Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 138
1.
Malar J ; 21(1): 169, 2022 Jun 03.
Article En | MEDLINE | ID: mdl-35659684

BACKGROUND: The enantiomers of the 8-aminoquinoline anti-malarial primaquine have different pharmacological properties. Development of an analytical method for simultaneous quantification of the enantiomers of primaquine and its metabolite, carboxyprimaquine, will support clinical pharmacometric assessments. METHODS: A simple and sensitive method consisting of liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was developed for simultaneous and enantiospecific determination of primaquine and its metabolite, carboxyprimaquine, in human plasma. Stable isotopes were used as internal standards to compensate for potential interference and matrix effects. Plasma samples (100 µL) were precipitated with 1% formic acid in acetonitrile followed by phospholipid removal solid phase extraction. Primaquine and carboxyprimaquine enantiomers were separated on a Chiralcel OD-3R (150 mm × 4.6 mm; I.D. 3 µm) column using a LC gradient mode. For separation of racemic primaquine and carboxyprimaquine, the LC method was modified and validated using a reverse phase column (Hypersil Gold 100 mm × 4.6 mm; I.D. 3 µm) and a mobile phase composed of 10 mM ammonium acetate buffer, pH 3.5 and acetonitrile in the isocratic mode. Method validation was performed according to regulatory guidelines. RESULTS: The calibration range was set to 0.571-260 ng/mL and 2.44-2,500 ng/mL for primaquine and carboxyprimaquine enantiomers, respectively, resulting in a correlation coefficient (r2) ≥ 0.0998 for all calibration curves. The intra- and inter-day assay precisions were < 10% and the accuracy was between 94.7 to 103% for all enantiomers of primaquine and carboxyprimaquine. The enantiospecific method was also modified and validated to quantify racemic primaquine and carboxyprimaquine, reducing the total run time from 30 to 8 min. The inter-, intra-day assay precision of the racemic quantification method was < 15%. The absolute recoveries of primaquine and carboxyprimaquine were between 70 and 80%. Stability was demonstrated for up to 2 years in - 80 °C. Both the enantiomeric and racemic LC-MS/MS methods were successfully implemented in pharmacokinetic studies in healthy volunteers. CONCLUSIONS: Simple, sensitive and accurate LC-MS/MS methods for the quantification of enantiomeric and racemic primaquine and carboxyprimaquine in human plasma were validated successfully and implemented in clinical routine drug analysis.


Primaquine , Tandem Mass Spectrometry , Acetonitriles , Chromatography, High Pressure Liquid , Chromatography, Liquid/methods , Humans , Primaquine/analogs & derivatives , Reproducibility of Results , Tandem Mass Spectrometry/methods
2.
Antimicrob Agents Chemother ; 66(3): e0182121, 2022 03 15.
Article En | MEDLINE | ID: mdl-34978892

The active metabolites of primaquine, in particular 5-hydroxyprimaquine, likely responsible for the clearance of dormant hypnozoites, are produced through the hepatic CYP450 2D6 (CYP2D6) enzymatic pathway. With the inherent instability of 5-hydroxyprimaquine, a stable surrogate, 5,6-orthoquinone, can now be detected and measured in the urine as part of primaquine pharmacokinetic studies. This study performed CYP450 2D6 genotyping and primaquine pharmacokinetic testing, to include urine 5,6-orthoquinone, in 27 healthy adult Cambodians, as a preliminary step to prepare for future clinical studies assessing primaquine efficacy for Plasmodium vivax infections. The CYP2D6 *10 reduced activity allele was found in 57% of volunteers, and the CYP2D6 genotypes were dominated by *1/*10 (33%) and *10/*10 (30%). Predicted phenotypes were evenly split between Normal Metabolizer (NM) and Intermediate Metabolizer (IM) except for one volunteer with a gene duplication and unclear phenotype, classifying as either IM or NM. Median plasma primaquine (PQ) area under the curve (AUC) was lower in the NM group (460 h*ng/mL) compared to the IM group (561 h*ng/mL), although not statistically significant. Similar to what has been found in the US study, no 5,6-orthoquinone was detected in the plasma. The urine creatinine-corrected 5,6-orthoquinone AUC in the NM group was almost three times higher than in the IM group, with peak measurements (Tmax) at 4 h. Although there is variation among individuals, future studies examining the relationship between the levels of urine 5,6-orthoquinone and primaquine radical cure efficacy could result in a metabolism biomarker predictive of radical cure.


Antimalarials , Malaria, Vivax , Adult , Antimalarials/pharmacokinetics , Antimalarials/therapeutic use , Asian People , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Humans , Malaria, Vivax/drug therapy , Plasmodium vivax/genetics , Primaquine/analogs & derivatives , Primaquine/pharmacokinetics , Primaquine/therapeutic use
3.
Antimicrob Agents Chemother ; 65(11): e0130221, 2021 10 18.
Article En | MEDLINE | ID: mdl-34398667

Primaquine is the only widely available drug for radical cure of Plasmodium vivax malaria. There is uncertainty whether the pharmacokinetic properties of primaquine are altered significantly in childhood or not. Patients with uncomplicated P. vivax malaria and with normal glucose-6-phosphate dehydrogenase were randomized to receive either chloroquine (25 mg base/kg of body weight) or dihydroartemisinin-piperaquine (dihydroartemisinin at 7 mg/kg and piperaquine at 55 mg/kg) plus primaquine, given either as 0.5 mg base/kg/day for 14 days or 1 mg/kg/day for 7 days. Predose day 7 venous plasma concentrations of chloroquine, desethylchloroquine, piperaquine, primaquine, and carboxyprimaquine were measured. Methemoglobin levels were measured at frequent intervals. Day 7 primaquine and carboxyprimaquine concentrations were available for 641 patients. After adjustment for the milligram-per-kilogram primaquine daily dose, day of sampling, partner drug, and fever clearance, there was a significant nonlinear relationship between age and trough primaquine and carboxyprimaquine concentrations and daily methemoglobin levels. Compared to adults 30 years of age, children 5 years of age had trough primaquine concentrations that were 0.53 (95% confidence interval [CI], 0.39 to 0.73)-fold lower, trough carboxyprimaquine concentrations that were 0.45 (95% CI, 0.35 to 0.55)-fold lower, and day 7 methemoglobin levels that were 0.87 (95% CI, 0.58 to 1.27)-fold lower. Increasing plasma concentrations of piperaquine and chloroquine and poor metabolizer CYP 2D6 alleles were associated with higher day 7 primaquine and carboxyprimaquine plasma concentrations. Higher blood methemoglobin concentrations were associated with a lower risk of recurrence. Young children have lower primaquine and carboxyprimaquine exposures and lower levels of methemoglobinemia than adults. Young children may need higher weight-adjusted primaquine doses than adults. (This study has been registered at ClinicalTrials.gov under identifier NCT01640574.).


Antimalarials , Malaria, Vivax , Adult , Antimalarials/therapeutic use , Child , Child, Preschool , Chloroquine/therapeutic use , Humans , Malaria, Vivax/drug therapy , Primaquine/analogs & derivatives , Primaquine/therapeutic use
4.
Molecules ; 26(13)2021 Jun 30.
Article En | MEDLINE | ID: mdl-34208832

The rapid emergence of drug resistance to the current antimalarial agents has led to the urgent need for the discovery of new and effective compounds. In this work, a series of 5-phenoxy primaquine analogs with 8-aminoquinoline core (7a-7h) was synthesized and investigated for their antimalarial activity against Plasmodium falciparum. Most analogs showed improved blood antimalarial activity compared to the original primaquine. To further explore a drug hybrid strategy, a conjugate compound between tetraoxane and the representative 5-phenoxy-primaquine analog 7a was synthesized. In our work, the hybrid compound 12 exhibited almost a 30-fold increase in the blood antimalarial activity (IC50 = 0.38 ± 0.11 µM) compared to that of primaquine, with relatively low toxicity against mammalian cells (SI = 45.61). Furthermore, we found that these 5-phenoxy primaquine analogs and the hybrid exhibit significant heme polymerization inhibition, an activity similar to that of chloroquine, which could contribute to their improved antimalarial activity. The 5-phenoxy primaquine analogs and the tetraoxane hybrid could serve as promising candidates for the further development of antimalarial agents.


Antimalarials , Erythrocytes/parasitology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/growth & development , Primaquine , Tetraoxanes , Adult , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antimalarials/pharmacology , Erythrocytes/metabolism , Erythrocytes/pathology , Female , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/pathology , Male , Middle Aged , Primaquine/analogs & derivatives , Primaquine/chemical synthesis , Primaquine/chemistry , Primaquine/pharmacology , Tetraoxanes/chemical synthesis , Tetraoxanes/chemistry , Tetraoxanes/pharmacology
5.
Brasília; CONITEC; mar. 2021.
Non-conventional Pt | BRISA | ID: biblio-1178759

CONITEC CONTEXTO: A malária é uma doença infecciosa parasitária aguda causada por protozoários do gênero Plasmodium, transmitidos ao homem pela picada da fêmea do mosquito Anopheles darlingi. O período de incubação da condição varia de 7 a 14 dias e a crise aguda é caracterizada por episódios de calafrios, febre e sudorese, geralmente acompanhados de cefaleia, mialgia, náuseas e vômitos. De acordo com o Relatório Mundial da Malária, 228 milhões de casos foram reportados, no ano de 2019, representando um grave problema de saúde pública para o mundo. No Brasil, a área endêmica compreende a região amazônica brasileira. Em 2019, foram notificadas no território nacional 157.454 casos de malária, uma redução de 19,1% em relação a 2018, quando foram registrados 194.572 casos da doença. Já a deficiência de glicose-6-fosfato desidrogenase (G6PD) é uma anomalia hereditária ligada ao cromossomo X, que acomete majoritariamente homens (hemizigóticos). Estima-se que afeta aproximadamente 400 milhões de pessoas em todo o mundo e a prevalência varia de 5% a 25% em áreas endêmicas, como África, Oriente Médio e Ásia. Essa enzima desempenha papel importante na sobrevivência dos eritrócitos: está envolvida na via da pentose fosfato (PPP) e fornece NADPH (nicotina adenina dinucleótido fosfato reduzido) e GSH (glutationa reduzida). GSH pode reagir com peróxido de hidrogênio (H2O2) e reduzir para H2O. Isso ajuda a proteger os eritrócitos de espécies reativas de oxigênio, que resultam em estresse oxidativo e consequentemente, hemólise. A principal preocupação de segurança com relação à tafenoquina é o alto risco de anemia hemolítica aguda (AHA) em pacientes com deficiência de G6PD (atividade da enzima < 30% do normal), que pode resultar em óbitos em indivíduos com menos de 10% da atividade enzimática normal. TECNOLOGIA: Tafenoquina (Kozenis®). PERGUNTAS DE PESQUISA: A tafenoquina 300 mg em dose única é eficaz e segura para cura radical (prevenção de recidiva) da malária por Plasmodium vivax? O teste quantitativo da atividade da enzima glicose-6-fosfato desidrogenase (G6PD) é sensível e específico na detecção da atividade da G6PD em pacientes com diagnóstico confirmado de malária por Plasmodium vivax? EVIDÊNCIAS CIENTÍFICAS: Foram incluídos dois estudos no corpo da evidência, duas revisões sistemáticas com meta-análise, de qualidade da evidência moderada a grave. No que tange às evidências que respondem à primeira pergunta de pesquisa, referente ao medicamento, para os desfechos avaliados nos grupos tafenoquina versus nenhum tratamento antihipinozoíto: tafenoquina reduziu a recidiva em comparação com não tratamento (RR 0,32, IC 95% 0,12 a 0,88); não houve mortes durante acompanhamento; não foram encontradas diferenças significativas entre tafenoquina e nenhum tratamento anti-hipnozoíto (RR 1,34, IC 95% 0,63 a 2,84) e não foram observadas diferença entre os grupos avaliados em relação ao número ou tipo de eventos adversos relatados com exceção de dor de cabeça. O grupo tratado com tafenoquina mais cloroquina demonstrou pouca ou nenhuma diferença na ocorrência geral de eventos adversos em comparação com cloroquina isolada (RR 0,96, IC 95% 0,81 a 1,13). Para o desfechos avaliados que compararam tafenoquina versus primaquina: não foi identificada diferença estatisticamente significativa na prevenção de recaídas entre os pacientes dos tratamentos (RR 1,04, IC 95% 0,8 a 1,34), assim, tafenoquina é possivelmente tão eficaz quanto primaquina; também não foi identificada diferença estatisticamente significativa entre os tratamentos em pacientes sem deficiência de G6PD; entre os EA mais comuns estão a queda no nível de hemoglobina e prolongamento QT assintomático (RR 1,41, IC 95% 0,70 a 2,83); para avaliação de qualquer evento adverso tafenoquina não apresentou diferença em todos os tipos de EA em comparação com primaquina, incluindo anemia e queda do nível de hemoglobina (RR 1,01, IC 95% 0,89 a 1,14). Para avaliação da acurácia do teste quantitativo de G6PD, a sensibilidade combinada do teste foi de 0,96 (IC 95% 0,90 a 0,99) e a especificidade combinada foi de 0,95 (IC 95% 0,92 a 0,96), sendo que o desempenho combinado não variou significativamente, independentemente do tipo de amostra sanguínea. CONSIDERAÇÕES: Com nível de certeza moderado, a dose única de tafenoquina 300 mg não teve diferença significativa quando comparado com o tratamento de primaquina 15 mg/dia por 14 dias. Também não foi identificada diferença significativa em relação aos eventos adversos graves e gerais entre os outros grupos comparadores, com uma certeza de evidência moderada a alta. O perfil de segurança dos dois tratamentos foi semelhante e ambos causaram declínios no nível de hemoglobina, no entanto de fácil manejo, entre os pacientes com atividade normal da enzima G6PD. Também com nível de certeza da evidência moderado, o teste quantitativo de atividade da enzima G6PD performou valores de sensibilidade e especificidade maiores que 95%. As razões de verossimilhança positiva e negativa sugerem que o teste é adequado para confirmação da atividade da enzima, bem como auxilia na exclusão de casos em que há deficiência de G6PD em um limiar de 30% de atividade enzimática. RECOMENDAÇÃO PRELIMINAR DA CONITEC: Diante do exposto, a Conitec, em sua 94ª reunião ordinária, realizada no dia 04 de fevereiro de 2021, deliberou que a matéria fosse disponibilizada em consulta pública com recomendação preliminar favorável à incorporação, no SUS, da tafenoquina para o tratamento, ou cura radical, de malária causada pelo Plasmodium vivax em pacientes com 16 anos de idade ou mais e atividade enzimática maior que 70% de glicose-6-fosfato desidrogenase (G6PD), confirmada por meio de teste rápido validado. Os membros do plenário concordaram que, embora a evidência de eficácia disponível em literatura tenha sido avaliada de boa qualidade, esta apresenta dados modestos, se considerado o principal desfecho que avalia o desempenho da tafenoquina versus primaquina. Para tanto, faz-se necessária a obtenção de dados de efetividade que serão coletados após condução do estudo observacional TRuST, focalizado nas cidades de Manaus e Porto Velho. A matéria foi disponibilizada em consulta pública. CONSULTA PÚBLICA: A Consulta Pública nº 04/2021 foi realizada entre os dias 08/02/2021 a 1º/03/2021. Foram recebidas 87 contribuições, sendo 32 pelo formulário para contribuições técnico-científicas e 55 pelo formulário para contribuições sobre experiência ou opinião de pacientes, familiares, amigos ou cuidadores de pacientes, profissionais de saúde ou pessoas interessadas no tema. As contribuições recebidas foram majoritariamente a favor da recomendação preliminar da Conitec, de incorporação de uso das tecnologias no SUS. RECOMENDAÇÃO FINAL: Os membros do plenário presentes na 95ª reunião ordinária da Conitec, no dia 03 de março de 2021, deliberaram, por unanimidade, recomendar a incorporação da tafenoquina para o tratamento, ou cura radical, de malária causada pelo Plasmodium vivax em pacientes com 16 anos de idade ou mais e atividade enzimática de glicose-6- fosfato desidrogenase (G6PD) maior que 70%, confirmada por meio de teste rápido quantitativo de G6PD. A recomendação para incorporação do medicamento está condicionada à apresentação de dados de mundo real ao final do estudo de 12 meses. Cabe informar que não foram adicionadas na consulta pública referências que alterassem a análise da evidência apresentada no relatório preliminar. Foi assinado o Registro de Deliberação nº 591/2021. DECISÃO: Incorporar a tafenoquina para tratamento de pacientes com malária por Plasmodium vivax condicionada à apresentação de dados de mundo real ao final do estudo, do Sistema Único de Saúde - SUS, conforme Portaria nº 07, publicada no Diário Oficial da União nº 48, seção 1, página 170, em 12 de março de 2021.


Humans , Plasmodium vivax/drug effects , Primaquine/analogs & derivatives , Malaria/drug therapy , Technology Assessment, Biomedical , Unified Health System , Brazil , Cost-Benefit Analysis/economics
7.
Article En | MEDLINE | ID: mdl-33387859

The antimalarial drug primaquine (PQ) causes methemoglobinemia and hemolysis in individuals with a genetic deficiency of glucose 6-phosphate dehydrogenase. Reactive oxygen species (ROS) generated by redox cycling of the metabolite primaquine-5,6-orthoquinone (POQ) in erythrocytes has been attributed to be responsible for the toxicity of PQ. Carboxyprimaquine (CPQ), the major human plasma metabolite of PQ, can also form the analogous carboxyprimaquine-5,6-orthoquinone (CPOQ) metabolite, which can also generate ROS in erythrocytes by redox cycling, thus contributing to the hematotoxicity of this drug. In order to study these pathways and characterize such effects in vivo, methods are needed for characterization and quantification of POQ and CPOQ in human erythrocytes. The purpose of this work was to develop a validated method for the quantitative determination of CPOQ and POQ metabolites in human erythrocytes, suitable for clinical studies of PQ metabolism. Several liquid-liquid extraction methods using different organic solvents had been investigated. The solvent mixture of water-methanol-acetonitrile (9:9:5, v/v) was shown to yield the best results for the two analytes. Chromatographic analysis of POQ and CPOQ in human erythrocytes was achieved on a high strength silica (HSS) column and gradient elution (water and acetonitrile, both containing 0.1% formic acid) by ultra-high-performance liquid chromatography coupled with tandem mass spectrometry (UHPLC-MS/MS). Quantitative estimation of POQ and CPOQ was executed by monitoring ion pairs of m/z 260.23 > 175.03 and m/z 275.19 > 175.04, respectively. The method, which was validated for precision, accuracy, selectivity, and linearity, was successfully applied for the quantitative determination of POQ and CPOQ, the key metabolites of PQ in human erythrocytes in PQ clinical study.


Chromatography, High Pressure Liquid/methods , Primaquine/analogs & derivatives , Primaquine/blood , Tandem Mass Spectrometry/methods , Erythrocytes/chemistry , Humans , Limit of Detection , Linear Models , Reproducibility of Results
8.
Rev Inst Med Trop Sao Paulo ; 60: e66, 2018 Oct 25.
Article En | MEDLINE | ID: mdl-30379233

In the last two years, a substantial increase in the number of malaria vivax cases has occurred in the Brazilian Amazon basin. The adequate exposure of hypnozoites to primaquine is a matter of interest as these dormant forms are responsible for the maintenance or even the increase of malaria burden in endemic areas. The aim of this study was to estimate the levels of primaquine and carboxyprimaquine in whole blood samples of patients with P. vivax treated with chloroquine and an abbreviated regimen of primaquine (0.5 mg/kg/d for 7 days), with adequate clinical and parasitological outcomes after 180 days of follow-up. A total of 40 male patients met the criteria for inclusion in the study. Primaquine and carboxyprimaquine were measured by high-performance liquid chromatography. The levels of primaquine in whole blood samples ranged from 40-238 ng/mL, 42-196 ng/mL and 42-150 ng/mL on days 1, 3 and 7. The levels of carboxyprimaquine in whole blood samples ranged from 87-234 ng/mL, 96-252 ng/mL and 74-448 ng/mL on days 1, 3 and 7. These data provide a reliable estimation of exposure of the infecting parasite to primaquine. Based on the regional pattern of relapse, the estimated blood levels of primaquine can be considered effective against hypnozoites of the local circulating strains of P. vivax.


Antimalarials/blood , Malaria, Vivax/blood , Malaria, Vivax/drug therapy , Primaquine/analogs & derivatives , Primaquine/blood , Adult , Antimalarials/administration & dosage , Brazil , Chromatography, High Pressure Liquid , Drug Administration Schedule , Drug Therapy, Combination , Follow-Up Studies , Humans , Male , Primaquine/administration & dosage , Prospective Studies
9.
Drug Metab Pharmacokinet ; 33(6): 250-257, 2018 Dec.
Article En | MEDLINE | ID: mdl-30366777

Cytochrome P450 2D6 (CYP2D6) is responsible for the metabolic activation of primaquine, an antimalarial drug. CYP2D6 is genetically polymorphic, and these polymorphisms are associated with interindividual variations observed in the therapeutic efficacy of primaquine. To further understand this association, we performed in vitro enzymatic analyses of the wild-type CYP2D6.1 and 49 CYP2D6 allelic variants, which were expressed in 293FT cells, using primaquine as a substrate. The concentrations of CYP2D6 variant holoenzymes were measured by using carbon monoxide (CO)-reduced difference spectroscopy, and the wild type and 27 variants showed a peak at 450 nm. The kinetic parameters Km, Vmax, and intrinsic clearance (Vmax/Km) of primaquine 5-hydroxylation were characterized. The kinetic parameters of the wild type and 16 variants were measured, but the values for the remaining 33 variants could not be determined because of low metabolite concentrations. Among the variants, six (i.e., CYP2D6.17, .18, .35, .39, .53, and .70) showed significantly reduced intrinsic clearance compared with that of CYP2D6.1. Three-dimensional structural modeling analysis was performed to elucidate the mechanism of changes in the kinetics of CYP2D6 variants. Our findings provide insights into the allele-specific activity of CYP2D6 for primaquine, which could be clinically useful for malaria treatment and eradication efforts.


Alleles , Cytochrome P-450 CYP2D6/genetics , Genetic Variation/genetics , Primaquine/metabolism , Cytochrome P-450 CYP2D6/metabolism , HEK293 Cells , Humans , Hydroxylation , Molecular Structure , Primaquine/analogs & derivatives , Primaquine/chemistry
10.
Eur J Med Chem ; 158: 68-81, 2018 Oct 05.
Article En | MEDLINE | ID: mdl-30199706

Emergence of drug resistance and targeting all stages of the parasite life cycle are currently the major challenges in antimalarial chemotherapy. Molecular hybridization combining two scaffolds in a single molecule is an innovative strategy for achieving these goals. In this work, a series of novel quinoxaline 1,4-di-N-oxide hybrids containing either chloroquine or primaquine pharmacophores was designed, synthesized and tested against both chloroquine sensitive and multidrug resistant strains of Plasmodium falciparum. Only chloroquine-based compounds exhibited potent blood stage activity with compounds 4b and 4e being the most active and selective hybrids at this parasite stage. Based on their intraerythrocytic activity and selectivity or their chemical nature, seven hybrids were then evaluated against the liver stage of Plasmodium yoelii, Plasmodium berghei and Plasmodium falciparum infections. Compound 4b was the only chloroquine-quinoxaline 1,4-di-N-oxide hybrid with a moderate liver activity, whereas compound 6a and 6b were identified as the most active primaquine-based hybrids against exoerythrocytic stages, displaying enhanced liver activity against P. yoelii and P. berghei, respectively, and better SI values than primaquine. Although both primaquine-quinoxaline 1,4-di-N-oxide hybrids slightly reduced the infection of mosquitoes, they inhibited sporogony of P. berghei and compound 6a showed 92% blocking of transmission. In vivo liver efficacy assays revealed that compound 6a showed causal prophylactic activity affording parasitaemia reduction of up to 95% on day 4. Absence of genotoxicity and in vivo acute toxicity were also determined. These results suggest the approach of primaquine-quinoxaline 1,4-di-N-oxide hybrids as new potential dual-acting antimalarials for further investigation.


Antimalarials/chemistry , Antimalarials/pharmacology , Chloroquine/analogs & derivatives , Chloroquine/pharmacology , Plasmodium/drug effects , Primaquine/analogs & derivatives , Primaquine/pharmacology , Animals , Antimalarials/therapeutic use , Chloroquine/therapeutic use , Female , Hep G2 Cells , Humans , Life Cycle Stages/drug effects , Malaria/drug therapy , Malaria/prevention & control , Mice, Inbred BALB C , Plasmodium/physiology , Primaquine/therapeutic use , Quinoxalines/chemistry , Quinoxalines/pharmacology , Quinoxalines/therapeutic use
11.
Clin Infect Dis ; 67(7): 1000-1007, 2018 09 14.
Article En | MEDLINE | ID: mdl-29590311

Background: Primaquine is the only drug providing radical cure of Plasmodium vivax malaria. It is not recommended for breastfeeding women as it causes hemolysis in glucose-6-phosphate dehydrogenase (G6PD)-deficient individuals, and breast milk excretion and thus infant exposure are not known. Methods: Healthy G6PD-normal breastfeeding women with previous P. vivax infection and their healthy G6PD-normal infants between 28 days and 2 years old were enrolled. Mothers took primaquine 0.5 mg/kg/day for 14 days. Primaquine and carboxyprimaquine concentrations were measured in maternal venous plasma, capillary plasma, and breast milk samples and infant capillary plasma samples taken on days 0, 3, 7, and 13. Results: In 20 mother-infant pairs, primaquine concentrations were below measurement thresholds in all but 1 infant capillary plasma sample (that contained primaquine 2.6 ng/mL), and carboxyprimaquine was likewise unmeasurable in the majority of infant samples (maximum value 25.8 ng/mL). The estimated primaquine dose received by infants, based on measured breast milk levels, was 2.98 µg/kg/day (ie, ~0.6% of a hypothetical infant daily dose of 0.5 mg/kg). There was no evidence of drug-related hemolysis in the infants. Maternal levels were comparable to levels in nonlactating patients, and adverse events in mothers were mild. Conclusions: The concentrations of primaquine in breast milk are very low and therefore very unlikely to cause adverse effects in the breastfeeding infant. Primaquine should not be withheld from mothers breastfeeding infants or young children. More information is needed in neonates. Clinical Trials Registration: NCT01780753.


Antimalarials/pharmacokinetics , Antimalarials/therapeutic use , Malaria, Vivax/drug therapy , Milk, Human/chemistry , Primaquine/pharmacokinetics , Primaquine/therapeutic use , Adolescent , Adult , Antimalarials/blood , Antimalarials/chemistry , Area Under Curve , Breast Feeding , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Lactation , Male , Primaquine/analogs & derivatives , Primaquine/blood , Primaquine/chemistry , Primaquine/metabolism , Young Adult
12.
Cochrane Database Syst Rev ; 2: CD008152, 2018 02 02.
Article En | MEDLINE | ID: mdl-29393511

BACKGROUND: The 8-aminoquinoline (8AQ) drugs act on Plasmodium falciparum gametocytes, which transmit malaria from infected people to mosquitoes. In 2012, the World Health Organization (WHO) recommended a single dose of 0.25 mg/kg primaquine (PQ) be added to malaria treatment schedules in low-transmission areas or those with artemisinin resistance. This replaced the previous recommendation of 0.75 mg/kg, aiming to reduce haemolysis risk in people with glucose-6-phosphate dehydrogenase deficiency, common in people living in malarious areas. Whether this approach, and at this dose, is effective in reducing transmission is not clear. OBJECTIVES: To assess the effects of single dose or short-course PQ (or an alternative 8AQ) alongside treatment for people with P. falciparum malaria. SEARCH METHODS: We searched the Cochrane Infectious Diseases Group Specialized Register; the Cochrane Central Register of Controlled Trials (CENTRAL), published in the Cochrane Library; and the WHO International Clinical Trials Registry Platform (ICRTP) portal using 'malaria*', 'falciparum', 'primaquine', '8-aminoquinoline', and eight 8AQ drug names as search terms. We checked reference lists of included trials, and contacted researchers and organizations. Date of last search: 21 July 2017. SELECTION CRITERIA: Randomized controlled trials (RCTs) or quasi-RCTs in children or adults, adding PQ (or alternative 8AQ) as a single dose or short course alongside treatment for P. falciparum malaria. DATA COLLECTION AND ANALYSIS: Two authors screened abstracts, applied inclusion criteria, and extracted data. We sought evidence on transmission (community incidence), infectiousness (people infectious and mosquitoes infected), and potential infectiousness (gametocyte measures assessed by microscopy or polymerase chain reaction [PCR]). We grouped trials into artemisinin and non-artemisinin treatments, and stratified by PQ dose (low, 0.2 to 0.25 mg/kg; moderate, 0.4 to 0.5 mg/kg; high, 0.75 mg/kg). We used GRADE, and absolute effects of infectiousness using trial control groups. MAIN RESULTS: We included 24 RCTs and one quasi-RCT, comprising 43 arms. Fourteen trials evaluated artemisinin treatments (23 arms), nine trials evaluated non-artemisinin treatments (13 arms), and two trials included both artemisinin and non-artemisinin arms (three and two arms, respectively). Two trial arms used bulaquine. Seven PQ arms used low dose (six with artemisinin), 11 arms used moderate dose (seven with artemisinin), and the remaining arms used high dose. Fifteen trials tested for G6PD status: 11 excluded participants with G6PD deficiency, one included only those with G6PD deficiency, and three included all, irrespective of status. The remaining 10 trials either did not test or did not report on testing.No cluster trials evaluating community effects on malaria transmission met the inclusion criteria.With artemisinin treatmentLow dose PQInfectiousness (participants infectious to mosquitoes) was reduced (day 3 or 4: RR 0.12, 95% CI 0.02 to 0.88, 3 trials, 105 participants; day 8: RR 0.34, 95% CI 0.07 to 1.58, 4 trials, 243 participants; low certainty evidence). This translates to a reduction in percentage of people infectious on day 3 or 4 from 14% to 2%, and, for day 8, from 4% to 1%; the waning infectiousness in the control group by day 8 making the absolute effect smaller by day 8. For gametocytes detected by PCR, there was little or no effect of PQ at day 3 or 4 (RR 1.02, 95% CI 0.87 to 1.21; 3 trials, 414 participants; moderate certainty evidence); with reduction at day 8 (RR 0.52, 95% CI 0.41 to 0.65; 4 trials, 532 participants; high certainty evidence). Severe haemolysis was infrequent, with or without PQ, in these groups with few G6PD-deficient individuals (RR 0.98, 95% CI 0.69 to 1.39; 4 trials, 752 participants, moderate certainty evidence).Moderate dose PQInfectiousness was reduced (day 3 or 4: RR 0.13, 95% CI 0.02 to 0.94; 3 trials, 109 participants; day 8 RR 0.33, 95% CI 0.07 to 1.57; 4 trials, 246 participants; low certainty evidence). Illustrative risk estimates for moderate dose were the same as low dose. The pattern and level of certainty of evidence with gametocytes detected by PCR was the same as low dose, and severe haemolysis was infrequent in both groups.High dose PQInfectiousness was reduced (day 4: RR 0.2, 95% CI 0.02 to 1.68, 1 trial, 101 participants; day 8: RR 0.18, 95% CI 0.02 to 1.41, 2 trials, 181 participants, low certainty evidence). The effects on gametocyte prevalence showed a similar pattern to moderate and low dose PQ. Trials did not systematically report evidence of haemolysis.With non-artemisinin treatmentTrials with non-artemisinin treatment have been conducted only for moderate and high dose PQ. With high dose, infectiousness appeared markedly reduced on day 5 (RR 0.09, 95% CI 0.01 to 0.62; 30 participants, very low certainty evidence), with similar reductions at day 8. For both moderate dose (two trials with 221 people) and high dose (two trials with 30 people), reduction in gametocytes (detected by microscopy) showed similar patterns as for artemisinin treatments, with little or no effect at day 4 or 5, and larger effects by day 8. No trials with non-artemisinin partner drugs systematically sought evidence of severe haemolysis.Two trials comparing bulaquine with PQ suggest bulaquine may have larger effects on gametocytes by microscopy on day 8 (RR 0.41, 95% CI 0.26 to 0.66; 2 trials, 112 participants). AUTHORS' CONCLUSIONS: A single low dose of PQ (0.25 mg/kg) added to artemisinin-based combination therapy for malaria reduces infectiousness of people to mosquitoes at day 3-4 and day 8, and appears as effective as higher doses. The absolute effect is greater at day 3 or 4, and smaller at day 8, in part because of the lower infectiousness in the control group. There was no evidence of increased haemolysis at 0.25 mg/kg, but few G6PD-deficient individuals were included in the trials. The effect on infectiousness precedes the effect of PQ on gametocyte prevalence. We do not know whether single dose PQ could reduce malaria transmission at community level.


Antimalarials/administration & dosage , Glucosephosphate Dehydrogenase Deficiency/diagnosis , Malaria, Falciparum/prevention & control , Primaquine/administration & dosage , Adult , Artemisinins/administration & dosage , Artemisinins/therapeutic use , Child , Chloroquine/administration & dosage , Chloroquine/therapeutic use , Drug Combinations , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Mefloquine/administration & dosage , Mefloquine/therapeutic use , Non-Randomized Controlled Trials as Topic , Plasmodium falciparum/drug effects , Primaquine/analogs & derivatives , Pyrimethamine/administration & dosage , Quinine/therapeutic use , Randomized Controlled Trials as Topic , Sulfadoxine/administration & dosage , Time Factors
13.
Article En | MEDLINE | ID: mdl-28289025

Low-dose primaquine is recommended to prevent Plasmodium falciparum malaria transmission in areas threatened by artemisinin resistance and areas aiming for malaria elimination. Community treatment campaigns with artemisinin-based combination therapy in combination with the gametocytocidal primaquine dose target all age groups, but no studies thus far have assessed the pharmacokinetics of this gametocytocidal drug in African children. We recruited 40 children participating in a primaquine efficacy trial in Burkina Faso to study primaquine pharmacokinetics. These children received artemether-lumefantrine and either a 0.25- or a 0.40-mg/kg primaquine dose. Seven blood samples were collected from each participant for primaquine and carboxy-primaquine plasma levels determinations: one sample was collected before primaquine administration and six after primaquine administration according to partially overlapping sampling schedules. Physiological population pharmacokinetic modeling was used to assess the impact of weight, age, and CYP2D6 genotype on primaquine and carboxy-primaquine pharmacokinetics. Despite linear weight normalized dosing, the areas under the plasma concentration-time curves and the peak concentrations for both primaquine and carboxy-primaquine increased with age and body weight. Children who were CYP2D6 poor metabolizers had higher levels of the parent compound, indicating a lower primaquine CYP2D6-mediated metabolism. Our data indicate that primaquine and carboxy-primaquine pharmacokinetics are influenced by age, weight, and CYP2D6 genotype and suggest that dosing strategies may have to be reconsidered to maximize the transmission-blocking properties of primaquine. (This study has been registered at ClinicalTrials.gov under registration no. NCT01935882.).


Antimalarials/therapeutic use , Cytochrome P-450 CYP2D6/genetics , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Primaquine/pharmacokinetics , Adolescent , Age Factors , Antimalarials/pharmacokinetics , Artemether, Lumefantrine Drug Combination , Artemisinins/therapeutic use , Body Weight , Burkina Faso , Child , Child, Preschool , Drug Combinations , Drug Resistance/genetics , Ethanolamines/therapeutic use , Female , Fluorenes/therapeutic use , Humans , Malaria, Falciparum/transmission , Male , Primaquine/analogs & derivatives , Primaquine/blood , Primaquine/therapeutic use
14.
Malar J ; 16(1): 110, 2017 03 09.
Article En | MEDLINE | ID: mdl-28279180

BACKGROUND: Primaquine is an anti-malarial used to prevent Plasmodium vivax relapses and malaria transmission. However, PQ metabolites cause haemolysis in patients deficient in the enzyme glucose-6-phosphate dehydrogenase (G6PD). Fifteen PQ-thiazolidinone derivatives, synthesized through one-post reactions from primaquine, arenealdehydes and mercaptoacetic acid, were evaluated in parallel in several biological assays, including ability to block malaria transmission to mosquitoes. RESULTS: All primaquine derivatives (PQ-TZs) exhibited lower cell toxicity than primaquine; none caused haemolysis to normal or G6PD-deficient human erythrocytes in vitro. Sera from mice pretreated with the test compounds thus assumed to have drug metabolites, caused no in vitro haemolysis of human erythrocytes, whereas sera from mice pretreated with primaquine did cause haemolysis. The ability of the PQ-TZs to block malaria transmission was evaluated based on the oocyst production and percentage of mosquitoes infected after a blood meal in drug pre-treated animals with experimental malaria caused by either Plasmodium gallinaceum or Plasmodium berghei; four and five PQ-TZs significantly inhibited sporogony in avian and in rodent malaria, respectively. Selected PQ-TZs were tested for their inhibitory activity on P. berghei liver stage development, in mice and in vitro, one compound (4m) caused a 3-day delay in the malaria pre-patent period. CONCLUSIONS: The compound 4m was the most promising, blocking malaria transmissions and reducing the number of exoerythrocytic forms of P. berghei (EEFs) in hepatoma cells in vitro and in mice in vivo. The same compound also caused a 3-day delay in the malaria pre-patent period.


Erythrocytes/parasitology , Glucosephosphate Dehydrogenase/metabolism , Malaria/drug therapy , Plasmodium berghei/drug effects , Plasmodium gallinaceum/drug effects , Primaquine/analogs & derivatives , Primaquine/pharmacology , Animals , Cell Line, Tumor , Chickens , Chlorocebus aethiops , Erythrocytes/drug effects , Hemolysis/drug effects , Hep G2 Cells , Humans , Malaria/transmission , Malaria, Avian/drug therapy , Malaria, Avian/transmission , Mice , Plasmodium berghei/growth & development , Plasmodium gallinaceum/growth & development
15.
Chem Biol Drug Des ; 90(2): 254-261, 2017 08.
Article En | MEDLINE | ID: mdl-28102941

Primaquine (PQ) is the only drug used to prevent relapse of malaria due to P. vivax and P. ovale, by eradicating the dormant liver form of the parasite (hypnozoites). The side-effects associated with PQ limits is uses in treatment of malaria. To overcome the premature oxidative deamination and to increase the life span of drug in the biological system, the novel glyco-conjugates of PQ were synthesized by coupling of primaquine with hexoses in phosphate buffer. The saccharide part of the hybrid molecules thought to direct the drug to the liver, where hypnozoites resides. All the synthesized compounds were fully characterized and evaluated for their radical curative activities. The three compounds viz glucoside (15a), galactoside (15b) and mannoside (15c) with high activity were tested for their activity in rhesus monkeys where the most active compound 15b showed twofold activity (100% radical curative activity at 1.92 mmol/kg) than the standard drug PQ diphosphate (3.861 mmol/kg). It is proposed that results from these studies may be advantageous to develop a new potent tissue schizonticide antimalarial compound.


Antimalarials/chemistry , Antimalarials/therapeutic use , Malaria/drug therapy , Plasmodium/drug effects , Primaquine/analogs & derivatives , Primaquine/therapeutic use , Animals , Antimalarials/chemical synthesis , Antimalarials/pharmacology , Female , Glycoconjugates/chemical synthesis , Glycoconjugates/chemistry , Glycoconjugates/pharmacology , Glycoconjugates/therapeutic use , Macaca mulatta , Malaria, Vivax/drug therapy , Male , Mice , Plasmodium cynomolgi/drug effects , Plasmodium vivax/drug effects , Primaquine/chemical synthesis , Primaquine/pharmacology
16.
Molecules ; 21(12)2016 Nov 28.
Article En | MEDLINE | ID: mdl-27916811

In this paper design and synthesis of a scaffold comprising primaquine (PQ) motif and cinnamic acid derivatives (CADs) bound directly (compounds 3a-k) or via a spacer (compounds 7a-k) are reported. In the first series of compounds, PQ and various CADs were connected by amide bonds and in the second series by acylsemicarbazide functional groups built from the PQ amino group, CONHNH spacer and the carbonyl group originating from the CADs. PQ-CAD amides 3a-k were prepared by a simple one-step condensation reaction of PQ with a series of CAD chlorides (method A) or benzotriazolides 2 (method B). The synthesis of acylsemicarbazides 7a-k included activation of PQ with benzotriazole, preparation of PQ-semicarbazide 6 and its condensation with CAD chlorides 4. All synthesized PQ-CAD conjugates were evaluated for their anticancer, antiviral and antioxidative activities. Almost all compounds from series 3 were selective towards the MCF-7 cell line and active at micromolar concentrations. The o-fluoro derivative 3h showed high activity against HeLa, MCF-7 and in particular against the SW 620 cell line, while acylsemicarbazide 7f with a benzodioxole ring and 7c, 7g and especially 7j with methoxy-, chloro- or trifluoromethyl-substituents in the para position showed high selectivity and high inhibitory activity against MCF-7 cell line at micromolar (7c, 7f, 7g) and nanomolar (7j) levels. Acylsemicarbazide derivatives with trifluoromethyl group(s) 7i, 7j and 7k showed specific activity against human coronavirus (229E) at concentrations which did not alter the normal cell morphology. The same compounds exerted the most potent reducing activity in the DPPH test, together with 7d and 7g, while methoxy (compounds 7c-e), benzodioxole (7f), p-Cl (7g) and m-CF3 (7i) acylsemicarbazides and amide 3f presented the highest LP inhibition (83%-89%). The dimethoxy derivative 7d was the most potent LOX inhibitor (IC50 = 10 µΜ). The performed biological tests gave evidence of acylsemicarbazide functional group as superior binding group in PQ-CAD conjugates.


Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cinnamates/pharmacology , Primaquine/pharmacology , Adenoviridae/drug effects , Animals , Antioxidants/chemical synthesis , Antioxidants/pharmacology , Cell Line, Tumor , Cinnamates/chemistry , Coronavirus/drug effects , HeLa Cells , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Humans , MCF-7 Cells , Mice , Neoplasms/drug therapy , Primaquine/analogs & derivatives , Primaquine/chemistry , Vaccinia virus/drug effects
17.
Antimicrob Agents Chemother ; 60(10): 5906-13, 2016 10.
Article En | MEDLINE | ID: mdl-27458212

Individuals with glucose-6-phosphate dehydrogenase (G6PD) deficiency (G6PDd) are at risk for developing hemolytic anemia when given the antimalarial drug primaquine (PQ). The WHO Evidence Review Group released a report suggesting that mass administration of a single dose of PQ at 0.25 mg of base/kg of body weight (mpk) (mouse equivalent of 3.125 mpk) could potentially reduce malaria transmission based on its gametocytocidal activity and could be safely administered to G6PD-deficient individuals, but there are limited safety data available confirming the optimum single dose of PQ. A single-dose administration of PQ was therefore assessed in our huRBC-SCID mouse model used to predict hemolytic toxicity with respect to G6PD deficiency. In this model, nonobese diabetic (NOD)/SCID mice are engrafted with human red blood cells (huRBC) from donors with the African or Mediterranean variant of G6PDd (A-G6PDd or Med-G6PDd, respectively) and demonstrate dose-dependent sensitivity to PQ. In mice engrafted with A-G6PD-deficient huRBC, single-dose PQ at 3.125, 6.25, or 12.5 mpk had no significant loss of huRBC compared to the vehicle control group. In contrast, in mice engrafted with Med-G6PDd huRBC, a single dose of PQ at 3.125, 6.25, or 12.5 mpk resulted in a significant, dose-dependent loss of huRBC compared to the value for the vehicle control group. Our data suggest that administration of a single low dose of 0.25 mpk of PQ could induce hemolytic anemia in Med-G6PDd individuals but that use of single-dose PQ at 0.25 mpk as a gametocytocidal drug to block transmission would be safe in areas where A-G6PDd predominates.


Antimalarials/administration & dosage , Glucosephosphate Dehydrogenase Deficiency/parasitology , Malaria/transmission , Primaquine/administration & dosage , Animals , Antimalarials/pharmacokinetics , Disease Models, Animal , Erythrocyte Transfusion , Erythrocytes/drug effects , Humans , Mice, SCID , Primaquine/analogs & derivatives , Primaquine/pharmacokinetics
18.
Malar J ; 15: 224, 2016 Apr 19.
Article En | MEDLINE | ID: mdl-27093859

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.


Hepatocytes/metabolism , Primaquine/analogs & derivatives , Primaquine/metabolism , Carbon Isotopes/analysis , Chromatography, High Pressure Liquid , Half-Life , Humans , Kinetics , Stereoisomerism , Tandem Mass Spectrometry
19.
Fundam Clin Pharmacol ; 30(1): 58-69, 2016 Feb.
Article En | MEDLINE | ID: mdl-26501210

New primaquine (PQ) urea and semicarbazide derivatives 1-4 were screened for the first time for central nervous system (CNS) and antimalarial activity. Behavioural tests were performed on mice. In vitro cytotoxicity on L-6 cells and activity against erythrocytic stages of Plasmodium falciparum was determined. Compound 4 inhibited 'head-twitch' responses and decreased body temperature of mice, which suggests some involvement of the serotonergic system. Compound 4 protected mice against clonic seizures and was superior in the antimalarial test. A hybrid of two PQ urea 2 showed a strong antimalarial activity, confirming the previous findings of the high activity of bis(8-aminoquinolines) and other bisantimalarial drugs. All the compounds decreased the locomotor activity of mice, what suggests their weak depressive effects on the CNS, while PQ derivatives 1 and 2 increased amphetamine-induced hyperactivity. None of the compounds impaired coordination, what suggests a lack of their neurotoxicity. All the tested compounds presented an antinociceptive activity in the 'writhing' test. Compounds 3 and 4 were active in nociceptive tests, and those effects were reversed by naloxone. Compound 4 could be a useful lead compound in the development of CNS active agents and antimalarials, whereas compound 3 may be considered as the most promising lead for new antinociceptive agents.


Antimalarials/pharmacology , Central Nervous System/drug effects , Plasmodium falciparum/drug effects , Primaquine/pharmacology , Semicarbazides/pharmacology , Urea/pharmacology , Animals , Antimalarials/chemistry , Behavior, Animal/drug effects , Cell Survival/drug effects , Cells, Cultured , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Male , Mice , Molecular Structure , Myoblasts/drug effects , Primaquine/analogs & derivatives , Primaquine/chemistry , Semicarbazides/chemistry , Toxicity Tests, Acute , Urea/analogs & derivatives , Urea/chemistry
20.
J Mol Graph Model ; 62: 235-244, 2015 Nov.
Article En | MEDLINE | ID: mdl-26521207

Primaquine is a traditional antimalarial drug with low parasitic resistance and generally good acceptance at higher doses, which has been used for over 60 years in malaria treatment. However, several limitations related to its hematotoxicity have been reported. It is believed that this toxicity comes from the hydroxylation of the C-5 and C-6 positions of its 8-aminoquinoline ring before binding to the molecular target: the quinone reductase II (NQO2) human protein. In this study we propose primaquine derivatives, with substitution at position C-6 of the 8-aminoquinoline ring, planned to have better binding to NQO2, compared to primaquine, but with a reduced toxicity related to the C-5 position being possible to be oxidized. On this sense the proposed analogues were suggested in order to reduce or inhibit hydroxylation and further oxidation to hemotoxic metabolites. Five C-6 substituted primaquine analogues were selected by de novo design and further submitted to docking and molecular dynamics simulations. Our results suggest that all analogues bind better to NQO2 than primaquine and may become better antimalarials. However, the analogues 3 and 4 are predicted to have a better activity/toxicity balance.


Enzyme Inhibitors/chemistry , Primaquine/analogs & derivatives , Primaquine/chemistry , Quinone Reductases/chemistry , Catalytic Domain , Humans , Hydrogen Bonding , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Structure, Secondary , Quinone Reductases/antagonists & inhibitors , Thermodynamics
...