Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799598

ABSTRACT

We sought to develop a cell-based cytotoxicity assay using human hepatocytes, which reflect the effects of drug-metabolizing enzymes on cytotoxicity. In this study, we generated luminescent human hepatoblastoma HepG2 cells using the mouse artificial chromosome vector, in which click beetle luciferase alone or luciferase and major drug-metabolizing enzymes (CYP2C9, CYP2C19, CYP2D6, and CYP3A4) are expressed, and monitored the time-dependent changes of CYP-mediated cytotoxicity expression by bioluminescence measurement. Real-time bioluminescence measurement revealed that compared with CYP-non-expressing cells, the luminescence intensity of CYP-expressing cells rapidly decreased when the cells were treated with low concentrations of aflatoxin B1 or primaquine, which exhibits cytotoxicity in the presence of CYP3A4 or CYP2D6, respectively. Using kinetics data obtained by the real-time bioluminescence measurement, we estimated the time-dependent changes of 50% inhibitory concentration (IC50) values in the aflatoxin B1- and primaquine-treated cell lines. The first IC50 value was detected much earlier and at a lower concentration in primaquine-treated CYP-expressing HepG2 cells than in primaquine-treated CYP-non-expressing cells, and the decrease of IC50 values was much faster in the former than the latter. Thus, we successfully monitored time- and concentration-dependent dynamic changes of CYP-mediated cytotoxicity expression in CYP-expressing luminescent HepG2 cells by means of real-time bioluminescence measurement.


Subject(s)
Aflatoxin B1/toxicity , Founder Effect , Luminescent Measurements/methods , Primaquine/toxicity , Time-Lapse Imaging/methods , Xenobiotics/toxicity , Animals , Cell Line, Tumor , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Gene Expression Regulation , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Luciferases/genetics , Luciferases/metabolism , Luminescence , Mice
2.
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
3.
Malar J ; 13: 148, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24742291

ABSTRACT

BACKGROUND: Primaquine, the only available drug effective against Plasmodium falciparum sexual stages, induces also a dose-dependent haemolysis, especially in glucose-6-phosphate dehydrogenase deficient (G6PDd) individuals. Therefore, it is important to determine the prevalence of this deficiency in areas that would potentially benefit from its use. The prevalence of G6PD deficiency by genotype and enzyme activity was determined in healthy school children in The Gambia. METHODS: Blood samples from primary school children collected during a dry season malaria survey were screened for G6PDd and malaria infection. Genotypes for allele mutations reported in the country; 376, 202A-, 968A- and 542 were analysed while enzyme activity (phenotype) was assayed using a semi-quantitative commercial test kit. Enzyme activity values were fitted in a finite mixture model to determine the distribution and calculate a cut-off for deficiency. The association between genotype and phenotype for boys and girls as well as the association between mutant genotype and deficient phenotype was analysed. RESULTS: Samples from 1,437 children; 51% boys were analysed. The prevalence of P. falciparum malaria infection was 14%. The prevalence of the 202A-, 968 and 542 mutations was 1.8%, 2.1% and 1.0%, respectively, and higher in boys than in girls. The prevalence of G6PDd phenotype was 6.4% (92/1,437), 7.8% (57/728) in boys and 4.9% (35/709) in girls with significantly higher odds in the former (OR 1.64, 95% CI 1.05, 2.53, p = 0.026). The deficient phenotype was associated with reduced odds of malaria infection (OR 0.77, 95% CI 0.36, 1.62, p = 0.49). CONCLUSIONS: There is a weak association between genotype and phenotype estimates of G6PDd prevalence. The phenotype expression of deficiency represents combinations of mutant alleles rather than specific mutations. Genotype studies in individuals with a deficient phenotype would help identify alleles responsible for haemolysis.


Subject(s)
Antimalarials/toxicity , Genotype , Glucosephosphate Dehydrogenase Deficiency/epidemiology , Glucosephosphate Dehydrogenase/genetics , Primaquine/toxicity , Adolescent , Alleles , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Gambia/epidemiology , Glucosephosphate Dehydrogenase/metabolism , Hemolysis , Humans , Malaria, Falciparum/epidemiology , Male , Plasmodium falciparum/isolation & purification , Polymerase Chain Reaction , Prevalence
4.
PLoS Med ; 9(11): e1001339, 2012.
Article in English | MEDLINE | ID: mdl-23152723

ABSTRACT

BACKGROUND: Primaquine is a key drug for malaria elimination. In addition to being the only drug active against the dormant relapsing forms of Plasmodium vivax, primaquine is the sole effective treatment of infectious P. falciparum gametocytes, and may interrupt transmission and help contain the spread of artemisinin resistance. However, primaquine can trigger haemolysis in patients with a deficiency in glucose-6-phosphate dehydrogenase (G6PDd). Poor information is available about the distribution of individuals at risk of primaquine-induced haemolysis. We present a continuous evidence-based prevalence map of G6PDd and estimates of affected populations, together with a national index of relative haemolytic risk. METHODS AND FINDINGS: Representative community surveys of phenotypic G6PDd prevalence were identified for 1,734 spatially unique sites. These surveys formed the evidence-base for a Bayesian geostatistical model adapted to the gene's X-linked inheritance, which predicted a G6PDd allele frequency map across malaria endemic countries (MECs) and generated population-weighted estimates of affected populations. Highest median prevalence (peaking at 32.5%) was predicted across sub-Saharan Africa and the Arabian Peninsula. Although G6PDd prevalence was generally lower across central and southeast Asia, rarely exceeding 20%, the majority of G6PDd individuals (67.5% median estimate) were from Asian countries. We estimated a G6PDd allele frequency of 8.0% (interquartile range: 7.4-8.8) across MECs, and 5.3% (4.4-6.7) within malaria-eliminating countries. The reliability of the map is contingent on the underlying data informing the model; population heterogeneity can only be represented by the available surveys, and important weaknesses exist in the map across data-sparse regions. Uncertainty metrics are used to quantify some aspects of these limitations in the map. Finally, we assembled a database of G6PDd variant occurrences to inform a national-level index of relative G6PDd haemolytic risk. Asian countries, where variants were most severe, had the highest relative risks from G6PDd. CONCLUSIONS: G6PDd is widespread and spatially heterogeneous across most MECs where primaquine would be valuable for malaria control and elimination. The maps and population estimates presented here reflect potential risk of primaquine-associated harm. In the absence of non-toxic alternatives to primaquine, these results represent additional evidence to help inform safe use of this valuable, yet dangerous, component of the malaria-elimination toolkit. Please see later in the article for the Editors' Summary.


Subject(s)
Antimalarials/toxicity , Gene Frequency , Geography, Medical/methods , Glucosephosphate Dehydrogenase Deficiency/epidemiology , Glucosephosphate Dehydrogenase/genetics , Hemolysis , Primaquine/toxicity , Bayes Theorem , Female , Geographic Mapping , Glucosephosphate Dehydrogenase/metabolism , Humans , Malaria/epidemiology , Male , Models, Genetic , Prevalence , Risk Assessment , Sex Distribution
5.
Chem Res Toxicol ; 24(9): 1476-85, 2011 Sep 19.
Article in English | MEDLINE | ID: mdl-21699254

ABSTRACT

The electron affinities (EA) of the 8-aminoquinoline antimalarial drug primaquine and several of its metabolites were studied using the density functional theory method. We first considered six substituents at the 5-position, -CH(3), -OH, -OCH(3), -Ph, -OPh, and -CHO. We found that in the gas phase the adiabatic EAs are similar to that of the parent primaquine for the -CH(3), -OH, and -OCH(3) substituents. In contrast, the -Ph, -OPh, and -CHO substituents all markedly increase the adiabatic EA. However, only the -CHO substituted compound is predicted to form a stable covalently bound radical anion in the gas phase due to its significant positive vertical EA relative to that of the parent primaquine. In addition, when the 8-position is substituted by the N-hydroxyl group or a quinone-imine structure is formed, the electron capture ability is significantly increased. In aqueous solution, all these molecules have significantly larger adiabatic EAs than in the gas phase. In addition, all of the vertical EAs are positive in aqueous solution. The implications of these findings for contributing to our mechanistic understanding of the red cell toxicity of 8-aminoquinoline compounds are further discussed.


Subject(s)
Aminoquinolines/chemistry , Antimalarials/chemistry , Free Radicals/chemistry , Primaquine/analogs & derivatives , Primaquine/chemistry , Aminoquinolines/toxicity , Anions/chemistry , Anions/toxicity , Antimalarials/toxicity , Electrons , Free Radicals/toxicity , Models, Molecular , Primaquine/toxicity , Quantum Theory
6.
Toxicol Appl Pharmacol ; 241(1): 14-22, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19616568

ABSTRACT

Primaquine, an 8-aminoquinoline, is the drug of choice for radical cure of relapsing malaria. Use of primaquine is limited due to its hemotoxicity, particularly in populations with glucose-6-phosphate dehydrogenase deficiency [G6PD(-)]. Biotransformation appears to be central to the anti-infective and hematological toxicities of primaquine, but the mechanisms are still not well understood. Metabolic studies with primaquine have been hampered due to the reactive nature of potential hemotoxic metabolites. An in vitro metabolism-linked hemotoxicity assay has been developed. Co-incubation of the drug with normal or G6PD(-) erythrocytes, microsomes or recombinant cytochrome P(450) (CYP) isoforms has allowed in situ generation of potential hemotoxic metabolite(s), which interact with the erythrocytes to generate hemotoxicity. Methemoglobin formation, real-time generation of reactive oxygen intermediates (ROIs) and depletion of reactive thiols were monitored as multiple biochemical end points for hemotoxicity. Primaquine alone did not produce any hemotoxicity, while a robust increase was observed in methemoglobin formation and generation of ROIs by primaquine in the presence of human or mouse liver microsomes. Multiple CYP isoforms (CYP2E1, CYP2B6, CYP1A2, CYP2D6 and CYP3A4) variably contributed to the hemotoxicity of primaquine. This was further confirmed by significant inhibition of primaquine hemotoxicity by the selective CYP inhibitors, namely thiotepa (CYP2B6), fluoxetine (CYP2D6) and troleandomycin (CYP3A4). Primaquine caused similar methemoglobin formation in G6PD(-) and normal human erythrocytes. However, G6PD(-) erythrocytes suffered higher oxidative stress and depletion of thiols than normal erythrocytes due to primaquine toxicity. The results provide significant insights regarding CYP isoforms contributing to hemotoxicity and may be useful in controlling toxicity of primaquine to increase its therapeutic utility.


Subject(s)
Antimalarials/toxicity , Cytochrome P-450 Enzyme System/metabolism , Erythrocytes/drug effects , Microsomes, Liver/drug effects , Primaquine/toxicity , Animals , Antimalarials/metabolism , Enzyme Inhibitors/pharmacology , Female , Fluoxetine/pharmacology , Glucosephosphate Dehydrogenase/metabolism , Humans , Isoenzymes/metabolism , Male , Methemoglobin/drug effects , Methemoglobin/metabolism , Mice , Mice, Inbred ICR , Microsomes, Liver/metabolism , Oxidative Stress/drug effects , Primaquine/metabolism , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Thiotepa/pharmacology , Troleandomycin/pharmacology
7.
J Spec Oper Med ; 19(3): 14-16, 2019.
Article in English | MEDLINE | ID: mdl-31539427

ABSTRACT

Presumptive antirelapse therapy (PART) with primaquine for Plasmodium vivax malaria postdeployment is an important component of the US military Force Health Protection plan. While primaquine is well tolerated in the majority of cases, we present a unique case of an active duty Army Ranger without glucose-6-phosphatase dehydrogenase or cytochrome b5 reductase (b5R) deficiencies who developed symptomatic methemoglobinemia while taking PART following a deployment to Afghanistan.


Subject(s)
Methemoglobinemia/chemically induced , Methemoglobinemia/diagnosis , Military Personnel , Primaquine/toxicity , Afghan Campaign 2001- , Delayed Diagnosis , Humans
8.
Mol Cell Biol ; 14(4): 2419-28, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7511206

ABSTRACT

Chloroquine (CQ)-resistant (CQR) Plasmodium falciparum malaria parasites show a strong decrease in CQ accumulation in comparison with chloroquine-sensitive parasites. Controversy exists over the role of the plasmodial pfmdr1 gene in the CQR phenotype. pfmdr1 is a member of the superfamily of ATP-binding cassette transporters. Other members of this family are the mammalian multidrug resistance genes and the CFTR gene. We have expressed the pfmdr1-encoded protein, Pgh1, in CHO cells and Xenopus oocytes. CHO cells expressing the Pgh1 protein demonstrated an increased, verapamil-insensitive susceptibility to CQ. Conversely, no increase in drug susceptibility to primaquine, quinine, adriamycin, or colchicine was observed in Pgh1-expressing cells. CQ uptake experiments revealed an increased, ATP-dependent accumulation of CQ in Pgh1-expressing cells over the level in nonexpressing control cells. The increased CQ accumulation in Pgh1-expressing cells coincided with an enhanced in vivo inhibition of lysosomal alpha-galactosidase by CQ. CHO cells expressing Pgh1 carrying two of the CQR-associated Pgh1 amino acid changes (S1034C and N1042D) did not display an increased CQ sensitivity. Immunofluorescence experiments revealed an intracellular localization of both mutant and wild-type forms of Pgh1. We conclude from our results that wild-type Pgh1 protein can mediate an increased intracellular accumulation of CQ and that this function is impaired in CQR-associated mutant forms of the protein. We speculate that the Pgh1 protein plays an important role in CQ import in CQ-sensitive malaria parasites.


Subject(s)
ATP-Binding Cassette Transporters , Chloroquine/toxicity , Drug Resistance/genetics , Gene Expression , Genes, Protozoan , Plasmodium falciparum/genetics , Protozoan Proteins/biosynthesis , Adenosine Triphosphate/metabolism , Animals , Base Sequence , Biological Transport , CHO Cells , Cell Survival/drug effects , Chloroquine/metabolism , Colchicine/toxicity , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator , DNA, Protozoan/genetics , DNA, Protozoan/metabolism , Doxorubicin/toxicity , Female , Membrane Proteins/genetics , Oligonucleotides, Antisense , Oocytes/metabolism , Plasmodium falciparum/drug effects , Primaquine/toxicity , Protozoan Proteins/metabolism , Quinine/toxicity , Restriction Mapping , Verapamil/pharmacology , Vinblastine/toxicity , Xenopus laevis
9.
Photochem Photobiol ; 83(6): 1415-27, 2007.
Article in English | MEDLINE | ID: mdl-18028216

ABSTRACT

This article describes the results of a coupled photophysical and photobiological study aimed at understanding the phototoxicity mechanism of the antimalarial drugs amodiaquine (AQ), primaquine (PQ) and chloroquine (CQ). Photophysical experiments were carried out in aqueous solutions by steady-state and time-resolved spectrometric techniques to obtain information on the different decay pathways of the excited states of the drugs and on the transient species formed upon laser irradiation. The results showed that all three drugs possess very low fluorescence quantum yields (10(-2)-10(-4)). Laser flash photolysis experiments proved the occurrence of photoionization processes leading to the formation of a radical cation in all three systems. In the case of AQ the lowest triplet state was also detected. Together with the photophysical properties the photobiological properties of the antimalarial drugs were investigated under UV irradiation, on various biological targets through a series of in vitro assays. Phototoxicity on mouse 3T3 fibroblast and human keratinocyte cell lines NCTC-2544 was detected for PQ and CQ but not for AQ. In particular, PQ- and CQ-induced apoptosis was revealed by the externalization of phosphatidylserine. Furthermore, upon UV irradiation, the drugs caused significant variations of the mitochondrial potential (Deltapsi(mt)) measured by flow cytometry. The photodamages produced by the drugs were also evaluated on proteins, lipids and DNA. The combined approaches were useful in understanding the mechanism of phototoxicity induced by these antimalarial drugs.


Subject(s)
Amodiaquine/chemistry , Chloroquine/chemistry , Primaquine/chemistry , Amodiaquine/toxicity , Animals , Antimalarials , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Chloroquine/toxicity , DNA/genetics , DNA Damage , Humans , Lipid Peroxidation/drug effects , Lipid Peroxidation/radiation effects , Lysosomes/drug effects , Lysosomes/radiation effects , Mice , Mitochondria/drug effects , Mitochondria/radiation effects , Molecular Structure , Photobiology , Photochemistry , Primaquine/toxicity , Reactive Oxygen Species/metabolism , Salmon , Spectrophotometry
10.
Toxicology ; 239(1-2): 96-107, 2007 Sep 24.
Article in English | MEDLINE | ID: mdl-17686563

ABSTRACT

Primaquine (PQ), a clinically important derivative of 8-aminoquinoline used against the hepatic stages (hypnozoites) of Plasmodium vivax and Plasmodium ovale, was studied to evaluate and compare between mRNA expression, and biochemical and histological parameters of hepatic stress in adult Swiss mice (Mus musculus). Following single oral dose of PQ (40 mg/kg, bw), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) along with hematoxylin and eosin stained liver sections did not show any signs of hepatic stress at 6, 12 and 24 h except for ALT activity at 6h. However, analysis at RNA transcript level revealed consistent and significant deregulation (p<0.01 and two-fold) of 16 probes corresponding to important cellular processes such as protein transportation, transcription regulation, intracellular signaling, protein synthesis, hematopoiesis, cell adhesion and cell proliferation. Pathway analysis identified large number of affected genes corresponding to 40 Gene Ontology terms having a z score greater than 2. These results indicate that PQ at high doses may affect gene expression in liver and may produce undesirable outcomes if consumed for longer durations.


Subject(s)
Antimalarials/toxicity , Gene Expression/drug effects , Liver/drug effects , Oligonucleotide Array Sequence Analysis/methods , Primaquine/toxicity , Toxicogenetics/methods , Administration, Oral , Alanine Transaminase/blood , Animals , Animals, Outbred Strains , Aspartate Aminotransferases/blood , Gene Expression Profiling , Liver/metabolism , Liver/pathology , Male , Mice , Oxidative Stress/drug effects , Oxidative Stress/genetics , RNA, Messenger/metabolism
11.
Chem Biol Interact ; 255: 63-73, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-26477383

ABSTRACT

The generation of reactive metabolites from therapeutic agents is one of the major mechanisms of drug-induced liver injury (DILI). In order to evaluate metabolism-related toxicity and improve drug efficacy and safety, we generated a battery of HepG2-derived cell lines that express 14 cytochrome P450s (CYPs) (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5 and 3A7) individually using a lentiviral expression system. The expression/production of a specific CYP in each cell line was confirmed by an increased abundance of the CYP at both mRNA and protein levels. Moreover, the enzymatic activities of representative CYPs in the corresponding cell lines were also measured. Using our CYP-expressed HepG2 cells, the toxicity of three drugs that could induce DILI (amiodarone, chlorpromazine and primaquine) was assessed, and all of them showed altered (increased or decreased) toxicity compared to the toxicity in drug-treated wild-type HepG2 cells. CYP-mediated drug toxicity examined in our cell system is consistent with previous reports, demonstrating the potential of these cells for assessing metabolism-related drug toxicity. This cell system provides a practical in vitro approach for drug metabolism screening and for early detection of drug toxicity. It is also a surrogate enzyme source for the enzymatic characterization of a particular CYP that contributes to drug-induced liver toxicity.


Subject(s)
Amiodarone/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 Enzyme System/metabolism , Liver/drug effects , Antimalarials/toxicity , Antipsychotic Agents/toxicity , Cell Line , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Chlorpromazine/toxicity , Cytochrome P-450 Enzyme Inhibitors/toxicity , Cytochrome P-450 Enzyme System/genetics , Drug Evaluation, Preclinical/methods , Gene Expression , Hep G2 Cells , Humans , Liver/metabolism , Liver/pathology , Primaquine/toxicity , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics
12.
J Clin Pharmacol ; 45(3): 346-51, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15703369

ABSTRACT

Hypersensitivity adverse drug reactions are much more common among patients with acquired immunodeficiency syndrome (AIDS) than in the general population. High rates of hypersensitivity reactions to clindamycin have been noted. To investigate the role of reactive metabolites in these reactions, the authors studied toxicity of clindamycin and sulphamethoxazole (SMX) and their metabolites in uninfected and human immunodeficiency virus (HIV)-infected MOLT3 cells. Infected and uninfected cells were incubated with clindamycin or sulphamethoxazole hydroxylamine in increasing concentrations; reactive metabolites were generated by coincubation of cells and drug with murine microsomes and a microsomal activating system. Over a concentration range of 0 to 400 microM SMX-HA, there was a significant concentration-dependent increase in cell death in HIV-infected compared to uninfected cells (28%+/-3% vs 8%+/-5% at 400 microM, P < .05). In contrast, coincubation of cells with clindamycin, microsomes, and a microsomal activating system, as well as combinations of primaquine or pyrimethamine, was not associated with an increase in cell death among infected compared to uninfected cells. No concentration-toxicity was demonstrated. These data support the role of reactive metabolites in adverse drug reactions to sulfonamides during HIV infection, whereas alternate mechanism(s) may be responsible for increased rates of adverse drug reactions to clindamycin among patients with AIDS.


Subject(s)
Anti-Infective Agents/toxicity , Cell Survival/drug effects , Clindamycin/toxicity , HIV/drug effects , Sulfamethoxazole/analogs & derivatives , T-Lymphocytes/drug effects , Animals , Biotransformation , Cell Line , Clindamycin/metabolism , Coculture Techniques , Drug Combinations , Drug Hypersensitivity/etiology , HIV/pathogenicity , Mice , Microsomes , Primaquine/toxicity , Pyrimethamine/toxicity , Sulfamethoxazole/metabolism , Sulfamethoxazole/toxicity , T-Lymphocytes/virology
13.
FEBS Lett ; 223(1): 77-81, 1987 Oct 19.
Article in English | MEDLINE | ID: mdl-3666145

ABSTRACT

Photooxidation of primaquine (1) and 5-hydroxyprimaquine (5) afforded a blue dye for which o-quinone structure 4 was elaborated. Similar oxidation of N-ethoxyacetylprimaquine (10) afforded o-quinone 11. Tissue schizontocidal activity of 4 and 11, and bisquinolylmethine 3 prepared earlier, showed that none of them had noteworthy antimalarial activity, but all three produced methemoglobin.


Subject(s)
Primaquine , Animals , Biotransformation , Cell Survival/drug effects , Hemolysis , Liver/drug effects , Methemoglobin , Oxidation-Reduction , Plasmodium/drug effects , Primaquine/analogs & derivatives , Primaquine/toxicity , Structure-Activity Relationship
14.
J Med Chem ; 29(9): 1765-9, 1986 Sep.
Article in English | MEDLINE | ID: mdl-3528491

ABSTRACT

The antimalarial effect of intravenously administered primaquine (PQ) can be improved and its toxicity diminished by linking it to a macromolecular carrier protein. A thiol-containing primaquine derivative 8-[[4-(2-amino-3-mercaptopropionamido)-1-methylbutyl]amino]-6- methoxyquinoline was synthesized. This compound could readily be linked via a disulfide bond to a carrier protein containing (pyridyldithio)propionate groups. The derivative was coupled to serum albumin as well as to serum albumin that contained covalently linked lactose residues. The protein-drug conjugates were tested for their antimalarial activity in mice inoculated with Plasmodium berghei. The causal prophylactic activity of the conjugate with the lactosaminated serum albumin was 2 times higher than that of the free drug; the mean causal prophylactic doses (CPD50) were 6 and 13 mg of primaquine base/kg, respectively. Moreover, its acute lethal toxicity had decreased at least 6.5-fold (mean lethal dose (LD50) greater than 85 mg of primaquine base/kg). The therapeutic index of this conjugate was at least 12 times higher than that of the free drug. This allowed the administration of a dose that cured 100% of the animals (17.5 mg of primaquine base/kg), in a single injection. With unmodified serum albumin the conjugate showed an increased therapeutic efficacy (the CPD50 was approximately 10 mg of primaquine base/kg) and a strongly reduced lethal toxicity.


Subject(s)
Malaria/drug therapy , Primaquine/analogs & derivatives , Serum Albumin, Bovine , Animals , Disulfides , Drug Stability , Lethal Dose 50 , Mice , Plasmodium berghei , Primaquine/administration & dosage , Primaquine/therapeutic use , Primaquine/toxicity , Serum Albumin/administration & dosage , Serum Albumin, Bovine/administration & dosage , Sulfhydryl Compounds
15.
J Med Chem ; 28(12): 1959-62, 1985 Dec.
Article in English | MEDLINE | ID: mdl-4068013

ABSTRACT

Two isomeric sulfur-interrupted 8-amino side chain analogues of 4-methyl-5-[m-(trifluoromethyl)phenoxy]primaquine (2) were prepared and tested for antimalarial activity. The compounds were evaluated for blood schizonticidal activity against Plasmodium berghei in mice and radical curative activity against Plasmodium cynomolgi in rhesus monkeys. In addition, they were evaluated for causal prophylactic activity against Plasmodium berghei yoelii in mice. Both compounds were more active and less toxic than primaquine in the P. berghei screen. One of the compounds showed radical curative activity similar to primaquine but was less active than 2. One of the compounds was active at 160 mg/kg in the P. berghei yoelii screen; the other was not active.


Subject(s)
Aminoquinolines/therapeutic use , Malaria/drug therapy , Aminoquinolines/chemical synthesis , Aminoquinolines/toxicity , Animals , Chemical Phenomena , Chemistry , Macaca mulatta , Malaria/prevention & control , Mice , Plasmodium , Primaquine/therapeutic use , Primaquine/toxicity
16.
J Med Chem ; 47(2): 285-7, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14711300

ABSTRACT

To eliminate an unwarranted metabolic pathway of the quinoline ring, a set of two compounds, where C-2 position of the antimalarial drug primaquine is blocked by metabolically stable bulky alkyl group are synthesized. Compound 2 [R = C(CH(3))(3)] of the series has produced excellent antimalarial efficacy against P. berghei and highly virulent multidrug-resistant P. yoelii nigeriensis strain in vivo. Compound 2 was also evaluated for methemoglobin (MetHb) toxicity. This study describes the discovery of a highly potent blood-schizontocidal antimalarial analogue 2, completely devoid of MetHb toxicity.


Subject(s)
Antimalarials/chemical synthesis , Methemoglobin/analysis , Primaquine/analogs & derivatives , Primaquine/chemical synthesis , Animals , Antimalarials/pharmacology , Antimalarials/toxicity , Malaria/blood , Malaria/mortality , Malaria/parasitology , Mice , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects , Primaquine/pharmacology , Primaquine/toxicity , Structure-Activity Relationship
17.
Biochem Pharmacol ; 31(21): 3501-7, 1982 Nov 01.
Article in English | MEDLINE | ID: mdl-7150371

ABSTRACT

Plasma clearance, urinary excretion and tissue distribution of radiolabeled free (FPQ) and liposome-entrapped Primaquine (LPQ) in mice were monitored for 2 hr following intravenous administration. FPQ is eliminated very rapidly from the plasma and excreted predominantly in the urine, probably largely in a metabolized form. In decreasing order of magnitude, pronounced accumulation of label occurs in the liver, kidneys, lungs and skeletal muscle. Less than 1 per cent of the total initial dose is recovered in other tissues. Partial erythrocytic sequestration results in drug levels higher and more persistent in blood cells than in the plasma. Compared to the free drug form, Primaquine entrapped within negatively charged liposomes of the cholesterol-rich multilamellar type exhibits a prolonged plasmatic half-life and, within the observation period, excretion is 8-fold reduced. Liver accumulation of label is doubled, accounting for close to 50% of the injected dose; splenic uptake is tripled, while accumulation in the lungs, kidneys, heart and brain is drastically reduced. These differences in pharmacodynamic behaviour may explain why liposomal entrapment leads to diminished acute Primaquine toxicity.


Subject(s)
Liposomes/administration & dosage , Primaquine/metabolism , Animals , Injections, Intravenous , Kinetics , Male , Mice , Phosphatidylcholines , Phosphatidylethanolamines , Phosphatidylserines , Primaquine/administration & dosage , Primaquine/blood , Primaquine/toxicity , Tissue Distribution
18.
Biochem Pharmacol ; 47(2): 309-16, 1994 Jan 20.
Article in English | MEDLINE | ID: mdl-8304975

ABSTRACT

The oxidative activities of primaquine [6-methoxy-8-(4-amino-1-methylbutylamino)quinoline] and its metabolites, the quinone-imine derivatives of 5-hydroxyprimaquine [5-hydroxy-6-methoxy-8-(4-amino-1-methylbutylamino)quinoline] and 5-hydroxydemethylprimaquine [5-hydroxy-6-demethyl-8-(4-amino-1-methylbutylamino)quinoline], 6-methoxy-8-amino quinoline and hydrogen peroxide, were studied on rat erythrocytes in vitro and in vivo. In both cases, the most effective metabolites in oxidizing hemoglobin and depleting non-protein sulfhydryl groups from erythrocytes were the quinone-imine derivatives of the ring-hydroxylated metabolites, 5-hydroxyprimaquine and 5-hydroxydemethyl-primaquine. The latter quinone-imines were shown by light absorption spectroscopy and oxygen consumption studies to be able to oxidize purified rat hemoglobin to methemoglobin but to be unable to react directly with reduced glutathione. In agreement with these results, no radical adduct was detected by electron paramagnetic resonance spectroscopy in incubations of rat erythrocytes with the quinone-imines and the spin-trap 5,5-dimethyl-1-pyrroline-N-oxide; metabolite-derived free radicals were detected instead. Taken together, the results suggest that 5-hydroxyprimaquine and 5-hydroxydemethylprimaquine are important metabolites in the expression of primaquine hemotoxicity, in contrast to 6-methoxy-8-aminoquinoline. Additionally, the results indicate that hydrogen peroxide is the ultimate oxidant formed from the ring-hydroxylated metabolites by redox-cycling of the corresponding quinone-imine derivatives both in vitro and in vivo.


Subject(s)
Erythrocytes/metabolism , Primaquine/metabolism , Animals , Biotransformation , Cyclic N-Oxides , Electron Spin Resonance Spectroscopy , Erythrocytes/drug effects , Hydrogen Peroxide/metabolism , Imines/isolation & purification , Male , Methemoglobin/analysis , Oxidation-Reduction , Primaquine/analogs & derivatives , Primaquine/toxicity , Quinones/isolation & purification , Rats , Rats, Wistar
19.
Biochem Pharmacol ; 37(13): 2683-90, 1988 Jul 01.
Article in English | MEDLINE | ID: mdl-2839199

ABSTRACT

The interaction of certain metabolites of the 8-aminoquinoline antimalarial primaquine with both normal and glucose-6-phosphate dehydrogenase (G6PD)-deficient erythrocytes and with haemoglobin preparations was studied in an attempt to elucidate the mechanisms of methaemoglobin formation and haemolytic anaemia associated with the use of primaquine. Studies using erythrocytes revealed that oxidation of haemoglobin and reduced glutathione (GSH) was due to the metabolites rather than the parent drug. Incubation of free haemoglobin with 5-hydroxylated metabolites of primaquine also led to oxidation of oxyhaemoglobin and GSH. Oxidation of GSH also occurred in the absence of oxyhaemoglobin. The results suggest a dual mechanism for these oxidative effects, involving autoxidation of the 5-hydroxy-8-aminoquinolines and their coupled oxidation with oxyhaemoglobin. The initial products of these processes would be drug metabolite free radicals, superoxide radical anions, hydrogen peroxide and methaemoglobin. Further free radical reactions would lead to oxidation of GSH, more haemoglobin and probably other cellular constituents. NADPH had no effect on the oxidative effects of the primaquine metabolites in these experiments. In the G6PD-deficient erythrocyte, the oxidation of haemoglobin and GSH leads to Heinz body formation and eventually to haemolysis, the mechanisms of which are as yet unclear. The possible role of oxygen free radicals in the mode of action of 8-aminoquinolines against the malaria parasite is also briefly discussed.


Subject(s)
Erythrocytes/drug effects , Primaquine/blood , Aminoquinolines/pharmacology , Ditiocarb/pharmacology , Edetic Acid/pharmacology , Glucosephosphate Dehydrogenase Deficiency/blood , Glutathione/blood , Methemoglobin/metabolism , Oxidation-Reduction , Oxyhemoglobins/metabolism , Primaquine/analogs & derivatives , Primaquine/pharmacology , Primaquine/toxicity
20.
Am J Trop Med Hyg ; 41(6): 638-42, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2641641

ABSTRACT

Methemoglobin toxicities of primaquine and compound N1-(3-acetyl-4-5-dihydro-2-furanyl)-N4-(6-methoxy-8-quinolinyl) 1,4-pentanediamine, CDRI Code 80/53, have been compared in beagles. Primaquine administration at 3 mg/kg for 7 days produced significantly high (P less than 0.001) methemoglobinemia and the levels increased 10.55-fold. Compound 80/53 at 3.75 mg/kg x 7 days produced a marginal increase in methemoglobinemia (3.24-fold; P less than 0.02). The methemoglobin formed by primaquine administration was 3.65-fold (P less than 0.001) higher than that formed after administration of compound 80/53. There was no significant change in other hematological parameters and liver function tests.


Subject(s)
Aminoquinolines/toxicity , Antimalarials/toxicity , Methemoglobinemia/chemically induced , Primaquine/toxicity , Animals , Appetite/drug effects , Body Weight/drug effects , Dogs , Female , Male
SELECTION OF CITATIONS
SEARCH DETAIL