Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Mol Cancer Ther ; 20(10): 1893-1903, 2021 10.
Article in English | MEDLINE | ID: mdl-34376582

ABSTRACT

Developing effective treatments for colorectal cancers through combinations of small-molecule approaches and immunotherapies present intriguing possibilities for managing these otherwise intractable cancers. During a broad-based, screening effort against multiple colorectal cancer cell lines, we identified indole-substituted quinolines (ISQ), such as N7,N7 -dimethyl-3-(1-methyl-1H-indol-3-yl)quinoline-2,7-diamine (ISQ-1), as potent in vitro inhibitors of several cancer cell lines. We found that ISQ-1 inhibited Wnt signaling, a main driver in the pathway governing colorectal cancer development, and ISQ-1 also activated adenosine monophosphate kinase (AMPK), a cellular energy-homeostasis master regulator. We explored the effect of ISQs on cell metabolism. Seahorse assays measuring oxygen consumption rate (OCR) indicated that ISQ-1 inhibited complex I (i.e., NADH ubiquinone oxidoreductase) in the mitochondrial, electron transport chain (ETC). In addition, ISQ-1 treatment showed remarkable synergistic depletion of oncogenic c-Myc protein level in vitro and induced strong tumor remission in vivo when administered together with BI2536, a polo-like kinase-1 (Plk1) inhibitor. These studies point toward the potential value of dual drug therapies targeting the ETC and Plk-1 for the treatment of c-Myc-driven cancers.


Subject(s)
Amodiaquine/analogs & derivatives , Cell Cycle Proteins/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , Drug Synergism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Amodiaquine/pharmacology , Animals , Apoptosis , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Male , Mice , Mice, Nude , Proto-Oncogene Proteins c-myc/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
2.
Article in English | MEDLINE | ID: mdl-34364298

ABSTRACT

Amodiaquine is a drug used for treatment of malaria and is often used in combination with artesunate in areas where malaria parasites are still susceptible to amodiaquine. Liquid chromatography tandem-mass spectrometry was used to quantify amodiaquine and its active metabolite, desethylamodiaquine, in plasma samples. A low sample volume of 100 µl, and high-throughput extraction technique using a supported liquid extraction (SLE+) technique on an automated liquid handler platform for faster sample processing are some of the advantages of this method. Separation of amodiaquine from desethylamodiaquine was achieved using a reversed phase Zorbax SB-CN 50 mm × 4.6 mm, I.D. 3.5 µm column with acetonitrile and 20 mM ammonium formate with 1% formic acid pH ~ 2.6 (15-85, v/v) as mobile phase. The absolute recoveries of amodiaquine and desethylamodiaquine were 66% to 76%, and their isotope label internal standard were in the range of 73% to 85%. Validation results of the developed method demonstrated intra-batch and inter-batch precisions within the acceptance criteria range of ± 15.0%. There were no matrix or carry-over effects observed. The lower limit of quantification was 1.08 ng/ml for amodiaquine and 1.41 ng/ml for desethylamodiaquine. The method showed robust and accurate performance with high sensitivity. Thus, the validated method was successfully implemented and applied in the evaluation of a clinical trial where participants received artemether-lumefantrine plus amodiaquine twice daily for three days (amodiaquine dose of 10 mg base/kg/day).


Subject(s)
Amodiaquine/analogs & derivatives , Amodiaquine/blood , Antimalarials/blood , Amodiaquine/isolation & purification , Amodiaquine/pharmacokinetics , Antimalarials/isolation & purification , Antimalarials/pharmacokinetics , Chromatography, Liquid , High-Throughput Screening Assays , Humans , Limit of Detection , Linear Models , Liquid-Liquid Extraction , Reproducibility of Results , Tandem Mass Spectrometry
3.
Malar J ; 20(1): 18, 2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33407454

ABSTRACT

BACKGROUND: Accurate measurement of anti-malarial drug concentrations in therapeutic efficacy studies is essential to distinguish between inadequate drug exposure and anti-malarial drug resistance, and to inform optimal anti-malarial dosing in key target population groups. METHODS: A sensitive and selective LC-MS/MS method was developed and validated for the simultaneous determination of amodiaquine and its active metabolite, desethylamodiaquine, and used to describe their pharmacokinetic parameters in Ghanaian patients with uncomplicated falciparum malaria treated with the fixed-dose combination, artesunate-amodiaquine. RESULTS: The day-28 genotype-adjusted adequate clinical and parasitological response rate in 308 patients studied was > 97% by both intention-to-treat and per-protocol analysis. After excluding 64 patients with quantifiable amodiaquine concentrations pre-treatment and 17 with too few quantifiable concentrations, the pharmacokinetic analysis included 227 patients (9 infants, 127 aged 1-4 years, 91 aged ≥ 5 years). Increased median day-3 amodiaquine concentrations were associated with a lower risk of treatment failure [HR 0.87 (95% CI 0.78-0.98), p = 0.021]. Amodiaquine exposure (median AUC0-∞) was significantly higher in infants (4201 ng h/mL) and children aged 1-5 years (1994 ng h/mL) compared to older children and adults (875 ng h/mL, p = 0.001), even though infants received a lower mg/kg amodiaquine dose (median 25.3 versus 33.8 mg/kg in older patients). Desethylamodiaquine AUC0-∞ was not significantly associated with age. No significant safety concerns were identified. CONCLUSIONS: Efficacy of artesunate-amodiaquine at currently recommended dosage regimens was high across all age groups. Reassuringly, amodiaquine and desethylamodiaquine exposure was not reduced in underweight-for-age young children or those with high parasitaemia, two of the most vulnerable target populations. A larger pharmacokinetic study with close monitoring of safety, including full blood counts and liver function tests, is needed to confirm the higher amodiaquine exposure in infants, understand any safety implications and assess whether dose optimization in this vulnerable, understudied population is needed.


Subject(s)
Amodiaquine/analogs & derivatives , Amodiaquine/pharmacokinetics , Antimalarials/pharmacokinetics , Malaria, Falciparum/prevention & control , Adolescent , Adult , Aged , Aged, 80 and over , Amodiaquine/administration & dosage , Artemisinins/administration & dosage , Child , Child, Preschool , Chromatography, Liquid/methods , Drug Combinations , Female , Ghana , Humans , Infant , Malaria, Falciparum/parasitology , Male , Middle Aged , Tandem Mass Spectrometry/methods , Young Adult
4.
Malar J ; 19(1): 238, 2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32631416

ABSTRACT

BACKGROUND: Since 2014, seasonal malaria chemoprevention (SMC) with amodiaquine-sulfadoxine-pyrimethamine (AQ-SP) has been implemented on a large scale during the high malaria transmission season in Burkina Faso. This paper reports the prevalence of microscopic and submicroscopic malaria infection at the outset and after the first round of SMC in children under 5 years old in Bama, Burkina Faso, as well as host and parasite factors involved in mediating the efficacy and tolerability of SMC. METHODS: Two sequential cross-sectional surveys were conducted in late July and August 2017 during the first month of SMC in a rural area in southwest Burkina Faso. Blood smears and dried blood spots were collected from 106 to 93 children under five, respectively, at the start of SMC and again 3 weeks later. Malaria infection was detected by microscopy and by PCR from dried blood spots. For all children, day 7 plasma concentrations of desethylamodiaquine (DEAQ) were measured and CYP2C8 genetic variants influencing AQ metabolism were genotyped. Samples were additionally genotyped for pfcrt K76T and pfmdr1 N86Y, molecular markers associated with reduced amodiaquine susceptibility. RESULTS: 2.8% (3/106) of children were positive for Plasmodium falciparum infection by microscopy and 13.2% (14/106) by nested PCR within 2 days of SMC administration. Three weeks after SMC administration, in the same households, 4.3% (4/93) of samples were positive by microscopy and 14.0% (13/93) by PCR (p = 0.0007). CYP2C8*2, associated with impaired amodiaquine metabolism, was common with an allelic frequency of 17.1% (95% CI 10.0-24.2). Day 7 concentration of DEAQ ranged from 0.48 to 362.80 ng/mL with a median concentration of 56.34 ng/mL. Pfmdr1 N86 predominated at both time points, whilst a non-significant trend towards a higher prevalence of pfcrt 76T was seen at week 3. CONCLUSION: This study showed a moderate prevalence of low-level malaria parasitaemia in children 3 weeks following SMC during the first month of administration. Day 7 concentrations of the active DEAQ metabolite varied widely, likely reflecting variability in adherence and possibly metabolism. These findings highlight factors that may contribute to the effectiveness of SMC in children in a high transmission setting.


Subject(s)
Amodiaquine/analogs & derivatives , Antimalarials/blood , Cytochrome P-450 CYP2C8/genetics , Drug Resistance/genetics , Genes, Protozoan/drug effects , Malaria, Falciparum/prevention & control , Polymorphism, Genetic/drug effects , Amodiaquine/blood , Amodiaquine/therapeutic use , Antimalarials/therapeutic use , Burkina Faso/epidemiology , Chemoprevention , Child, Preschool , Cross-Sectional Studies , Female , Humans , Infant , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Male , Plasma/chemistry
5.
Malar J ; 19(1): 8, 2020 Jan 06.
Article in English | MEDLINE | ID: mdl-31906948

ABSTRACT

BACKGROUND: Artemisinin-based combination therapy (ACT) is recommended to improve malaria treatment efficacy and limit drug-resistant parasites selection in malaria endemic areas. 5 years after they were adopted, the efficacy and safety of artemether-lumefantrine (AL) and artesunate-amodiaquine (ASAQ), the first-line treatments for uncomplicated malaria were assessed in Burkina Faso. METHODS: In total, 440 children with uncomplicated Plasmodium falciparum malaria were randomized to receive either AL or ASAQ for 3 days and were followed up weekly for 42 days. Blood samples were collected to investigate the ex vivo susceptibility of P. falciparum isolates to lumefantrine, dihydroartemisinin (the active metabolite of artemisinin derivatives) and monodesethylamodiaquine (the active metabolite of amodiaquine). The modified isotopic micro test technique was used to determine the 50% inhibitory concentration (IC50) values. Primary endpoints were the risks of treatment failure at days 42. RESULTS: Out of the 440 patients enrolled, 420 (95.5%) completed the 42 days follow up. The results showed a significantly higher PCR unadjusted cure rate in ASAQ arm (71.0%) than that in the AL arm (49.8%) on day 42, and this trend was similar after correction by PCR, with ASAQ performing better (98.1%) than AL (91.1%). Overall adverse events incidence was low and not significantly different between the two treatment arms. Ex vivo results showed that 6.4% P. falciparum isolates were resistant to monodesthylamodiaquine. The coupled in vivo/ex vivo analysis showed increased IC50 values for lumefantrine and monodesethylamodiaquine at day of recurrent parasitaemia compared to baseline values while for artesunate, IC50 values remained stable at baseline and after treatment failure (p > 0.05). CONCLUSION: These findings provide substantial evidence that AL and ASAQ are highly efficacious for the treatment of uncomplicated malaria in children in Burkina Faso. However, the result of P. falciparum susceptibility to the partner drugs advocates the need to regularly replicate such surveillance studies. This would be particularly indicated when amodiaquine is associated in seasonal malaria chemoprophylaxis (SMC) mass drug administration in children under 5 years in Burkina Faso. Trial registration clinicaltrials, NCT00808951. Registered 05 December 2008,https://clinicaltrials.gov/ct2/show/NCT00808951?cond=NCT00808951&rank=1.


Subject(s)
Amodiaquine/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Artemisinins/therapeutic use , Drug Therapy, Combination/methods , Malaria, Falciparum/drug therapy , Adolescent , Amodiaquine/administration & dosage , Amodiaquine/analogs & derivatives , Antimalarials/therapeutic use , Artemether, Lumefantrine Drug Combination/administration & dosage , Artemisinins/administration & dosage , Artesunate/therapeutic use , Burkina Faso , Child , Child, Preschool , Drug Combinations , Female , Humans , Infant , Inhibitory Concentration 50 , Lumefantrine/therapeutic use , Male , Mass Drug Administration , Plasmodium falciparum/drug effects , Plasmodium falciparum/isolation & purification , Treatment Failure , Treatment Outcome
6.
Eur J Drug Metab Pharmacokinet ; 45(1): 81-88, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31642009

ABSTRACT

BACKGROUND AND OBJECTIVE: MAMA decoction (MD) is an antimalarial product prepared from the leaves of Mangifera indica L. (Anacardiaceae), Alstonia boonei De Wild (Apocynaceae), Morinda lucida Benth (Rubiaceae) and Azadirachta indica A. Juss (Meliaceae). A previous report showed that MD enhanced the efficacy of amodiaquine (AQ) in malaria-infected mice, thus suggesting a herb-drug interaction. The present study hence evaluated the effect of MD on the disposition of AQ in mice with a view to investigating a possible pharmacokinetic interaction. METHODS: In a 3-period study design, three groups of mice (n = 72) were administered oral doses of AQ (10 mg/kg/day) alone, concurrently with MD (120 mg/kg/day), and in the 3rd period, mice were given AQ after a 3-day pre-treatment with MD. Blood samples were collected between 0 and 96 h for quantification of AQ and its major metabolite, desethylamodiaquine, by a validated high-performance liquid chromatography method. RESULTS: Maximum concentrations of AQ increased by 12% with the concurrent dosing of MD and by 85% in the group of mice pre-treated with MD. The exposure and half-life of desethylamodiaquine increased by approximately 11% and 21%, respectively, with concurrent administration. Corresponding increases of approximately 20% and 33% of desethylamodiaquine were also observed in mice pre-treated with MD. CONCLUSION: MD influenced the pharmacokinetics of AQ and desethylamodiaquine, its major metabolite. The increase in the half-life and systemic exposure of AQ following its co-administration with MD may provide a basis for the enhanced pharmacological effect of the combination in an earlier study in Plasmodium-infected mice.


Subject(s)
Amodiaquine/pharmacokinetics , Antimalarials/pharmacokinetics , Malaria/drug therapy , Plant Extracts/pharmacology , Amodiaquine/analogs & derivatives , Amodiaquine/blood , Amodiaquine/pharmacology , Animals , Antimalarials/blood , Antimalarials/pharmacology , Drug Combinations , Drug Interactions , Female , Herb-Drug Interactions , Male , Mice , Models, Animal , Plant Leaves/chemistry
7.
Toxicol In Vitro ; 62: 104669, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31629065

ABSTRACT

Amodiaquine (ADQ), an antimalarial drug used in endemic areas, has been reported to be associated with liver toxicity; however, the mechanism underlying its hepatoxicity remains unclear. In this study, we examined the cytotoxicity of ADQ and its major metabolite N-desethylamodiaquine (NADQ) and the effect of cytochrome P450 (CYP)-mediated metabolism on ADQ-induced cytotoxicity. After a 48-h treatment, ADQ and NADQ caused cytotoxicity and induced apoptosis in HepG2 cells; NADQ was slightly more toxic than ADQ. ADQ treatment decreased the levels of anti-apoptotic Bcl-2 family proteins, which was accompanied by an increase in the levels of pro-apoptotic Bcl-2 family proteins, indicating that ADQ-induced apoptosis was mediated by the Bcl-2 family. NADQ treatment markedly increased the phosphorylation of JNK, extracellular signal-regulated kinase (ERK1/2), and p38, indicating that NADQ-induced apoptosis was mediated by MAPK signaling pathways. Metabolic studies using microsomes obtained from HepG2 cell lines overexpressing human CYPs demonstrated that CYP1A1, 2C8, and 3A4 were the major enzymes that metabolized ADQ to NADQ and that CYP1A2, 1B1, 2C19, and 3A5 also metabolized ADQ, but to a lesser extent. The cytotoxicity of ADQ was increased in CYP2C8 and 3A4 overexpressing HepG2 cells compared to HepG2/CYP vector cells, confirming that NADQ was more toxic than ADQ. Moreover, treatment of CYP2C8 and 3A4 overexpressing HepG2 cells with ADQ increased the phosphorylation of JNK, ERK1/2, and p38, but not the expression of Bcl-2 family proteins. Our findings indicate that ADQ and its major metabolite NADQ induce apoptosis, which is mediated by members of the Bcl-2 family and the activation of MAPK signaling pathways, respectively.


Subject(s)
Amodiaquine/analogs & derivatives , Apoptosis/drug effects , Amodiaquine/toxicity , Cell Survival/drug effects , Cytochrome P-450 Enzyme System/metabolism , Hep G2 Cells , Humans , Isoenzymes/metabolism , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism
8.
Clin Pharmacol Ther ; 107(5): 1179-1188, 2020 05.
Article in English | MEDLINE | ID: mdl-31652336

ABSTRACT

Poor adherence to seasonal malaria chemoprevention (SMC) might affect the protective effectiveness of SMC. Here, we evaluated the population pharmacokinetic properties of amodiaquine and its active metabolite, desethylamodiaquine, in children receiving SMC under directly observed ideal conditions (n = 136), and the adherence of SMC at an implementation phase in children participating in a case-control study to evaluate SMC effectiveness (n = 869). Amodiaquine and desethylamodiaquine concentration-time profiles were described simultaneously by two-compartment and three-compartment disposition models, respectively. The developed methodology to evaluate adherence showed a sensitivity of 65-71% when the first dose of SMC was directly observed and 71-73% when no doses were observed in a routine programmatic setting. Adherence simulations and measured desethylamodiaquine concentrations in the case-control children showed complete adherence (all doses taken) in < 20% of children. This result suggests that more efforts are needed urgently to improve the adherence to SMC among children in this area.


Subject(s)
Amodiaquine/administration & dosage , Antimalarials/administration & dosage , Malaria/prevention & control , Medication Adherence/statistics & numerical data , Models, Biological , Africa , Amodiaquine/analogs & derivatives , Amodiaquine/pharmacokinetics , Antimalarials/pharmacokinetics , Case-Control Studies , Chemoprevention , Child, Preschool , Female , Humans , Infant , Male , Prospective Studies , Seasons
9.
J Vector Borne Dis ; 56(3): 221-230, 2019.
Article in English | MEDLINE | ID: mdl-32655071

ABSTRACT

BACKGROUND & OBJECTIVES: Due to the rapid increase of drug resistance in Plasmodium parasites, there is a pressing need of developing new antiplasmodial drugs. In this study, new amodiaquine (AQ) analogs were synthesized, followed by an evaluation of their antiplasmodial activity. METHODS: A new series of quinoline derivatives containing N-alkyl (piperazin-1-yl)methyl benzamidine moiety was synthesized by reacting 4-[(4-(7-chloroquinolin-4-yl)piperazin-1-yl)methyl]benzonitrile with appropriate primary amines. The synthesized compounds were investigated for inhibitory activity by inhibition test of heme detoxification (ITHD). Their antiplasmodial activity was then evaluated using the classical 4-day suppressive test (Peter's test) against Plasmodium berghei-infected mice (ANKA strain). RESULTS: The results showed that the percentage of heme detoxification inhibition in the active compounds was 90%. The most promising analogs, N-butyl-4-[(4-(7-chloroquinolin-4-yl)piperazin-1-yl)methyl]benzamidine (compound 1e), and 4-[(4-(7-chloroquinolin-4-yl)piperazin-1-yl)methyl)]-N-(4-methylpentan-2-yl)benzamidine (compound 1f) displayed 97.65 and 99.18% suppressions at the doses of 75 and 50 mg/kg/day, respectively. Further, the mean survival time of the mice treated with these compounds was higher than that of the negative control group. INTERPRETATION & CONCLUSION: The newly synthesized amodiaquine analogs presented sufficient antiplasmodial activity with excellent suppressions and high in vitro heme detoxification inhibition. Higher mean survival time of the mice treated with synthetic compounds further confirmed the in vivo antimalarial activity of these new AQ analogs. Therefore, these compounds have the potential to replace common drugs from 4-aminoquinoline class. However, further investigations such as pharmacokinetic evaluations, cytotoxicity, toxicity, and formulation seem to be necessary.


Subject(s)
Amodiaquine/therapeutic use , Antimalarials/therapeutic use , Plasmodium berghei/drug effects , Amodiaquine/analogs & derivatives , Amodiaquine/chemical synthesis , Animals , Antimalarials/chemical synthesis , Drug Resistance , Female , Malaria/drug therapy , Mice , Mice, Inbred BALB C
10.
J Clin Pharm Ther ; 43(5): 626-632, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29920710

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Moringa oleifera (MO) Lam (Moringaceae) is commonly used as food supplement and as medicine in most African countries where malaria is also endemic. Therefore, co-administration of MO with antimalarials is a possibility. This study investigated the effects of MO leaves powder on the pharmacokinetics of amodiaquine (AQ) in human subjects. METHODS: Twenty healthy volunteers were recruited for the 3-period study. In the first period, a single dose of AQ tablet (10 mg/kg) was administered orally after an overnight fast. After a 7-day washout period, AQ was co-administered with MO. For the third period, each subject took 3 g MO once daily for 7 days and on the 8th day, MO was co-administered with AQ. The plasma concentrations of amodiaquine and desethylamodiaquine (DEAQ) were simultaneously determined using a validated HPLC method. RESULTS AND DISCUSSION: The results showed a significant decrease (P = .037) in the Cmax of AQ after concurrent administration (CA) with MO, whereas after pretreatment (PT), there was a 32% decrease in the Cmax of AQ. For the metabolite, DEAQ, Cmax increased significantly (P = .006) by 79.36%, and Cmax in PT was significantly higher than (P = .001) that of the CA arm of the study. AUC of DEAQ increased significantly by 40.4% (P = .006) and by 188% (P = .001) after CA and PT, respectively. WHAT IS NEW AND CONCLUSION: The study established pharmacokinetic interaction between AQ and MO when given together or following a long period of ingestion of MO. This may have clinical implications for malaria therapy.


Subject(s)
Amodiaquine/pharmacokinetics , Moringa oleifera/adverse effects , Plant Leaves/adverse effects , Powders/adverse effects , Adult , Amodiaquine/analogs & derivatives , Antimalarials/pharmacokinetics , Female , Healthy Volunteers , Herb-Drug Interactions/physiology , Humans , Male , Tablets/pharmacokinetics , Young Adult
11.
Article in English | MEDLINE | ID: mdl-29760133

ABSTRACT

There are limited data on the pharmacokinetic and safety profiles of artesunate-amodiaquine in human immnunodeficiency virus-infected (HIV+) individuals receiving antiretroviral therapy. In a two-step intensive sampling pharmacokinetic trial, we compared the area under the concentration-time curve from 0 to 28 days (AUC0-28) of an active metabolite of amodiaquine, desethylamodiaquine, and treatment-emergent adverse events between antiretroviral therapy-naive HIV+ adults and those taking nevirapine and ritonavir-boosted lopinavir-based antiretroviral therapy. In step 1, malaria-uninfected adults (n = 6/arm) received half the standard adult treatment regimen of artesunate-amodiaquine. In step 2, another cohort (n = 25/arm) received the full regimen. In step 1, there were no safety signals or significant differences in desethylamodiaquine AUC0-28 among participants in the ritonavir-boosted lopinavir, nevirapine, and antiretroviral therapy-naive arms. In step 2, compared with those in the antiretroviral therapy-naive arm, participants in the ritonavir-boosted lopinavir arm had 51% lower desethylamodiaquine AUC0-28, with the following geometric means (95% confidence intervals [CIs]): 23,822 (17,458 to 32,506) versus 48,617 (40,787 to 57,950) ng · h/ml (P < 0.001). No significant differences in AUC0-28 were observed between nevirapine and antiretroviral therapy-naive arms. Treatment-emergent transaminitis was higher in the nevirapine (20% [5/25]) than the antiretroviral therapy-naive (0.0% [0/25]) arm (risk difference, 20% [95% CI, 4.3 to 35.7]; P = 0.018). The ritonavir-boosted lopinavir antiretroviral regimen was associated with reduced desethylamodiaquine exposure, which may compromise artesunate-amodiaquine's efficacy. Coadministration of nevirapine and artesunate-amodiaquine may be associated with hepatoxicity.


Subject(s)
Amodiaquine/analogs & derivatives , Anti-HIV Agents/therapeutic use , Antimalarials/adverse effects , Antimalarials/pharmacokinetics , Artemisinins/adverse effects , Artemisinins/pharmacokinetics , HIV Infections/drug therapy , Malaria, Falciparum/drug therapy , Adult , Amodiaquine/adverse effects , Amodiaquine/pharmacokinetics , Amodiaquine/therapeutic use , Antimalarials/therapeutic use , Artemisinins/therapeutic use , Drug Combinations , Drug Interactions , Drug Therapy, Combination/adverse effects , Female , Humans , Lopinavir/therapeutic use , Malawi , Male , Nevirapine/therapeutic use , Ritonavir/therapeutic use
12.
Am J Trop Med Hyg ; 99(1): 65-72, 2018 07.
Article in English | MEDLINE | ID: mdl-29741150

ABSTRACT

The pharmacokinetics (PK) and ex vivo activity (pharmacodynamics [PD]) of two artemisinin combination therapies (ACTs) (artemisinin-piperaquine [ARN-PPQ] [Artequick®] and artesunate-amodiaquine [ARS-AQ] [Coarsucam™]) in healthy Vietnamese volunteers were compared following 3-day courses of the ACTs for the preselection of the drugs for falciparum malaria therapy. For PK analysis, serial plasma samples were collected from two separate groups of 22 volunteers after ACT administration. Of these volunteers, ex vivo activity was assessed in plasma samples from seven volunteers who received both ACTs. The area under the concentration-time curve (AUC0-∞) was 3.6-fold higher for dihydroartemisinin (active metabolite of ARS) than that for ARN, whereas the AUC0-∞ of desethylamodiaquine (active metabolite of AQ) was 2.0-fold lower than that of PPQ. Based on the 50% inhibitory dilution values of the volunteers' plasma samples collected from 0.25 to 3 hours after the last dose, the ex vivo activity of ARS-AQ was 2.9- to 16.2-fold more potent than that of ARN-PPQ against the drug-sensitive D6 Plasmodium falciparum line. In addition, at 1.5, 4.0, and 24 hours after the last dose, the ex vivo activity of ARS-AQ was 20.8-, 3.5-, and 8.5-fold more potent than that of ARN-PPQ against the ARN-sensitive MRA1239 line. By contrast, at 1.5 hours, the ex vivo activity of ARS-AQ was 5.4-fold more active than that of ARN-PPQ but had similar activities at 4 and 24 hours against the ARN-resistant MRA1240 line. The PK-PD data suggest that ARS-AQ possesses superior antimalarial activity than that of ARN-PPQ and would be the preferred ACT for further in vivo efficacy testing in multidrug-resistant falciparum malaria areas.


Subject(s)
Amodiaquine/pharmacokinetics , Antimalarials/pharmacokinetics , Artemisinins/pharmacokinetics , Plasmodium falciparum/drug effects , Quinolines/pharmacokinetics , Amodiaquine/analogs & derivatives , Amodiaquine/blood , Amodiaquine/pharmacology , Antimalarials/blood , Antimalarials/pharmacology , Area Under Curve , Artemisinins/blood , Artemisinins/pharmacology , Biotransformation , Drug Administration Schedule , Drug Combinations , Half-Life , Healthy Volunteers , Humans , Malaria, Falciparum/drug therapy , Male , Parasitic Sensitivity Tests , Plasmodium falciparum/growth & development , Quinolines/blood , Quinolines/pharmacology , Tablets , Young Adult
13.
Eur J Med Chem ; 148: 498-506, 2018 Mar 25.
Article in English | MEDLINE | ID: mdl-29126722

ABSTRACT

Many people are affected by Malaria around the world, and the parasite is developing resistance against available drugs. Currently, isoquine and N-tert-butyl isoquine are some of the most promising antimalarial candidates that have already reached Phase I and II clinical trials, respectively. Nevertheless, pharmacodynamic studies have demonstrated that isoquine is highly sensitive to form O-glucuronide metabolite, which may affect its accumulation in tissues. To avoid the O-glucuronide formation and its negative influence in the accumulation process, a series of novel five dehydroxy isoquine derivatives were designed and prepared herein as potential antimalarial agents. By a simple three-step procedure, five dehydroxy isoquines were prepared and subsequently examined on the inhibition of haemozoin formation, the main target of the 4-aminoquinolines. Four derivatives displayed significant inhibitory activities at low IC50 values from 1.66 to 1.86 µM comparable to CQ. On the basis of the results, these four compounds were subsequently tested against Plasmodium berghei ANKA model in mice, showing to be as active as CQ with significant curative responses and parasitemia suppression in mice infected. On the other hand, these four compounds showed an acceptable non specific cytotoxicity on murine peritoneal macrophague and human erythrocyte cells. Thus, the presented data indicate that the dehydroxy isoquines 4b, 4c and 4e constitute promising cost-effective leads for the development of new antiplasmodial targeted at blood-stage malaria parasites.


Subject(s)
Antimalarials/chemistry , Plasmodium berghei/drug effects , Aminoquinolines , Amodiaquine/analogs & derivatives , Animals , Antimalarials/pharmacology , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Macrophages/drug effects , Macrophages/parasitology , Mice , Parasitemia/drug therapy
14.
Article in English | MEDLINE | ID: mdl-28923866

ABSTRACT

Dihydroartemisinin-piperaquine (DP) has demonstrated excellent efficacy for the treatment and prevention of malaria in Uganda. However, resistance to both components of this regimen has emerged in Southeast Asia. The efficacy of artemether-lumefantrine, the first-line regimen to treat malaria in Uganda, has also been excellent, but continued pressure may select for parasites with decreased sensitivity to lumefantrine. To gain insight into current drug sensitivity patterns, ex vivo sensitivities were assessed and genotypes previously associated with altered drug sensitivity were characterized for 58 isolates collected in Tororo, Uganda, from subjects presenting in 2016 with malaria from the community or as part of a clinical trial comparing DP chemoprevention regimens. Compared to community isolates, those from trial subjects had lower sensitivities to the aminoquinolines chloroquine, monodesethyl amodiaquine, and piperaquine and greater sensitivities to lumefantrine and mefloquine, an observation consistent with DP selection pressure. Compared to results for isolates from 2010 to 2013, the sensitivities of 2016 community isolates to chloroquine, amodiaquine, and piperaquine improved (geometric mean 50% inhibitory concentrations [IC50] = 248, 76.9, and 19.1 nM in 2010 to 2013 and 33.4, 14.9, and 7.5 nM in 2016, respectively [P < 0.001 for all comparisons]), the sensitivity to lumefantrine decreased (IC50 = 3.0 nM in 2010 to 2013 and 5.4 nM in 2016 [P < 0.001]), and the sensitivity to dihydroartemisinin was unchanged (IC50 = 1.4 nM). These changes were accompanied by decreased prevalence of transporter mutations associated with aminoquinoline resistance and low prevalence of polymorphisms recently associated with resistance to artemisinins or piperaquine. Antimalarial drug sensitivities are changing in Uganda, but novel genotypes associated with DP treatment failure in Asia are not prevalent.


Subject(s)
Antimalarials/therapeutic use , Drug Resistance/genetics , Malaria, Falciparum/drug therapy , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Plasmodium falciparum/drug effects , Protozoan Proteins/genetics , Adolescent , Amodiaquine/analogs & derivatives , Amodiaquine/therapeutic use , Artemisinins/therapeutic use , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Child , Child, Preschool , Chloroquine/therapeutic use , Ethanolamines/therapeutic use , Female , Fluorenes/therapeutic use , Gene Expression , Humans , Infant , Inhibitory Concentration 50 , Lumefantrine , Malaria, Falciparum/parasitology , Male , Mefloquine/therapeutic use , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Mutation , Parasitic Sensitivity Tests , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Quinolines/therapeutic use , Uganda , Young Adult
15.
Toxicol Lett ; 275: 83-91, 2017 Jun 05.
Article in English | MEDLINE | ID: mdl-28478157

ABSTRACT

Amodiaquine (AQ), an antimalarial drug, widely prescribed in endemic areas of Africa and Asia, is used in combination with artesunate as recommended by the WHO. However, due to its idiosyncratic hepatotoxicity and agranulocytosis, the therapeutic use has been discontinued in most countries. Oxidative bioactivation to protein-reactive quinonimines (QIs) by hepatic cytochrome P450s and myeloperoxidase (MPO) have been suggested to be important mechanisms underlying AQ idiosyncratic toxicity. However, the inactivation of the reactive QIs by detoxifying enzymes such as human glutathione S-transferases (GSTs) and NAD(P)H:quinone oxidoreducatase 1 (NQO1) has not been characterized yet. In the present study, the activities of 15 recombinant human GSTs and NQO1 in the inactivation of reactive QIs of AQ and its pharmacological active metabolite, N-desethylamodiaquine (DEAQ) were investigated. The results showed that GSTP1-1, GSTA4-4, GSTM4-4, GSTM2-2 and GSTA2-2 (activity in decreasing order) were active isoforms in catalyzing GSH conjugation of reactive QIs of AQ and DEAQ. Additionally, NQO1 was shown to inactivate these QIs by reduction. Simulation of the variability of cytosolic GST-activity based on the hepatic GST contents from 22 liver donors, showed a large variation in cytosolic inactivation of QIs by GSH, especially at a reduced GSH-concentration. In conclusion, the present study demonstrates that a low hepatic expression of the active GSTs and NQO1 may increase the susceptibility of patients to AQ idiosyncratic hepatotoxicity.


Subject(s)
Amodiaquine/analogs & derivatives , Chemical and Drug Induced Liver Injury/metabolism , Glutathione Transferase/metabolism , Microsomes, Liver/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Amodiaquine/metabolism , Amodiaquine/toxicity , Biocatalysis , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/etiology , Escherichia coli/genetics , Glutathione Transferase/genetics , Humans , In Vitro Techniques , Isoenzymes , Microsomes, Liver/enzymology , NAD(P)H Dehydrogenase (Quinone)/genetics , Recombinant Proteins , Transfection
16.
Malar J ; 16(1): 118, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28302108

ABSTRACT

BACKGROUND: In response to increasing resistance to anti-malarial drugs, Senegal adopted artemisinin-based combination therapy (ACT) as the first-line treatment for uncomplicated malaria in 2006. However, resistance of Plasmodium falciparum parasites to artemisinin derivatives, characterized by delayed parasite clearance after treatment with ACT or artesunate monotherapy, has recently emerged and rapidly spread in Southeast Asia. After 10 years of stability with rates ranging from 5.6 to 11.8%, the prevalence of parasites with reduced susceptibility in vitro to monodesethylamodiaquine, the active metabolite of an ACT partner drug, increased to 30.6% in 2014 in Dakar. Additionally, after a decrease of the in vitro chloroquine resistance in Dakar in 2009-2011, the prevalence of parasites that showed in vitro chloroquine resistance increased again to approximately 50% in Dakar since 2013. The aim of this study was to follow the evolution of the susceptibility to ACT partners and other anti-malarial drugs in 2015 in Dakar. An in vitro test is the only method currently available to provide an early indication of resistance to ACT partners. RESULTS: Thirty-two P. falciparum isolates collected in 2015 in Dakar were analysed using a standard ex vivo assay based on an HRP2 ELISA. The prevalence of P. falciparum parasites with reduced susceptibility in vitro to monodesethylamodiaquine, chloroquine, mefloquine, doxycycline and quinine was 28.1, 46.9, 45.2, 31.2 and 9.7%, respectively. None of the parasites were resistant to lumefantrine, piperaquine, pyronaridine, dihydroartemisinin and artesunate. These results confirm an increase in the reduced susceptibility to monodesethylamodiaquine observed in 2014 in Dakar and the chloroquine resistance observed in 2013. The in vitro resistance seems to be established in Dakar. Additionally, the prevalence of parasites with reduced susceptibility to doxycycline has increased two-fold compared to 2014. CONCLUSIONS: The establishment of a reduced susceptibility to monodesethylamodiaquine as well as chloroquine resistance, and the emergence of a reduced susceptibility to doxycycline are disturbing. The in vitro and in vivo surveillance of anti-malarial drugs must be implemented in Senegal.


Subject(s)
Amodiaquine/analogs & derivatives , Antimalarials/pharmacology , Chloroquine/pharmacology , Drug Resistance , Plasmodium falciparum/drug effects , Amodiaquine/pharmacology , Artemisinins/pharmacology , Drug Therapy, Combination , Senegal
17.
Article in English | MEDLINE | ID: mdl-28052850

ABSTRACT

Polymorphisms and the overexpression of transporter genes, especially of the ATP-binding cassette superfamily, have been involved in antimalarial drug resistance. The objective of this study was to use 77 Senegalese Plasmodium falciparum isolates to evaluate the association between the number of Asn residues in the polymorphic microsatellite region of the Plasmodium falciparum multidrug resistance 6 gene (Pfmdr6) and the ex vivo susceptibility to antimalarials. A significant association was observed between the presence of 7 or 9 Asn repeats and reduced susceptibility to quinine.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Antimalarials/pharmacology , Drug Resistance/genetics , Plasmodium falciparum/genetics , Polymorphism, Genetic , Protozoan Proteins/genetics , Quinine/pharmacology , Amodiaquine/analogs & derivatives , Amodiaquine/pharmacology , Artemisinins/pharmacology , Artesunate , Asparagine/metabolism , Chloroquine/pharmacology , Doxycycline/pharmacology , Ethanolamines/pharmacology , Fluorenes/pharmacology , Gene Expression , Humans , Inhibitory Concentration 50 , Lumefantrine , Malaria, Falciparum/parasitology , Mefloquine/pharmacology , Naphthyridines/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Plasmodium falciparum/isolation & purification , Protein Isoforms/genetics , Quinolines/pharmacology , Repetitive Sequences, Amino Acid , Senegal
18.
Br J Clin Pharmacol ; 83(3): 572-583, 2017 03.
Article in English | MEDLINE | ID: mdl-27718269

ABSTRACT

AIMS: Oxidative bioactivation of amodiaquine (AQ) by cytochrome P450s to a reactive quinoneimine is considered as an important mechanism underlying its idiosyncratic hepatotoxicity. However, because internal exposure to its major metabolite N-desethylamodiaquine (DEAQ) is up to 240-fold higher than AQ, bioactivation of DEAQ might significantly contribute to covalent binding. The aim of the present study was to compare the kinetics of bioactivation of AQ and DEAQ by human liver microsomes (HLM) and to characterize the CYPs involved in bioactivation of AQ and DEAQ. METHODS: Glutathione was used to trap reactive metabolites formed in incubations of AQ and DEAQ with HLM and recombinant human cytochrome P450s (hCYPs). Kinetics of bioactivation of AQ and DEAQ in HLM and involvement of hCYPs were characterized by measuring corresponding glutathione conjugates (AQ-SG and DEAQ-SG) using a high-performance liquid chromatography method. RESULTS: Bioactivation of AQ and DEAQ in HLM both exhibited Michaelis-Menten kinetics. For AQ bioactivation, enzyme kinetical parameters were Km , 11.5 ± 2.0 µmol l-1 , Vmax , 59.2 ± 3.2 pmol min-1  mg-1 and CLint , 5.15 µl min-1  mg-1 . For DEAQ, parameters for bioactivation were Km , 6.1 ± 1.3 µmol l-1 , Vmax , 5.5 ± 0.4 pmol min-1  mg-1 and CLint 0.90 µl min-1  mg-1 . Recombinant hCYPs and inhibition studies with HLM showed involvement of CYP3A4, CYP2C8, CYP2C9 and CYP2D6 in bioactivation. CONCLUSIONS: The major metabolite DEAQ is likely to be quantitatively more important than AQ with respect to hepatic exposure to reactive metabolites in vivo. High expression of CYP3A4, CYP2C8, CYP2C9, and CYP2D6 may be risk factors for hepatotoxicity caused by AQ-therapy.


Subject(s)
Activation, Metabolic/genetics , Amodiaquine/analogs & derivatives , Amodiaquine/pharmacokinetics , Microsomes, Liver/enzymology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Glutathione/metabolism , Humans , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism
19.
Antiviral Res ; 137: 141-150, 2017 01.
Article in English | MEDLINE | ID: mdl-27889529

ABSTRACT

In a previous study, twelve antimalarial compounds, amodiaquine (AQ) and derivatives, were shown to have potent anti-dengue viral (DENV) activity by using the stable DENV2 Renilla luciferase reporter replicon expressing BHK-21 cells, infectivity (plaque), and the qRT-PCR assays. In this study, we performed molecular modeling on these compounds to determine their stereo-electronic properties required for optimal antiviral activity. Based on the similarity of calculated stereo-electronic profiles, specifically the electrostatic potential profiles of the compounds, and in silico screening of related compounds from literature, we identified three additional compounds, Quinacrine (QC), Mefloquine (MQ), and GSK369796. Analysis of their antiviral activities indicated that all three compounds have high anti-DENV activity in the DENV2 replicon expressing cells with EC50 values of 5.30 ± 1.31 µM (QC), 3.22 ± 0.37 µM (MQ), and 5.06 ± 0.86 µM (GSK369796). The infectivity assays revealed the EC50 values of 7.09 ± 1.67 µM (QC), 4.36 ± 0.31 µM (MQ) and 3.03 ± 0.35 µM (GSK369796). The mode of action of these compounds is through inhibition of autophagy, thereby affecting DENV2 replication. Moreover, these compounds also showed antiviral activity against the rapidly emerging Zika virus (ZIKV) with EC50 values of 2.27 ± 0.14 µM (QC), 3.95 ± 0.21 µM (MQ), and 2.57 ± 0.09 µM (GSK369796).


Subject(s)
Antimalarials/pharmacology , Antiviral Agents/pharmacology , Dengue Virus/drug effects , Zika Virus/drug effects , Amodiaquine/analogs & derivatives , Amodiaquine/chemistry , Amodiaquine/pharmacology , Antimalarials/chemistry , Autophagy/drug effects , Computer Simulation , Dengue Virus/physiology , Drug Discovery , Humans , Mefloquine/chemistry , Mefloquine/pharmacology , Quinacrine/chemistry , Quinacrine/pharmacology , Replicon/drug effects , Virus Replication/drug effects , Zika Virus/physiology
20.
PLoS One ; 11(8): e0160091, 2016.
Article in English | MEDLINE | ID: mdl-27483471

ABSTRACT

Antimalarial chloroquine (CQ) prevents haematin detoxication when CQ-base concentrates in the acidic digestive vacuole through protonation of its p-aminopyridine (pAP) basic aromatic nitrogen and sidechain diethyl-N. CQ export through the variant vacuolar membrane export channel, PFCRT, causes CQ-resistance in Plasmodium falciparum but 3-methyl CQ (sontochin SC), des-ethyl amodiaquine (DAQ) and bis 4-aminoquinoline piperaquine (PQ) are still active. This is determined by changes in drug accumulation ratios in parasite lipid (LAR) and in vacuolar water (VAR). Higher LAR may facilitate drug binding to and blocking PFCRT and also aid haematin in lipid to bind drug. LAR for CQ is only 8.3; VAR is 143,482. More hydrophobic SC has LAR 143; VAR remains 68,523. Similarly DAQ with a phenol substituent has LAR of 40.8, with VAR 89,366. In PQ, basicity of each pAP is reduced by distal piperazine N, allowing very high LAR of 973,492, retaining VAR of 104,378. In another bis quinoline, dichlorquinazine (DCQ), also active but clinically unsatisfactory, each pAP retains basicity, being insulated by a 2-carbon chain from a proximal nitrogen of the single linking piperazine. While LAR of 15,488 is still high, the lowest estimate of VAR approaches 4.9 million. DCQ may be expected to be very highly lysosomotropic and therefore potentially hepatotoxic. In 11 pAP antimalarials a quadratic relationship between logLAR and logResistance Index (RI) was confirmed, while log (LAR/VAR) vs logRI for 12 was linear. Both might be used to predict the utility of structural modifications.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Hemin/antagonists & inhibitors , Plasmodium falciparum/drug effects , Vacuoles/drug effects , Amodiaquine/analogs & derivatives , Amodiaquine/chemistry , Amodiaquine/metabolism , Amodiaquine/pharmacology , Antimalarials/metabolism , Biological Transport , Chloroquine/analogs & derivatives , Chloroquine/chemistry , Chloroquine/metabolism , Chloroquine/pharmacology , Drug Design , Drug Resistance , Heme/antagonists & inhibitors , Heme/metabolism , Hemin/metabolism , Hydrophobic and Hydrophilic Interactions , Plasmodium falciparum/metabolism , Quinolines/chemistry , Quinolines/metabolism , Quinolines/pharmacology , Structure-Activity Relationship , Vacuoles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...