Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.389
Filtrer
1.
BMC Pharmacol Toxicol ; 25(1): 38, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38978151

RÉSUMÉ

BACKGROUND: Dihydroartemisinin-piperaquine (DHP) recently showed superior effectiveness over sulfadoxine-pyrimethamine for malaria intermittent preventive treatment in pregnancy (IPTp). We investigated day 7 piperaquine pharmacokinetics and its therapeutic efficacy in preventing malaria during pregnancy. METHODS: Malaria-free (mRDT) pregnant women (n = 400) who received monthly IPTp-DHP were enrolled and followed till delivery. Day 7 Plasma piperaquine concentrations were determined after each IPTp dose using UPLC/MS/MS. IPTp outcomes (symptomatic malaria and parasitemia during pregnancy, placental malaria, and maternal malaria at delivery) were monitored. Linear mixed model and Cox regression were used to assess predictors of day 7 piperaquine concentration and treatment outcome, respectively. RESULTS: The incidences of symptomatic malaria and parasitemia during pregnancy per 100 person-year at risk were 2 and 33, respectively. The prevalence of histopathologically confirmed placental malaria and maternal malaria at delivery were 3% and 9.8%, respectively. Repeated monthly IPTp-DHP resulted in significantly increased day 7 plasma piperaquine concentration (p < 0.001). Following the 1st, 2nd, and 3rd monthly IPTp-DHP doses, the proportions of women with day 7 piperaquine concentration below the therapeutic threshold (< 30 ng/mL) were 6.1%, 4.1% and 3.6%, respectively. Factors such as maternal age, body weight and trimester were not significant predictors of day 7 piperaquine concentration. However, having a low day 7 piperaquine plasma concentration (< 30 ng/mL) was significantly associated with a higher risk of parasitemia during pregnancy (p = 0.004). CONCLUSION: Lower day 7 piperaquine plasma concentration is a risk factor for parasitemia during pregnancy. Single plasma sampling at day 7 can be used to monitor piperaquine effectiveness during IPTp-DHP. TRIAL REGISTRATION: Registered 09/12/2016, PACTR201612001901313.


Sujet(s)
Antipaludiques , Paludisme , Complications parasitaires de la grossesse , Quinoléines , Humains , Femelle , Grossesse , Quinoléines/pharmacocinétique , Quinoléines/sang , Quinoléines/usage thérapeutique , Quinoléines/administration et posologie , Antipaludiques/pharmacocinétique , Antipaludiques/usage thérapeutique , Antipaludiques/sang , Antipaludiques/administration et posologie , Adulte , Complications parasitaires de la grossesse/prévention et contrôle , Complications parasitaires de la grossesse/sang , Jeune adulte , Paludisme/prévention et contrôle , Paludisme/traitement médicamenteux , Artémisinines/pharmacocinétique , Artémisinines/usage thérapeutique , Artémisinines/administration et posologie , Artémisinines/sang , Parasitémie/sang , Parasitémie/prévention et contrôle , Résultat thérapeutique , Association médicamenteuse , Adolescent , Pipérazines
2.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000107

RÉSUMÉ

Even though several new targets (mostly viral infection) for drug repurposing of pyronaridine and artesunate have recently emerged in vitro and in vivo, inter-species pharmacokinetic (PK) data that can extend nonclinical efficacy to humans has not been reported over 30 years of usage. Since extrapolation of animal PK data to those of humans is essential to predict clinical outcomes for drug repurposing, this study aimed to investigate inter-species PK differences in three animal species (hamster, rat, and dog) and to support clinical translation of a fixed-dose combination of pyronaridine and artesunate. PK parameters (e.g., steady-state volume of distribution (Vss), clearance (CL), area under the concentration-time curve (AUC), mean residence time (MRT), etc.) of pyronaridine, artesunate, and dihydroartemisinin (an active metabolite of artesunate) were determined by non-compartmental analysis. In addition, one- or two-compartment PK modeling was performed to support inter-species scaling. The PK models appropriately described the blood concentrations of pyronaridine, artesunate, and dihydroartemisinin in all animal species, and the estimated PK parameters in three species were integrated for inter-species allometric scaling to predict human PKs. The simple allometric equation (Y = a × Wb) well explained the relationship between PK parameters and the actual body weight of animal species. The results from the study could be used as a basis for drug repurposing and support determining the effective dosage regimen for new indications based on in vitro/in vivo efficacy data and predicted human PKs in initial clinical trials.


Sujet(s)
Artémisinines , Artésunate , Repositionnement des médicaments , Naphtyridines , Artésunate/pharmacocinétique , Artésunate/pharmacologie , Repositionnement des médicaments/méthodes , Animaux , Rats , Chiens , Naphtyridines/pharmacocinétique , Naphtyridines/pharmacologie , Artémisinines/pharmacocinétique , Spécificité d'espèce , Humains , Modèles biologiques , Mâle , Antipaludiques/pharmacocinétique , Antipaludiques/pharmacologie
3.
Clin Transl Sci ; 17(7): e13865, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39020517

RÉSUMÉ

The urgent need for safe, efficacious, and accessible drug treatments to treat coronavirus disease 2019 (COVID-19) prompted a global effort to evaluate drug repurposing opportunities. Pyronaridine and amodiaquine are both components of approved antimalarials with in vitro activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In vitro activity does not always translate to clinical efficacy across a therapeutic dose range. This study applied available, verified, physiologically based pharmacokinetic (PBPK) models for pyronaridine, amodiaquine, and its active metabolite N-desethylamodiaquine (DEAQ) to predict drug concentrations in lung tissue relative to plasma or blood in the default healthy virtual population. Lung exposures were compared to published data across the reported range of in vitro EC50 values against SARS-CoV-2. In the multicompartment permeability-limited PBPK model, the predicted total Cmax in lung mass for pyronaridine was 34.2 µM on Day 3, 30.5-fold greater than in blood (1.12 µM) and for amodiaquine was 0.530 µM, 8.83-fold greater than in plasma (0.060 µM). In the perfusion-limited PBPK model, the DEAQ predicted total Cmax on Day 3 in lung mass (30.2 µM) was 21.4-fold greater than for plasma (1.41 µM). Based on the available in vitro data, predicted drug concentrations in lung tissue for pyronaridine and DEAQ, but not amodiaquine, appeared sufficient to inhibit SARS-CoV-2 replication. Simulations indicated standard dosing regimens of pyronaridine-artesunate and artesunate-amodiaquine have potential to treat COVID-19. These findings informed repurposing strategies to select the most relevant compounds for clinical investigation in COVID-19. Clinical data for model verification may become available from ongoing clinical studies.


Sujet(s)
Amodiaquine , Antipaludiques , Traitements médicamenteux de la COVID-19 , Repositionnement des médicaments , Poumon , SARS-CoV-2 , Humains , Antipaludiques/pharmacocinétique , Antipaludiques/administration et posologie , Amodiaquine/pharmacocinétique , Amodiaquine/administration et posologie , Amodiaquine/analogues et dérivés , SARS-CoV-2/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Naphtyridines/pharmacocinétique , Naphtyridines/administration et posologie , Naphtyridines/pharmacologie , Modèles biologiques , COVID-19/virologie , Antiviraux/pharmacocinétique , Antiviraux/administration et posologie , Simulation numérique
4.
ACS Infect Dis ; 10(7): 2419-2442, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38862127

RÉSUMÉ

ELQ-300 is a potent antimalarial drug with activity against blood, liver, and vector stages of the disease. A prodrug, ELQ-331, exhibits reduced crystallinity and improved in vivo efficacy in preclinical testing, and currently, it is in the developmental pipeline for once-a-week dosing for oral prophylaxis against malaria. Because of the high cost of developing a new drug for human use and the high risk of drug failure, it is prudent to have a back-up plan in place. Here we describe ELQ-596, a member of a new subseries of 3-biaryl-ELQs, with enhanced potency in vitro against multidrug-resistant Plasmodium falciparum parasites. ELQ-598, a prodrug of ELQ-596 with diminished crystallinity, is more effective vs murine malaria than its progenitor ELQ-331 by 4- to 10-fold, suggesting that correspondingly lower doses could be used to protect and cure humans of malaria. With a longer bloodstream half-life in mice compared to its progenitor, ELQ-596 highlights a novel series of next-generation ELQs with the potential for once-monthly dosing for protection against malaria infection. Advances in the preparation of 3-biaryl-ELQs are presented along with preliminary results from experiments to explore key structure-activity relationships for drug potency, selectivity, pharmacokinetics, and safety.


Sujet(s)
Antipaludiques , Plasmodium falciparum , Quinolinone , Antipaludiques/pharmacologie , Antipaludiques/composition chimique , Antipaludiques/pharmacocinétique , Animaux , Plasmodium falciparum/effets des médicaments et des substances chimiques , Souris , Quinolinone/pharmacologie , Quinolinone/composition chimique , Quinolinone/pharmacocinétique , Paludisme/traitement médicamenteux , Paludisme/prévention et contrôle , Humains , Promédicaments/pharmacologie , Promédicaments/composition chimique , Promédicaments/pharmacocinétique , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/prévention et contrôle , Femelle , Relation structure-activité
5.
Antimicrob Agents Chemother ; 68(7): e0033824, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38837364

RÉSUMÉ

The human malaria-Aotus monkey model has served the malaria research community since its inception in 1966 at the Gorgas Memorial Laboratory (GML) in Panama. Spanning over five decades, this model has been instrumental in evaluating the in vivo efficacy and pharmacokinetics of a wide array of candidate antimalarial drugs, whether used singly or in combination. The animal model could be infected with drug-resistant and susceptible Plasmodium falciparum and Plasmodium vivax strains that follow a characteristic and reproducible course of infection, remarkably like human untreated and treated infections. Over the years, the model has enabled the evaluation of several synthetic and semisynthetic endoperoxides, for instance, artelinic acid, artesunate, artemether, arteether, and artemisone. These compounds have been evaluated alone and in combination with long-acting partner drugs, commonly referred to as artemisinin-based combination therapies, which are recommended as first-line treatment against uncomplicated malaria. Further, the model has also supported the evaluation of the primaquine analog tafenoquine against blood stages of P. vivax, contributing to its progression to clinical trials and eventual approval. Besides, the P. falciparum/Aotus model at GML has also played a pivotal role in exploring the biology, immunology, and pathogenesis of malaria and in the characterization of drug-resistant P. falciparum and P. vivax strains. This minireview offers a historical overview of the most significant contributions made by the Panamanian owl monkey (Aotus lemurinus lemurinus) to malaria chemotherapy research.


Sujet(s)
Antipaludiques , Artémisinines , Modèles animaux de maladie humaine , Animaux , Antipaludiques/usage thérapeutique , Antipaludiques/pharmacocinétique , Antipaludiques/pharmacologie , Artémisinines/usage thérapeutique , Artémisinines/pharmacologie , Humains , Panama , Aotidae , Plasmodium falciparum/effets des médicaments et des substances chimiques , Paludisme/traitement médicamenteux , Plasmodium vivax/effets des médicaments et des substances chimiques , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/parasitologie , Artésunate/usage thérapeutique , Artésunate/pharmacologie , Artésunate/pharmacocinétique , Paludisme à Plasmodium vivax/traitement médicamenteux , Paludisme à Plasmodium vivax/parasitologie , Histoire du 20ème siècle , Aminoquinoléines
6.
Malar J ; 23(1): 176, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38840151

RÉSUMÉ

BACKGROUND: With only one 15 mg primaquine tablet registered by a stringent regulatory authority and marketed, more quality-assured primaquine is needed to meet the demands of malaria elimination. METHODS: A classic, two sequence, crossover study, with a 10-day wash out period, of 15 mg of IPCA-produced test primaquine tablets and 15 mg of Sanofi reference primaquine tablets was conducted. Healthy volunteers, aged 18-45 years, without glucose-6-phosphate dehydrogenase deficiency, a baseline haemoglobin ≥ 11 g/dL, creatinine clearance ≥ 70 mL/min/1.73 ms, and body mass index of 18.5-30 kg/m2 were randomized to either test or reference primaquine, administered on an empty stomach with 240 mL of water. Plasma primaquine and carboxyprimaquine concentrations were measured at baseline, then 0.25, 0.5, 0.75, 1.0, 1.25, 1.5, 1.75, 2.0, 2.333, 2.667, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 8.0, 10.0, 12.0, 16.0, 24.0, 36.0, 48.0 and 72.0 h by liquid chromatography coupled to tandem mass spectrometry. Primaquine pharmacokinetic profiles were evaluated by non-compartmental analysis and bioequivalence concluded if the 90% confidence intervals (CI) of geometric mean (GM) ratios of test vs. reference formulation for the peak concentrations (Cmax) and area under the drug concentration-time (AUC0-t) were within 80.00 to 125.00%. RESULTS: 47 of 50 volunteers, median age 33 years, completed both dosing rounds and were included in the bioequivalence analysis. For primaquine, GM Cmax values for test and reference formulations were 62.12 vs. 59.63 ng/mL, resulting in a GM ratio (90% CI) of 104.17% (96.92-111.96%); the corresponding GM AUC0-t values were 596.56 vs. 564.09 ngxh/mL, for a GM ratio of 105.76% (99.76-112.08%). Intra-subject coefficient of variation was 20.99% for Cmax and 16.83% for AUC0-t. Median clearances and volumes of distribution were similar between the test and reference products: 24.6 vs. 25.2 L/h, 189.4 vs. 191.0 L, whilst the median half-lives were the same, 5.2 h. CONCLUSION: IPCA primaquine was bioequivalent to the Sanofi primaquine. This opens the door to prequalification, registration in malaria endemic countries, and programmatic use for malaria elimination. Trial registration The trial registration reference is ISRCTN 54640699.


Sujet(s)
Antipaludiques , Études croisées , Primaquine , Équivalence thérapeutique , Primaquine/pharmacocinétique , Primaquine/administration et posologie , Humains , Antipaludiques/pharmacocinétique , Antipaludiques/administration et posologie , Adulte , Jeune adulte , Mâle , Femelle , Adolescent , Adulte d'âge moyen , Paludisme/traitement médicamenteux , Paludisme/prévention et contrôle , Volontaires sains , Comprimés
7.
Int J Antimicrob Agents ; 64(1): 107196, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38734217

RÉSUMÉ

With the spread of artemisinin resistance throughout Southeast Asia and now in Africa, the antimalarial drug pyronaridine is likely to become an increasingly important component of new antimalarial drug regimens. However, the antimalarial activity of pyronaridine in humans has not been completely characterised. This volunteer infection study aimed to determine the pharmacokinetic/pharmacodynamic (PK/PD) relationship of pyronaridine in malaria naïve adults. Volunteers were inoculated with Plasmodium falciparum-infected erythrocytes on day 0 and administered different single oral doses of pyronaridine on day 8. Parasitaemia and concentrations of pyronaridine were measured and standard safety assessments performed. Curative artemether-lumefantrine therapy was administered if parasite regrowth occurred, or on day 47 ± 2. Outcomes were parasite clearance kinetics, PK and PK/PD parameters from modelling. Ten participants were inoculated and administered 360 mg (n = 4), 540 mg (n = 4) or 720 mg (n = 1) pyronaridine. One participant was withdrawn without receiving pyronaridine. The time to maximum pyronaridine concentration was 1-2 h, the elimination half-life was 8-9 d, and the parasite clearance half-life was approximately 5 h. Parasite regrowth occurred with 360 mg (4/4 participants) and 540 mg (2/4 participants). Key efficacy parameters including the minimum inhibitory concentration (5.5 ng/mL) and minimum parasiticidal concentration leading to 90% of maximum effect (MPC90: 8 ng/mL) were derived from the PK/PD model. Adverse events considered related to pyronaridine were predominantly mild to moderate gastrointestinal symptoms. There were no serious adverse events. Data obtained in this study will support the use of pyronaridine in new antimalarial combination therapies by informing partner drug selection and dosing considerations.


Sujet(s)
Antipaludiques , Volontaires sains , Paludisme à Plasmodium falciparum , Naphtyridines , Parasitémie , Plasmodium falciparum , Humains , Antipaludiques/pharmacocinétique , Antipaludiques/usage thérapeutique , Antipaludiques/pharmacologie , Antipaludiques/administration et posologie , Naphtyridines/pharmacocinétique , Naphtyridines/usage thérapeutique , Naphtyridines/pharmacologie , Naphtyridines/administration et posologie , Plasmodium falciparum/effets des médicaments et des substances chimiques , Adulte , Mâle , Jeune adulte , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/parasitologie , Femelle , Parasitémie/traitement médicamenteux , Parasitémie/parasitologie , Érythrocytes/effets des médicaments et des substances chimiques , Érythrocytes/parasitologie , Administration par voie orale , Adulte d'âge moyen , Résultat thérapeutique
8.
Int J Antimicrob Agents ; 64(1): 107209, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38761871

RÉSUMÉ

OBJECTIVES: Malaria-induced alteration of physiological parameters and pharmacokinetic properties of antimalarial drugs may be clinically relevant. Whether and how malaria alters the disposition of piperaquine (PQ) was investigated in this study. METHODS: The effect of malaria on drug metabolism-related enzymes and PQ pharmacokinetic profiles was studied in Plasmodium yoelii-infected mice in vitro/in vivo. Whether the malaria effect was clinically relevant for PQ was evaluated using a validated physiologically-based pharmacokinetic model with malaria-specific scalars obtained in mice. RESULTS: The infection led to a higher blood-to-plasma partitioning (Rbp) for PQ, which was concentration-dependent and correlated to parasitemia. No significant change in plasma protein binding was found for PQ. Drug metabolism-related genes (CYPs/UDP-glucuronosyltransferase/nuclear receptor, except for CYP2a5) were downregulated in infected mice, especially at the acute phase. The plasma oral clearances (CL/F) of three probe substrates for CYP enzymes were significantly decreased (by ≥35.9%) in mice even with moderate infection. The validated physiologically-based pharmacokinetic model indicated that the hepatic clearance (CLH) of PQ was the determinant of its simulated CL/F, which was predicted to slightly decrease (by ≤23.6%) in severely infected mice but not in malaria patients. The result fitted well with the plasma pharmacokinetics of PQ in infected mice and literature data on malaria patients. The blood clearance of PQ was much lower than its plasma clearance due to its high Rbp. CONCLUSIONS: The malaria-induced alteration of drug metabolism was substrate-dependent, and its impact on the disposition of PQ and maybe other long-acting aminoquinoline antimalarials was not expected to be clinically relevant.


Sujet(s)
Antipaludiques , Modèles animaux de maladie humaine , Paludisme , Plasmodium yoelii , Quinoléines , Animaux , Quinoléines/pharmacocinétique , Paludisme/traitement médicamenteux , Paludisme/parasitologie , Plasmodium yoelii/effets des médicaments et des substances chimiques , Antipaludiques/pharmacocinétique , Antipaludiques/usage thérapeutique , Humains , Souris , Femelle , Parasitémie/traitement médicamenteux , Mâle , Pipérazines
9.
Eur J Pharm Sci ; 198: 106795, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38729224

RÉSUMÉ

The overarching premise of this investigation is that injectable, long-acting antimalarial medication would encourage adherence to a dosage regimen for populations at risk of contracting the disease. To advance support for this goal, we have developed oil-based formulations of ELQ-331 (a prodrug of ELQ-300) that perform as long-acting, injectable chemoprophylactics with drug loading as high as 160 mg/ml of ELQ-331. In a pharmacokinetic study performed with rats, a single intramuscular injection of 12.14 mg/kg maintained higher plasma levels than the previously established minimum fully protective plasma concentration (33.25 ng/ml) of ELQ-300 for more than 4 weeks. The formulations were well tolerated by the rats and the tested dose produced no adverse reactions. We believe that by extending the length of time between subsequent injections, these injectable oil-based solutions of ELQ-331 can offer a more accessible, low-cost option for long-acting disease prevention and reduced transmission in malaria-endemic regions and may also be of use to travelers.


Sujet(s)
Antipaludiques , Animaux , Antipaludiques/administration et posologie , Antipaludiques/pharmacocinétique , Injections musculaires , Mâle , Rats , Rat Sprague-Dawley , Préparations à action retardée/administration et posologie , Promédicaments/administration et posologie , Promédicaments/pharmacocinétique , Paludisme/traitement médicamenteux
10.
Malar J ; 23(1): 159, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773528

RÉSUMÉ

BACKGROUND: Primaquine (PQ) is the prototype 8-aminoquinoline drug, a class which targets gametocytes and hypnozoites. The World Health Organization (WHO) recommends adding a single low dose of primaquine to the standard artemisinin-based combination therapy (ACT) in order to block malaria transmission in regions with low malaria transmission. However, the haemolytic toxicity is a major adverse outcome of primaquine in glucose-6-phosphate dehydrogenase (G6PD)-deficient subjects. This study aimed to characterize the pharmacokinetic properties of primaquine and its major metabolites in G6PD-deficient subjects. METHODS: A single low-dose of primaquine (0.4-0.5 mg/kg) was administered in twenty-eight African males. Venous and capillary plasma were sampled up to 24 h after the drug administration. Haemoglobin levels were observed up to 28 days after drug administration. Only PQ, carboxy-primaquine (CPQ), and primaquine carbamoyl-glucuronide (PQCG) were present in plasma samples and measured using liquid chromatography mass spectrometry. Drug and metabolites' pharmacokinetic properties were investigated using nonlinear mixed-effects modelling. RESULTS: Population pharmacokinetic properties of PQ, CPQ, and PQCG can be described by one-compartment disposition kinetics with a transit-absorption model. Body weight was implemented as an allometric function on the clearance and volume parameters for all compounds. None of the covariates significantly affected the pharmacokinetic parameters. No significant correlations were detected between the exposures of the measured compounds and the change in haemoglobin or methaemoglobin levels. There was no significant haemoglobin drop in the G6PD-deficient patients after administration of a single low dose of PQ. CONCLUSIONS: A single low-dose of PQ was haematologically safe in this population of G6PD-normal and G6PD-deficient African males without malaria. Trial registration NCT02535767.


Sujet(s)
Antipaludiques , Déficit en glucose-6-phosphate-déshydrogénase , Primaquine , Adolescent , Adulte , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Antipaludiques/pharmacocinétique , Antipaludiques/sang , Antipaludiques/administration et posologie , Primaquine/pharmacocinétique , Primaquine/sang , Primaquine/administration et posologie
11.
Nat Commun ; 15(1): 3851, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38719803

RÉSUMÉ

Current guidelines advise against primaquine treatment for breastfeeding mothers to avoid the potential for haemolysis in infants with G6PD deficiency. To predict the haemolytic risk, the amount of drug received from the breast milk and the resulting infant drug exposure need to be characterised. Here, we develop a pharmacokinetic model to describe the drug concentrations in breastfeeding women using venous, capillary, and breast milk data. A mother-to-infant model is developed to mimic the infant feeding pattern and used to predict their drug exposures. Primaquine and carboxyprimaquine exposures in infants are <1% of the exposure in mothers. Therefore, even in infants with the most severe G6PD deficiency variants, it is highly unlikely that standard doses of primaquine (0.25-1 mg base/kg once daily given to the mother for 1-14 days) would cause significant haemolysis. After the neonatal period, primaquine should not be restricted for breastfeeding women (Clinical Trials Registration: NCT01780753).


Sujet(s)
Antipaludiques , Allaitement naturel , Lactation , Lait humain , Primaquine , Humains , Femelle , Primaquine/pharmacocinétique , Primaquine/administration et posologie , Antipaludiques/pharmacocinétique , Antipaludiques/administration et posologie , Nourrisson , Lait humain/composition chimique , Lait humain/métabolisme , Adulte , Nouveau-né , Hémolyse/effets des médicaments et des substances chimiques , Modèles biologiques
12.
Int J Pharm ; 658: 124204, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38710297

RÉSUMÉ

Pulsatile drug delivery is hardly achieved by conventional gastro-retentive dosage forms. Artesunate as a typical anti-malaria medicine needs oral pulsatile release. Here, artesunate-loaded pulsatile-release multi-unit gastro-retentive tablets (APGTs) were prepared with a semi-solid extrusion three-dimensional (3D) printing method. An APGT was composed of three units: artesunate-loaded immediate and delayed release units and a block unit. The matrix of the immediate/delayed release units consisted of polyvinylpyrrolidone (PVP) K30 and croscarmellose sodium, which improved the rapid release of artesunate when contacting water. The block unit consisted of octadecanol, hydroxypropyl methyl cellulose K15M, PVP K30, and poloxamer F68. APGTs showed multi-phase release in simulated gastric liquids (SGLs). The first immediate release phase continued for 1 h followed by a long block phase for 7 h. The second rapid release phase was initiated when the eroded holes in the block unit extended to the inner delayed release unit, and this phase continued for about 14 h. Low-density APGTs could ensure their long-term floating in the stomach. Oral APGTs remained in the rabbit stomach for about 20 h. 3D printing provides a new strategy for the preparation of oral pulsatile-release tablets.


Sujet(s)
Antipaludiques , Artésunate , Préparations à action retardée , Libération de médicament , Povidone , Impression tridimensionnelle , Comprimés , Artésunate/administration et posologie , Artésunate/composition chimique , Artésunate/pharmacocinétique , Animaux , Lapins , Antipaludiques/administration et posologie , Antipaludiques/composition chimique , Antipaludiques/pharmacocinétique , Povidone/composition chimique , Dérivés de l'hypromellose/composition chimique , Excipients/composition chimique , Systèmes de délivrance de médicaments , Administration par voie orale , Carboxyméthylcellulose de sodium/composition chimique , Poloxamère/composition chimique , Muqueuse gastrique/métabolisme
13.
Antimicrob Agents Chemother ; 68(5): e0009324, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38597636

RÉSUMÉ

Capillary samples offer practical benefits compared with venous samples for the measurement of drug concentrations, but the relationship between the two measures varies between different drugs. We measured the concentrations of lumefantrine, mefloquine, piperaquine in 270 pairs of venous plasma and concurrent capillary plasma samples collected from 270 pregnant women with uncomplicated falciparum or vivax malaria. The median and range of venous plasma concentrations included in this study were 447.5 ng/mL (8.81-3,370) for lumefantrine (day 7, n = 76, median total dose received 96.0 mg/kg), 17.9 ng/mL (1.72-181) for desbutyl-lumefantrine, 1,885 ng/mL (762-4,830) for mefloquine (days 3-21, n = 90, median total dose 24.9 mg/kg), 641 ng/mL (79.9-1,950) for carboxy-mefloquine, and 51.8 ng/mL (3.57-851) for piperaquine (days 3-21, n = 89, median total dose 52.2 mg/kg). Although venous and capillary plasma concentrations showed a high correlation (Pearson's correlation coefficient: 0.90-0.99) for all antimalarials and their primary metabolites, they were not directly interchangeable. Using the concurrent capillary plasma concentrations and other variables, the proportions of venous plasma samples predicted within a ±10% precision range was 34% (26/76) for lumefantrine, 36% (32/89) for desbutyl-lumefantrine, 74% (67/90) for mefloquine, 82% (74/90) for carboxy-mefloquine, and 24% (21/89) for piperaquine. Venous plasma concentrations of mefloquine, but not lumefantrine and piperaquine, could be predicted by capillary plasma samples with an acceptable level of agreement. Capillary plasma samples can be utilized for pharmacokinetic and clinical studies, but caution surrounding cut-off values is required at the individual level.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT01054248.


Sujet(s)
Antipaludiques , Luméfantrine , Paludisme à Plasmodium falciparum , Paludisme à Plasmodium vivax , Méfloquine , Pipérazines , Quinoléines , Humains , Femelle , Méfloquine/sang , Méfloquine/usage thérapeutique , Méfloquine/pharmacocinétique , Antipaludiques/sang , Antipaludiques/usage thérapeutique , Antipaludiques/pharmacocinétique , Grossesse , Quinoléines/sang , Quinoléines/pharmacocinétique , Quinoléines/usage thérapeutique , Luméfantrine/usage thérapeutique , Luméfantrine/sang , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/sang , Adulte , Paludisme à Plasmodium vivax/traitement médicamenteux , Paludisme à Plasmodium vivax/sang , Jeune adulte , Éthanolamines/sang , Éthanolamines/pharmacocinétique , Éthanolamines/usage thérapeutique , Fluorènes/sang , Fluorènes/usage thérapeutique , Fluorènes/pharmacocinétique , Adolescent
14.
J Pharm Biomed Anal ; 245: 116154, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38657367

RÉSUMÉ

Malaria remains a major health concern, aggravated by emerging resistance of the parasite to existing treatments. The World Health Organization recently endorsed the use of artesunate-pyronaridine to treat uncomplicated malaria. However, there is a lack of clinical pharmacokinetic (PK) data of pyronaridine, particularly in special populations such as children and pregnant women. Existing methods for the quantification of pyronaridine in biological matrices to support PK studies exhibit several drawbacks. These include limited sensitivity, a large sample volume required, and extensive analysis time. To overcome these limitations, an ultra-performance reversed-phase liquid chromatography tandem-mass spectrometry method to determine pyronaridine was developed and validated according to international guidelines. The method enabled fast and accurate quantification of pyronaridine in whole blood across a clinically relevant concentration range of 0.500-500 ng/mL (r2 ≥ 0.9963), with a required sample volume of 50 µL. Pyronaridine was extracted from whole blood using liquid-liquid extraction, effectively eliminating the matrix effect and preventing ion enhancement or suppression. The method achieved a satisfactory reproducible sample preparation recovery of 77%, accuracy (as bias) and precision were within ±8.2% and ≤5.3%, respectively. Stability experiments demonstrated that pyronaridine was stable for up to 315 days when stored at -70°C. Adjustments to the chromatographic system substantially reduced carry-over and improved sensitivity compared to prior methods. The method was successfully applied to quantify pyronaridine in whole blood samples from a selection of pregnant malaria patients participating in the PYRAPREG clinical trial (PACTR202011812241529) in the Democratic Republic of the Congo, demonstrating its suitability to support future PK studies. Furthermore, the enhanced sensitivity allows for the determination of pyronaridine up to 42 days post-treatment initiation, enabling assessment of the terminal elimination half-life.


Sujet(s)
Antipaludiques , Naphtyridines , Spectrométrie de masse en tandem , Humains , Antipaludiques/sang , Antipaludiques/pharmacocinétique , Antipaludiques/analyse , Spectrométrie de masse en tandem/méthodes , Naphtyridines/sang , Naphtyridines/pharmacocinétique , Naphtyridines/analyse , Chromatographie en phase liquide à haute performance/méthodes , Reproductibilité des résultats , Femelle , Extraction liquide-liquide/méthodes , Grossesse , Paludisme/traitement médicamenteux , Paludisme/sang , Chromatographie en phase inverse/méthodes
15.
Malar J ; 23(1): 125, 2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38685044

RÉSUMÉ

BACKGROUND: Despite efforts made to reduce morbidity and mortality associated with malaria, especially in sub-Saharan Africa, malaria continues to be a public health concern that requires innovative efforts to reach the WHO-set zero malaria agenda. Among the innovations is the use of artemisinin-based combination therapy (ACT) that is effective against Plasmodium falciparum. Generic artemether-lumefantrine (AL) is used to treat uncomplicated malaria after appropriate diagnosis. AL is metabolized by the cytochrome P450 family of enzymes, such as CYP2B6, CYP3A4 and CYP3A5, which can be under pharmacogenetic influence. Pharmacogenetics affecting AL metabolism, significantly influence the overall anti-malarial activity leading to variable therapeutic efficacy. This study focused on generic AL drugs used in malarial treatment as prescribed at health facilities and evaluated pharmacogenomic influences on their efficacy. METHODS: Patients who have been diagnosed with malaria and confirmed through RDT and microscopy were recruited in this study. Blood samples were taken on days 1, 2, 3 and 7 for parasite count and blood levels of lumefantrine, artemisinin, desbutyl-lumefantrine (DBL), and dihydroartemisinin (DHA), the active metabolites of lumefantrine and artemether, respectively, were analysed using established methods. Pharmacogene variation analysis was undertaken using iPLEX microarray and PCR-RFLP. RESULTS: A total of 52 patients completed the study. Median parasite density from day 1 to 7 ranged from 0-2666/µL of blood, with days 3 and 7 recording 0 parasite density. Highest median plasma concentration for lumefantrine and desbutyl lumefantrine, which are the long-acting components of artemisinin-based combinations, was 4123.75 ng/mL and 35.87 ng/mL, respectively. Day 7 plasma lumefantrine concentration across all generic ACT brands was ≥ 200 ng/mL which potentially accounted for the parasitaemia profile observed. Monomorphism was observed for CYP3A4 variants, while there were observed variations in CYP2B6 and CYP3A5 alleles. Among the CYP3A5 genotypes, significant differences in genotypes and plasma concentration for DBL were seen on day 3 between 1/*1 versus *1/*6 (p = 0.002), *1/*3 versus *1/*6 (p = 0.006) and *1/*7 versus *1/*6 (p = 0.008). Day 7 plasma DBL concentrations showed a significant difference between *1/*6 and *1/*3 (p = 0.026) expressors. CONCLUSIONS: The study findings show that CYP2B6 and CYP3A5 pharmacogenetic variations may lead to higher plasma exposure of AL metabolites.


Sujet(s)
Antipaludiques , Association d'artéméther et de luméfantrine , Artémisinines , Association médicamenteuse , Éthanolamines , Fluorènes , Humains , Antipaludiques/usage thérapeutique , Antipaludiques/pharmacocinétique , Association d'artéméther et de luméfantrine/usage thérapeutique , Femelle , Éthanolamines/usage thérapeutique , Éthanolamines/pharmacocinétique , Adolescent , Fluorènes/usage thérapeutique , Fluorènes/pharmacocinétique , Fluorènes/pharmacologie , Artémisinines/usage thérapeutique , Artémisinines/pharmacocinétique , Mâle , Ghana , Adulte , Jeune adulte , Enfant , Enfant d'âge préscolaire , Adulte d'âge moyen , Paludisme à Plasmodium falciparum/traitement médicamenteux , Paludisme à Plasmodium falciparum/parasitologie , Médicaments génériques/usage thérapeutique , Résultat thérapeutique , Pharmacogénétique , Sujet âgé , Nourrisson
16.
CPT Pharmacometrics Syst Pharmacol ; 13(5): 795-811, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38528724

RÉSUMÉ

We reported here on the development of a pharmacometric framework to assess patient adherence, by using two population-based approaches - the percentile and the Bayesian method. Three different dosing strategies were investigated in patients prescribed a total of three doses; (1) non-observed therapy, (2) directly observed administration of the first dose, and (3) directly observed administration of the first two doses. The percentile approach used population-based simulations to derive optimal concentration percentile cutoff values from the distribution of simulated drug concentrations at a specific time. This was done for each adherence scenario and compared to full adherence. The Bayesian approach calculated the posterior probability of each adherence scenario at a given drug concentration. The predictive performance (i.e., Youden index, receiver operating characteristic [ROC] curve) of both approaches were highly influenced by sample collection time (early was better) and interindividual variability (smaller was better). The complexity of the structural model and the half-life had a minimal impact on the predictive performance of these methods. The impact of the assay limitation (LOQ) on the predictive performance was relatively small if the fraction of LOQ data was less than 20%. Overall, the percentile method performed similar or better for adherence predictions compared to the Bayesian approach, with the latter showing slightly better results when investigating the adherence to the last dose only. The percentile approach showed acceptable adherence predictions (area under ROC curve > 0.74) when sampling the antimalarial drugs piperaquine at day 7 postdose and lumefantrine at day 3 postdose (i.e., 12 h after the last dose). This could be a highly useful approach when evaluating programmatic implementations of preventive and curative antimalarial treatment programs in endemic areas.


Sujet(s)
Antipaludiques , Théorème de Bayes , Adhésion au traitement médicamenteux , Humains , Antipaludiques/pharmacocinétique , Antipaludiques/administration et posologie , Adhésion au traitement médicamenteux/statistiques et données numériques , Paludisme/traitement médicamenteux , Femelle , Mâle , Adulte , Simulation numérique , Adulte d'âge moyen , Courbe ROC
17.
Antimicrob Agents Chemother ; 68(5): e0091523, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38517190

RÉSUMÉ

Primaquine is the mainstream antimalarial drug to prevent Plasmodium vivax relapses. However, this drug can induce hemolysis in patients with glucose-6-phosphate dehydrogenase deficiency. Nanostructure formulations of primaquine loaded with D-galactose were used as a strategy to target the drug to the liver and decrease the hemolytic risks. Nanoemulsion (NE-Pq) and nanochitosan (NQ-Pq) formulations of primaquine diphosphate containing D-galactose were prepared and characterized by their physicochemistry properties. Pharmacokinetic and biodistribution studies were conducted using Swiss Webster mice. A single dose of 10 mg/kg of each nanoformulation or free primaquine solution was administered by gavage to the animals, which were killed at 0.5, 1, 2, 4, 8, and 24 hours. Blood samples and tissues were collected, processed, and analyzed by high-performance liquid chromatography. The nanoformulation showed sizes around 200 nm (NE-Pq) and 400 nm (NQ-Pq) and physicochemical stability for over 30 days. Free primaquine solution achieved higher primaquine Cmax in the liver than NE-Pq or NQ-Pq at 0.5 hours. However, the half-life and mean residence time (MRT) of primaquine in the liver were three times higher with the NQ-Pq formulation than with free primaquine, and the volume distribution was four times higher. Conversely, primaquine's half-life, MRT, and volume distribution in the plasma were lower for NQ-Pq than for free primaquine. NE-Pq, on the other hand, accumulated more in the lungs but not in the liver. Galactose-coated primaquine nanochitosan formulation showed increased drug targeting to the liver compared to free primaquine and may represent a promising strategy for a more efficient and safer radical cure for vivax malaria.


Sujet(s)
Antipaludiques , Chitosane , Galactose , Foie , Primaquine , Primaquine/pharmacocinétique , Primaquine/composition chimique , Animaux , Souris , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Galactose/composition chimique , Chitosane/composition chimique , Antipaludiques/pharmacocinétique , Nanoparticules/composition chimique , Distribution tissulaire , Nanostructures/composition chimique , Mâle
18.
Biomed Chromatogr ; 38(5): e5844, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38326977

RÉSUMÉ

As first-line antimalarials used in the artemisinin combination therapy, artemisinin drugs exert their action inside red blood cells. However, the blood pharmacokinetic characteristics of artemisinin drugs have not been fully revealed owing to their built-in chemical instability initiated by Fe2+ released from hemoglobin, with limited information on their metabolites. In this study, liquid chromatography tandem high-resolution mass spectrometric (LC-HRMS) methods were developed for the quantification of two representative artemisinin drugs (artemisinin, ART; dihydroartemisinin, DHA) and their respective metabolite (deoxyartemisinin, D-ART; dihydroartemisinin glucuronide, DHA-Glu) in rat blood/plasma. The blood samples were pretreated with the stabilizer (0.4 m potassium dichromate and 3% EDTA-2Na). The methods displayed excellent specificity, linearity, accuracy and precision for ART (17.7-709.2 nm) and its metabolite D-ART (18.8-751.9 nm), and the linear range was 40.0-4,000.0 nm for both DHA and DHA-Glu. The methods were successfully applied to the pharmacokinetic studies of ART and DHA in rats. The blood-to-plasma ratio was 0.8-1.5 for ART, 1.0-1.5 for D-ART, 1.2-2.2 for DHA and 0.9-1.3 for DHA-Glu, which was time dependent. The results indicated that artemisinin drugs and their metabolites showed a high but different blood-to-plasma ratio, which should be considered when optimizating their dosing regimens or evaluating their clinical outcomes.


Sujet(s)
Artémisinines , Rat Sprague-Dawley , Spectrométrie de masse en tandem , Artémisinines/sang , Artémisinines/pharmacocinétique , Animaux , Rats , Reproductibilité des résultats , Mâle , Modèles linéaires , Chromatographie en phase liquide/méthodes , Spectrométrie de masse en tandem/méthodes , Antipaludiques/sang , Antipaludiques/pharmacocinétique , Limite de détection , Sensibilité et spécificité
19.
J Antimicrob Chemother ; 78(10): 2406-2418, 2023 10 03.
Article de Anglais | MEDLINE | ID: mdl-37638690

RÉSUMÉ

Pyronaridine-artesunate was recently strongly recommended in the 2022 update of the WHO Guidelines for the Treatment of Malaria, becoming the newest artemisinin-based combination therapy (ACT) for both uncomplicated Plasmodium falciparum and Plasmodium vivax malaria. Pyronaridine-artesunate, available as a tablet and paediatric granule formulations, is being adopted in regions where malaria treatment outcome is challenged by increasing chloroquine resistance. Pyronaridine is an old antimalarial agent that has been used for more than 50 years as a blood schizonticide, which exerts its antimalarial activity by interfering with the synthesis of the haemozoin pigment within the Plasmodium digestive vacuole. Pyronaridine exhibits a high blood-to-plasma distribution ratio due to its tendency to accumulate in blood cells. This feature is believed to play a crucial role in its pharmacokinetic (PK) properties and pharmacological activity. The PK characteristics of pyronaridine include rapid oral absorption, large volumes of distribution and low total body clearance, resulting in a long terminal apparent half-life. Moreover, differences in PK profiles have been observed between healthy volunteers and malaria-infected patients, indicating a potential disease-related impact on PK properties. Despite a long history, there is only limited knowledge of the clinical PK and pharmacodynamics of pyronaridine, particularly in special populations such as children and pregnant women. We here provide a comprehensive overview of the clinical pharmacology of pyronaridine in the treatment of malaria.


Sujet(s)
Antipaludiques , Paludisme à Plasmodium falciparum , Paludisme , Grossesse , Humains , Enfant , Femelle , Paludisme à Plasmodium falciparum/traitement médicamenteux , Antipaludiques/usage thérapeutique , Antipaludiques/pharmacocinétique , Paludisme/traitement médicamenteux , Naphtyridines/pharmacologie , Naphtyridines/usage thérapeutique , Chloroquine/usage thérapeutique
20.
Macromol Biosci ; 23(5): e2200518, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36999404

RÉSUMÉ

Uncomplicated malaria is effectively treated with oral artemisinin-based combination therapy (ACT). Yet, there is an unmet clinical need for the intravenous treatment of the more fatal severe malaria. There is no combination intravenous therapy for uncomplicated due to the nonavailability of a water-soluble partner drug for the artemisinin, artesunate. The currently available treatment is a two-part regimen split into an intravenous artesunate followed by the conventional oral ACT . In a novel application of polymer therapeutics, the aqueous insoluble antimalarial lumefantrine is conjugated to a carrier polymer to create a new water-soluble chemical entity suitable for intravenous administration in a clinically relevant formulation . The conjugate is characterized by spectroscopic and analytical techniques, and the aqueous solubility of lumefantrine is determined to have increased by three orders of magnitude. Pharmacokinetic studies in mice indicate that there is a significant plasma release of lumefantrine and production its metabolite desbutyl-lumefantrine (area under the curve of metabolite is ≈10% that of the parent). In a Plasmodium falciparum malaria mouse model, parasitemia clearance is 50% higher than that of reference unconjugated lumefantrine. The polymer-lumefantrine shows potential for entering the clinic to meet the need for a one-course combination treatment for severe malaria.


Sujet(s)
Antipaludiques , Luméfantrine , Paludisme , Polymères , Animaux , Souris , Administration par voie intraveineuse , Antipaludiques/administration et posologie , Antipaludiques/synthèse chimique , Antipaludiques/pharmacocinétique , Antipaludiques/usage thérapeutique , Antipaludiques/toxicité , Aire sous la courbe , Modèles animaux de maladie humaine , Association médicamenteuse , Luméfantrine/administration et posologie , Luméfantrine/analogues et dérivés , Luméfantrine/synthèse chimique , Luméfantrine/pharmacocinétique , Luméfantrine/usage thérapeutique , Luméfantrine/toxicité , Paludisme/traitement médicamenteux , Souris de lignée BALB C , Parasitémie , Plasmodium falciparum , Polymères/composition chimique , Polymères/pharmacologie , Polymères/usage thérapeutique , Solubilité , Eau/composition chimique , Mâle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE