PQ (m/z 480) was not affected by CQ/QN. CONCLUSION: The metabolite-specific interactions in the current studies seem at variance with earlier reports of the dependence of PQ on CYP2D6 metabolism, and enhanced PQ anti-malarial activity/reduced toxicity in the presence of CQ/QN. These results suggest a complex picture in which CQ/QN may shift metabolite pathway balances towards a profile that retains efficacy, while reducing the formation or availability of toxic metabolites to erythrocytes. Alternatively, these drugs may alter transport or distribution of PQ metabolites in a fashion that reduces toxicity while maintaining efficacy against the parasite.


Subject(s)
Antimalarials/metabolism , Antimalarials/pharmacology , Chloroquine/metabolism , Chloroquine/pharmacology , Drug Interactions , Primaquine/metabolism , Primaquine/pharmacology , Cytochrome P-450 CYP2D6/metabolism , Hepatocytes/metabolism , Humans , Metabolic Networks and Pathways , Monoamine Oxidase/metabolism , Primaquine/pharmacokinetics
9.
Malar J ; 15: 224, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27093859

ABSTRACT

BACKGROUND: The clinical utility of primaquine (PQ), used as a racemic mixture of two enantiomers, is limited due to metabolism-linked hemolytic toxicity in individuals with genetic deficiency in glucose-6-phosphate dehydrogenase. The current study investigated differential metabolism of PQ enantiomers in light of the suggestions that toxicity and efficacy might be largely enantioselective. METHODS: Stable isotope (13)C-labelled primaquine and its two enantiomers (+)-PQ, (-)-PQ were separately incubated with cryopreserved human hepatocytes. Time-tracked substrate depletion and metabolite production were monitored via UHPLC-MS/MS. RESULTS: The initial half-life of 217 and 65 min; elimination rate constants (λ) of 0.19 and 0.64 h(-1); intrinsic clearance (Clint) of 2.55 and 8.49 (µL/min)/million cells, which when up-scaled yielded Clint of 6.49 and 21.6 (mL/min)/kg body mass was obtained respectively for (+)- and (-)-PQ. The extrapolation of in vitro intrinsic clearance to in vivo human hepatic blood clearance, performed using the well-stirred liver model, showed that the rate of hepatic clearance of (+)-PQ was only 45 % that of (-)-PQ. Two major primary routes of metabolism were observed-oxidative deamination of the terminal amine and hydroxylations on the quinoline moiety of PQ. The major deaminated metabolite, carboxyprimaquine (CPQ) was preferentially generated from the (-)-PQ. Other deaminated metabolites including PQ terminal alcohol (m/z 261), a cyclized side chain derivative from the aldehyde (m/z 241), cyclized carboxylic acid derivative (m/z 257), a quinone-imine product of hydroxylated CPQ (m/z 289), CPQ glucuronide (m/z 451) and the glucuronide of PQ alcohol (m/z 437) were all preferentially generated from the (-)-PQ. The major quinoline oxidation product (m/z 274) was preferentially generated from (+)-PQ. In addition to the products of the two metabolic pathways, two other major metabolites were observed: a prominent glycosylated conjugate of PQ on the terminal amine (m/z 422), peaking by 30 min and preferentially generated by (+)-PQ; and the carbamoyl glucuronide of PQ (m/z 480) exclusively generated from (+)-PQ. CONCLUSION: Metabolism of PQ showed enantioselectivity. These findings may provide important information in establishing clinical differences in PQ enantiomers.


Subject(s)
Hepatocytes/metabolism , Primaquine/analogs & derivatives , Primaquine/metabolism , Carbon Isotopes/analysis , Chromatography, High Pressure Liquid , Half-Life , Humans , Kinetics , Stereoisomerism , Tandem Mass Spectrometry
10.
J Nat Prod ; 79(9): 2341-9, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27618204

ABSTRACT

Two new flavonoids, rac-6-formyl-5,7-dihydroxyflavanone (1) and 2',6'-dihydroxy-4'-methoxy-3'-methylchalcone (2), together with five known derivatives, rac-8-formyl-5,7-dihydroxyflavanone (3), 4',6'-dihydroxy-2'-methoxy-3'-methyldihydrochalcone (4), rac-7-hydroxy-5-methoxy-6-methylflavanone (5), 3'-formyl-2',4',6'-trihydroxy-5'-methyldihydrochalcone (6), and 3'-formyl-2',4',6'-trihydroxydihydrochalcone (7), were isolated from the leaves of Eugenia rigida. The individual (S)- and (R)-enantiomers of 1 and 3, together with the corresponding formylated flavones 8 (6-formyl-5,7-dihydroxyflavone) and 9 (8-formyl-5,7-dihydroxyflavone), as well as 2',4',6'-trihydroxychalcone (10), 3'-formyl-2',4',6'-trihydroxychalcone (11), and the corresponding 3'-formyl-2',4',6'-trihydroxydihydrochalcone (7) and 2',4',6'-trihydroxydihydrochalcone (12), were synthesized. The structures of the isolated and synthetic compounds were established via NMR, HRESIMS, and electronic circular dichroism data. In addition, the structures of 3, 5, and 8 were confirmed by single-crystal X-ray diffraction crystallography. The isolated and synthetic flavonoids were evaluated for their antimicrobial and cytotoxic activities against a panel of microorganisms and solid tumor cell lines.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Chalcones/isolation & purification , Chalcones/pharmacology , Eugenia/chemistry , Flavonoids/isolation & purification , Flavonoids/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Candida albicans/drug effects , Chalcones/chemistry , Cryptococcus neoformans/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Escherichia coli/drug effects , Flavanones , Flavonoids/chemistry , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Molecular Conformation , Molecular Structure , Mycobacterium avium Complex/drug effects , Nuclear Magnetic Resonance, Biomolecular , Plant Leaves/chemistry , Pseudomonas aeruginosa/drug effects , Puerto Rico , Staphylococcus aureus/drug effects
11.
Antimicrob Agents Chemother ; 59(7): 3864-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25870069

ABSTRACT

Cytochrome P450 (CYP) 2D metabolism is required for the liver-stage antimalarial efficacy of the 8-aminoquinoline molecule tafenoquine in mice. This could be problematic for Plasmodium vivax radical cure, as the human CYP 2D ortholog (2D6) is highly polymorphic. Diminished CYP 2D6 enzyme activity, as in the poor-metabolizer phenotype, could compromise radical curative efficacy in humans. Despite the importance of CYP 2D metabolism for tafenoquine liver-stage efficacy, the exact role that CYP 2D metabolism plays in the metabolism and pharmacokinetics of tafenoquine and other 8-aminoquinoline molecules has not been extensively studied. In this study, a series of tafenoquine pharmacokinetic experiments were conducted in mice with different CYP 2D metabolism statuses, including wild-type (WT) (reflecting extensive metabolizers for CYP 2D6 substrates) and CYPmouse 2D knockout (KO) (reflecting poor metabolizers for CYP 2D6 substrates) mice. Plasma and liver pharmacokinetic profiles from a single 20-mg/kg of body weight dose of tafenoquine differed between the strains; however, the differences were less striking than previous results obtained for primaquine in the same model. Additionally, the presence of a 5,6-ortho-quinone tafenoquine metabolite was examined in both mouse strains. The 5,6-ortho-quinone species of tafenoquine was observed, and concentrations of the metabolite were highest in the WT extensive-metabolizer phenotype. Altogether, this study indicates that CYP 2D metabolism in mice affects tafenoquine pharmacokinetics and could have implications for human tafenoquine pharmacokinetics in polymorphic CYP 2D6 human populations.


Subject(s)
Aminoquinolines/pharmacokinetics , Antimalarials/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Aminoquinolines/blood , Animals , Antimalarials/blood , Area Under Curve , Biotransformation , Cytochrome P-450 CYP2D6/metabolism , Half-Life , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Primaquine/pharmacokinetics
12.
Drug Metab Dispos ; 43(4): 571-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637634

ABSTRACT

Primaquine (PQ), a racemic drug, is the only treatment available for radical cure of relapsing Plasmodium vivax malaria and blocking transmission of P. falciparum malaria. Recent studies have shown differential pharmacologic and toxicologic profiles of individual PQ enantiomers in rodent, dog, and primate animal models. This study was conducted in six healthy adult human volunteers to determine the plasma pharmacokinetic profile of enantiomers of PQ and carboxyprimaquine (cPQ), the major plasma metabolite. The individuals were orally administered PQ diphosphate, equivalent to 45-mg base, 30 minutes after a normal breakfast. Blood samples were collected at different time intervals, and plasma samples were analyzed for enantiomers of PQ and cPQ. Plasma PQ concentrations were low and variable for both parent enantiomers and peaked around 2-4 hours. Peak (-)-(R)-PQ concentrations ranged from 121 ng/ml to 221 ng/ml, and peak (+)-(S)-PQ concentrations ranged from 168 ng/ml to 299 ng/ml. The cPQ concentrations were much higher and were surprisingly consistent from subject to subject. Essentially all the cPQ detected in plasma was (-)-cPQ. The peak concentrations of (-)-cPQ were observed at 8 hours (range: 1104-1756 ng/ml); however, very high concentrations were sustained through 24 hours. (+)-cPQ was two orders of magnitude lower than (-)-cPQ, and in a few subjects it was detected but only under the limit of quantification. In vitro studies with primary human hepatocytes also suggested more rapid metabolism of (-)-PQ compared with (+)-PQ. The results suggest more rapid metabolism of (-)-PQ to (-) cPQ compared with (+)-PQ. Alternatively, (+)-PQ or (+)-cPQ could be rapidly converted to another metabolite(s) or distributed to tissues. This is the first clinical report on enantioselective pharmacokinetic profiles of PQ and cPQ and supports further clinical evaluation of individual PQ enantiomers.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacokinetics , Hepatocytes/metabolism , Primaquine/analogs & derivatives , Administration, Oral , Adult , Antimalarials/blood , Cells, Cultured , Chromatography, High Pressure Liquid , Healthy Volunteers , Humans , Middle Aged , Molecular Structure , Primaquine/blood , Primaquine/chemistry , Primaquine/pharmacokinetics , Primary Cell Culture , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism , Structure-Activity Relationship
13.
Antimicrob Agents Chemother ; 58(12): 7283-91, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25267666

ABSTRACT

Primaquine (PQ) remains the sole available drug to prevent relapse of Plasmodium vivax malaria more than 60 years after licensure. While this drug was administered as a racemic mixture, prior studies suggested a pharmacodynamic advantage based on differential antirelapse activity and/or toxicities of its enantiomers. Oral primaquine enantiomers prepared using a novel, easily scalable method were given for 7 days to healthy rhesus macaques in a dose-rising fashion to evaluate their effects on the blood, liver, and kidneys. The enantiomers were then administered to Plasmodium cynomolgi-infected rhesus macaques at doses of 1.3 and 0.6 mg/kg of body weight/day in combination with chloroquine. The (-)-PQ enantiomer had higher clearance and apparent volume of distribution than did (+)-PQ and was more extensively converted to the carboxy metabolite. There is evidence for differential oxidative stress with a concentration-dependent rise in methemoglobin (MetHgb) with increasing doses of (+)-PQ greater than that seen for (-)-PQ. There was a marked, reversible hepatotoxicity in 2 of 3 animals dosed with (-)-PQ at 4.5 mg/kg. (-)-PQ in combination with chloroquine was successful in preventing P. cynomolgi disease relapse at doses of 0.6 and 1.3 mg/kg/day, while 1 of 2 animals receiving (+)-PQ at 0.6 mg/kg/day relapsed. While (-)-PQ was also associated with hepatotoxicity at higher doses as seen previously, this has not been identified as a clinical concern in humans during >60 years of use. Limited evidence for increased MetHgb generation with the (+) form in the rhesus macaque model suggests that it may be possible to improve the therapeutic window for hematologic toxicity in the clinic by separating primaquine into its enantiomers.


Subject(s)
Antimalarials/pharmacokinetics , Chloroquine/pharmacology , Malaria/drug therapy , Plasmodium cynomolgi/drug effects , Primaquine/pharmacokinetics , Animals , Antimalarials/blood , Antimalarials/chemistry , Antimalarials/pharmacology , Disease Models, Animal , Drug Administration Schedule , Drug Therapy, Combination , Humans , Kidney/drug effects , Liver/drug effects , Macaca mulatta , Malaria/blood , Malaria/parasitology , Malaria, Vivax , Male , Methemoglobin/metabolism , Oxidative Stress , Plasmodium cynomolgi/growth & development , Plasmodium vivax , Primaquine/blood , Primaquine/chemistry , Primaquine/pharmacology , Recurrence , Stereoisomerism
14.
Antimicrob Agents Chemother ; 58(8): 4737-44, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24913163

ABSTRACT

Hematotoxicity in individuals genetically deficient in glucose-6-phosphate dehydrogenase (G6PD) activity is the major limitation of primaquine (PQ), the only antimalarial drug in clinical use for treatment of relapsing Plasmodium vivax malaria. PQ is currently clinically used in its racemic form. A scalable procedure was developed to resolve racemic PQ, thus providing pure enantiomers for the first time for detailed preclinical evaluation and potentially for clinical use. These enantiomers were compared for antiparasitic activity using several mouse models and also for general and hematological toxicities in mice and dogs. (+)-(S)-PQ showed better suppressive and causal prophylactic activity than (-)-(R)-PQ in mice infected with Plasmodium berghei. Similarly, (+)-(S)-PQ was a more potent suppressive agent than (-)-(R)-PQ in a mouse model of Pneumocystis carinii pneumonia. However, at higher doses, (+)-(S)-PQ also showed more systemic toxicity for mice. In beagle dogs, (+)-(S)-PQ caused more methemoglobinemia and was toxic at 5 mg/kg of body weight/day given orally for 3 days, while (-)-(R)-PQ was well tolerated. In a novel mouse model of hemolytic anemia associated with human G6PD deficiency, it was also demonstrated that (-)-(R)-PQ was less hemolytic than (+)-(S)-PQ for the G6PD-deficient human red cells engrafted in the NOD-SCID mice. All these data suggest that while (+)-(S)-PQ shows greater potency in terms of antiparasitic efficacy in rodents, it is also more hematotoxic than (-)-(R)-PQ in mice and dogs. Activity and toxicity differences of PQ enantiomers in different species can be attributed to their different pharmacokinetic and metabolic profiles. Taken together, these studies suggest that (-)-(R)-PQ may have a better safety margin than the racemate in human.


Subject(s)
Antimalarials/pharmacokinetics , Hemolysis/drug effects , Malaria/drug therapy , Pneumonia, Pneumocystis/drug therapy , Primaquine/pharmacokinetics , Animals , Antimalarials/isolation & purification , Antimalarials/toxicity , Dogs , Erythrocyte Transfusion , Erythrocytes/drug effects , Erythrocytes/parasitology , Female , Glucosephosphate Dehydrogenase Deficiency/metabolism , Humans , Lethal Dose 50 , Malaria/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Inbred NOD , Mice, SCID , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Pneumocystis carinii/drug effects , Pneumocystis carinii/physiology , Pneumonia, Pneumocystis/microbiology , Primaquine/isolation & purification , Primaquine/toxicity , Stereoisomerism , Transplantation, Heterologous
15.
Malar J ; 13: 507, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25518709

ABSTRACT

BACKGROUND: Primaquine, currently the only approved drug for the treatment and radical cure of Plasmodium vivax malaria, is still used as a racemic mixture. Clinical use of primaquine has been limited due to haemolytic toxicity in individuals with genetic deficiency in glucose-6-phosphate dehydrogenase. Earlier studies have linked its therapeutic effects to CYP2D6-generated metabolites. The aim of the current study was to investigate the differential generation of the CYP2D6 metabolites by racemic primaquine and its individual enantiomers. METHODS: Stable isotope 13C-labelled primaquine and its two enantiomers were incubated with recombinant cytochrome-P450 supersomes containing CYP2D6 under optimized conditions. Metabolite identification and time-point quantitative analysis were performed using LC-MS/MS. UHPLC retention time, twin peaks with a mass difference of 6, MS-MS fragmentation pattern, and relative peak area with respect to parent compound were used for phenotyping and quantitative analysis of metabolites. RESULTS: The rate of metabolism of (+)-(S)-primaquine was significantly higher (50% depletion of 20 µM in 120 min) compared to (-)-(R)-primaquine (30% depletion) when incubated with CYP2D6. The estimated Vmax (µmol/min/mg) were 0.75, 0.98 and 0.42, with Km (µM) of 24.2, 33.1 and 21.6 for (±)-primaquine, (+)-primaquine and (-)-primaquine, respectively. Three stable mono-hydroxylated metabolites, namely, 2-, 3- and 4-hydroxyprimaquine (2-OH-PQ, 3-OH-PQ, and 4-OH-PQ), were identified and quantified. 2-OH-PQ was preferentially formed from (+)-primaquine in a ratio of 4:1 compared to (-)-primaquine. The racemic (±)-primaquine showed a pattern similar to the (-)-primaquine; 2-OH-PQ accounted for about 15-17% of total CYP2D6-mediated conversion of (+)-primaquine. In contrast, 4-OH-PQ was preferentially formed with (-)-primaquine (5:1), accounting for 22% of the total (-)-primaquine conversion. 3-OH-PQ was generated from both enantiomers and racemate. 5-hydroxyprimaquine was unstable. Its orthoquinone degradation product (twice as abundant in (+)-primaquine compared to (-)-primaquine) was identified and accounted for 18-20% of the CYP2D6-mediated conversion of (+)-primaquine. Other minor metabolites included dihydroxyprimaquine species, two quinone-imine products of dihydroxylated primaquine, and a primaquine terminal alcohol with variable generation from the individual enantiomers. CONCLUSION: The metabolism of primaquine by human CYP2D6 and the generation of its metabolites display enantio-selectivity regarding formation of hydroxylated product profiles. This may partly explain differential pharmacologic and toxicologic properties of primaquine enantiomers.


Subject(s)
Antimalarials/metabolism , Cytochrome P-450 CYP2D6/metabolism , Primaquine/metabolism , Antimalarials/chemistry , Chromatography, Liquid , Humans , Isotope Labeling , Kinetics , Plasmodium vivax , Primaquine/chemistry , Stereoisomerism , Substrate Specificity , Tandem Mass Spectrometry
16.
J Phys Chem A ; 118(29): 5501-7, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24956138

ABSTRACT

For antimalarial 8-aminoquinoline (8-AQ) drugs, the ionization potential (energy required to remove an electron) of their putative metabolites has been proposed to be correlated in part to their hemotoxicity potential. NPC1161 is a developmental candidate as an 8-AQ antimalarial drug. In this work, the ionization potentials (IPs) of the S-NPC1161 (NPC1161a) hydroxylated derivatives, which are possible metabolites derived from action of endogenous cytochrome P450 (CYP450) enzymes, were calculated at the B3LYP-SCRF(PCM)/6-311++G**//B3LYP/6-31G** level in water. The derivative hydroxylated at N1' (8-amino) was found to have the smallest IP of ∼ 430 kJ/mol, predicting that it would be the most hemotoxic. The calculated IPs of the derivatives hydroxylated at the C2 and C7 positions were ∼ 475 and ∼ 478 kJ/mol, respectively, whereas the calculated IPs of those hydroxylated at all other possible positions were between 480 and 490 kJ/mol. The homolytic bond dissociation energies (HBDEs) of all C-H/N-H bonds in NPC1161a were also calculated. The smaller HBDEs of the C-H/N-H bonds on the 8-amino side chain suggest that these positions are more easily hydroxylated compared to other sites. Molecular orbital analysis implies that the N1' position should be the most reactive center when NPC1161 approaches the heme in CYP450.


Subject(s)
Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Antimalarials/chemistry , Antimalarials/pharmacology , Heme/antagonists & inhibitors , Succinates/chemistry , Succinates/pharmacology , Aminoquinolines/metabolism , Antimalarials/metabolism , Heme/metabolism , Humans , Hydroxylation , Molecular Structure , Quantum Theory , Software , Stereoisomerism , Succinates/metabolism
17.
Plant Cell ; 22(3): 867-87, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20348430

ABSTRACT

Sorghum bicolor is considered to be an allelopathic crop species, producing phytotoxins such as the lipid benzoquinone sorgoleone, which likely accounts for many of the allelopathic properties of Sorghum spp. Current evidence suggests that sorgoleone biosynthesis occurs exclusively in root hair cells and involves the production of an alkylresorcinolic intermediate (5-[(Z,Z)-8',11',14'-pentadecatrienyl]resorcinol) derived from an unusual 16:3Delta(9,12,15) fatty acyl-CoA starter unit. This led to the suggestion of the involvement of one or more alkylresorcinol synthases (ARSs), type III polyketide synthases (PKSs) that produce 5-alkylresorcinols using medium to long-chain fatty acyl-CoA starter units via iterative condensations with malonyl-CoA. In an effort to characterize the enzymes responsible for the biosynthesis of the pentadecyl resorcinol intermediate, a previously described expressed sequence tag database prepared from isolated S. bicolor (genotype BTx623) root hairs was first mined for all PKS-like sequences. Quantitative real-time RT-PCR analyses revealed that three of these sequences were preferentially expressed in root hairs, two of which (designated ARS1 and ARS2) were found to encode ARS enzymes capable of accepting a variety of fatty acyl-CoA starter units in recombinant enzyme studies. Furthermore, RNA interference experiments directed against ARS1 and ARS2 resulted in the generation of multiple independent transformant events exhibiting dramatically reduced sorgoleone levels. Thus, both ARS1 and ARS2 are likely to participate in the biosynthesis of sorgoleone in planta. The sequences of ARS1 and ARS2 were also used to identify several rice (Oryza sativa) genes encoding ARSs, which are likely involved in the production of defense-related alkylresorcinols.


Subject(s)
Lipids/biosynthesis , Plant Proteins/metabolism , Plant Roots/enzymology , Polyketide Synthases/metabolism , Sorghum/genetics , Amino Acid Sequence , Benzoquinones , Escherichia coli/enzymology , Escherichia coli/genetics , Expressed Sequence Tags , Models, Molecular , Molecular Sequence Data , Phylogeny , Plant Proteins/genetics , Plant Roots/genetics , Polyketide Synthases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Resorcinols/metabolism , Sequence Alignment , Sorghum/enzymology
18.
Chem Res Toxicol ; 26(12): 1801-9, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24224488

ABSTRACT

Currently, the only clinically approved antimalarial drug to treat relapsing malaria is primaquine (PQ), yet PQ administration can cause life-threatening hemolytic anemia in some patients. In our efforts to understand the connection between PQ and methemoglobin formation, the effect of 5-substituted primaquine derivatives on the basicity of hemoglobin-bound O2 was investigated using various computational methods, including quantum mechanics/molecular mechanics (QM/MM) calculations, molecular dynamics simulations and density functional theory calculations, to determine the geometries, relative energies, spin densities, proton affinities and ionization potentials of various PQ derivatives and PQ···hemoglobin complexes. We found that the protein environment and solvent do not change our previously proposed methemoglobin formation mechanism that 5-hydroxyprimaquine donates an electron to O2, facilitating its conversion to H2O2 and generating methemoglobin. Because of 5-hydroxyprimaquine's ability to lose an electron by this mechanism, we then used different substituents at primaquine's 5-position and found that an electron-withdrawing group (EWG) increases the ionization potential of the corresponding derivative. As a result, the EWG-substituted derivatives make the hemoglobin-bound O2 less basic, because of their weaker electron-donating ability. These derivatives hence are predicted to have a lower propensity to generate methemoglobin, which can inform future design of less hemotoxic antimalarial drugs. We also carried out experimental measurement of methemoglobin formation for some of the 5-substituted derivatives.


Subject(s)
Aminoquinolines/chemistry , Methemoglobin/chemical synthesis , Primaquine/chemistry , Methemoglobin/chemistry , Molecular Dynamics Simulation , Molecular Structure , Quantum Theory
19.
J Chem Ecol ; 39(2): 262-70, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23314892

ABSTRACT

Leptospermone is a natural ß-triketone that specifically inhibits the enzyme p-hydrophyphenylpyruvate dioxygenase, the same molecular target site as that of the commercial herbicide mesotrione. The ß-triketone-rich essential oil of Leptospermum scoparium has both preemergence and postemergence herbicidal activity, resulting in bleaching of treated plants and dramatic growth reduction. Radiolabeled leptospermone was synthesized to investigate the in planta mechanism of action of this natural herbicide. Approximately 50 % of the absorbed leptospermone was translocated to the foliage suggesting rapid acropetal movement of the molecule. On the other hand, very little leptospermone was translocated away from the point of application on the foliage, indicating poor phloem mobility. These observations are consistent with the physico-chemical properties of leptospermone, such as its experimentally measured logP and pK a values, and molecular mass, number of hydrogen donors and acceptors, and number of rotatable bonds. Consequently, leptospermone is taken up readily by roots and translocated to reach its molecular target site. This provides additional evidence that the anecdotal observation of allelopathic suppression of plant growth under ß-triketone-producing species may be due to the release of these phytotoxins in soils.


Subject(s)
Digitaria/physiology , Herbicides/chemistry , Herbicides/metabolism , Leptospermum/chemistry , Oils, Volatile/chemistry , Oils, Volatile/metabolism , Biological Transport , Phloroglucinol/analogs & derivatives , Plant Leaves/physiology , Plant Roots/physiology
20.
J Labelled Comp Radiopharm ; 56(7): 341-3, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-24285435

ABSTRACT

In support of a program to identify toxic metabolites of the antimalarial, primaquine, its [(13)C6] analog was prepared from [(13)C6] anisole in seven steps.


Subject(s)
Antimalarials/chemical synthesis , Primaquine/chemical synthesis , Carbon Isotopes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL