Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 535
Filter
Add more filters

Publication year range
1.
Circ Res ; 132(9): e116-e133, 2023 04 28.
Article in English | MEDLINE | ID: mdl-36927079

ABSTRACT

BACKGROUND: Small-conductance Ca2+-activated K+ (SK)-channel inhibitors have antiarrhythmic effects in animal models of atrial fibrillation (AF), presenting a potential novel antiarrhythmic option. However, the regulation of SK-channels in human atrial cardiomyocytes and its modification in patients with AF are poorly understood and were the object of this study. METHODS: Apamin-sensitive SK-channel current (ISK) and action potentials were recorded in human right-atrial cardiomyocytes from sinus rhythm control (Ctl) patients or patients with (long-standing persistent) chronic AF (cAF). RESULTS: ISK was significantly higher, and apamin caused larger action potential prolongation in cAF- versus Ctl-cardiomyocytes. Sensitivity analyses in an in silico human atrial cardiomyocyte model identified IK1 and ISK as major regulators of repolarization. Increased ISK in cAF was not associated with increases in mRNA/protein levels of SK-channel subunits in either right- or left-atrial tissue homogenates or right-atrial cardiomyocytes, but the abundance of SK2 at the sarcolemma was larger in cAF versus Ctl in both tissue-slices and cardiomyocytes. Latrunculin-A and primaquine (anterograde and retrograde protein-trafficking inhibitors) eliminated the differences in SK2 membrane levels and ISK between Ctl- and cAF-cardiomyocytes. In addition, the phosphatase-inhibitor okadaic acid reduced ISK amplitude and abolished the difference between Ctl- and cAF-cardiomyocytes, indicating that reduced calmodulin-Thr80 phosphorylation due to increased protein phosphatase-2A levels in the SK-channel complex likely contribute to the greater ISK in cAF-cardiomyocytes. Finally, rapid electrical activation (5 Hz, 10 minutes) of Ctl-cardiomyocytes promoted SK2 membrane-localization, increased ISK and reduced action potential duration, effects greatly attenuated by apamin. Latrunculin-A or primaquine prevented the 5-Hz-induced ISK-upregulation. CONCLUSIONS: ISK is upregulated in patients with cAF due to enhanced channel function, mediated by phosphatase-2A-dependent calmodulin-Thr80 dephosphorylation and tachycardia-dependent enhanced trafficking and targeting of SK-channel subunits to the sarcolemma. The observed AF-associated increases in ISK, which promote reentry-stabilizing action potential duration shortening, suggest an important role for SK-channels in AF auto-promotion and provide a rationale for pursuing the antiarrhythmic effects of SK-channel inhibition in humans.


Subject(s)
Atrial Fibrillation , Animals , Humans , Atrial Fibrillation/metabolism , Apamin/metabolism , Apamin/pharmacology , Primaquine/metabolism , Primaquine/pharmacology , Calmodulin/metabolism , Heart Atria/metabolism , Myocytes, Cardiac/metabolism , Anti-Arrhythmia Agents/therapeutic use , Action Potentials/physiology , Small-Conductance Calcium-Activated Potassium Channels/metabolism
2.
Antimicrob Agents Chemother ; 68(4): e0120423, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38411047

ABSTRACT

Primaquine (PQ) is the main drug used to eliminate dormant liver stages and prevent relapses in Plasmodium vivax malaria. It also has an effect on the gametocytes of Plasmodium falciparum; however, it is unclear to what extent PQ affects P. vivax gametocytes. PQ metabolism involves multiple enzymes, including the highly polymorphic CYP2D6 and the cytochrome P450 reductase (CPR). Since genetic variability can impact drug metabolism, we conducted an evaluation of the effect of CYP2D6 and CPR variants on PQ gametocytocidal activity in 100 subjects with P. vivax malaria. To determine gametocyte density, we measured the levels of pvs25 transcripts in samples taken before treatment (D0) and 72 hours after treatment (D3). Generalized estimating equations (GEEs) were used to examine the effects of enzyme variants on gametocyte densities, adjusting for potential confounding factors. Linear regression models were adjusted to explore the predictors of PQ blood levels measured on D3. Individuals with the CPR mutation showed a smaller decrease in gametocyte transcript levels on D3 compared to those without the mutation (P = 0.02, by GEE). Consistent with this, higher PQ blood levels on D3 were associated with a lower reduction in pvs25 transcripts. Based on our findings, the CPR variant plays a role in the persistence of gametocyte density in P. vivax malaria. Conceptually, our work points to pharmacogenetics as a non-negligible factor to define potential host reservoirs with the propensity to contribute to transmission in the first days of CQ-PQ treatment, particularly in settings and seasons of high Anopheles human-biting rates.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Malaria, Vivax , Malaria , Humans , Antimalarials/pharmacology , Antimalarials/therapeutic use , Malaria, Vivax/drug therapy , Malaria, Falciparum/drug therapy , NADPH-Ferrihemoprotein Reductase , Chloroquine/pharmacology , Cytochrome P-450 CYP2D6/genetics , Artemisinins/pharmacology , Primaquine/pharmacology , Primaquine/therapeutic use , Malaria/drug therapy , Plasmodium falciparum , Plasmodium vivax/genetics
3.
J Pharmacol Exp Ther ; 391(1): 119-129, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39095205

ABSTRACT

Primaquine and Tafenoquine are the only approved drugs that can achieve a radical cure for Plasmodium vivax malaria but are contraindicated in patients who are deficient in glucose 6-phosphate dehydrogenase (G6PDd) due to risk of severe hemolysis from reactive oxygen species generated by redox cycling of drug metabolites. 5-hydroxyprimaquine and its quinoneimine cause robust redox cycling in red blood cells (RBCs) but are so labile as to not be detected in blood or urine. Rather, the quinoneimine is rapidly converted into primaquine-5,6-orthoquinone (5,6-POQ) that is then excreted in the urine. The extent to which 5,6-POQ contributes to hemolysis remains unclear, although some have suggested that it is a minor toxin that should be used predominantly as a surrogate to infer levels of 5-hydroxyprimaquine. In this report, we describe a novel humanized mouse model of the G6PD Mediterranean variant (hG6PDMed-) that recapitulates the human biology of RBC age-dependent enzyme decay, as well as an isogenic matched control mouse with human nondeficient G6PD hG6PDND In vitro challenge of RBCs with 5,6-POQ causes increased generation of superoxide and methemoglobin. Infusion of treated RBCs shows that 5,6-POQ selectively causes in vivo clearance of older hG6PDMed- RBCs. These findings support the hypothesis that 5,6-POQ directly induces hemolysis and challenges the notion that 5,6-POQ is an inactive metabolic waste product. Indeed, given the extreme lability of 5-hydroxyprimaquine and the relative stability of 5,6-POQ, these data raise the possibility that 5,6-POQ is a major hemolytic primaquine metabolite in vivo. SIGNIFICANCE STATEMENT: These findings demonstrate that 5,6-POQ, which has been considered an inert waste product of primaquine metabolism, directly induces ROS that cause clearance of older G6PDd RBCs. As 5,6-POQ is relatively stable compared with other active primaquine metabolites, these data support the hypothesis that 5,6-POQ is a major toxin in primaquine induced hemolysis. The findings herein also establish a new model of G6PDd and provide the first direct evidence, to our knowledge, that young G6PDd RBCs are resistant to primaquine-induced hemolysis.


Subject(s)
Erythrocytes , Glucosephosphate Dehydrogenase Deficiency , Hemolysis , Primaquine , Animals , Hemolysis/drug effects , Erythrocytes/metabolism , Erythrocytes/drug effects , Primaquine/pharmacology , Primaquine/metabolism , Mice , Humans , Glucosephosphate Dehydrogenase Deficiency/metabolism , Glucosephosphate Dehydrogenase/metabolism , Disease Models, Animal , Male , Antimalarials/pharmacology
4.
Bioorg Med Chem ; 104: 117714, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38582046

ABSTRACT

4,9-diaminoacridines with reported antiplasmodial activity were coupled to different trans-cinnamic acids, delivering a new series of conjugates inspired by the covalent bitherapy concept. The new compounds were more potent than primaquine against hepatic stages of Plasmodium berghei, although this was accompanied by cytotoxic effects on Huh-7 hepatocytes. Relevantly, the conjugates displayed nanomolar activities against blood stage P. falciparum parasites, with no evidence of hemolytic effects below 100 µM. Moreover, the new compounds were at least 25-fold more potent than primaquine against P. falciparum gametocytes. Thus, the new antiplasmodial hits disclosed herein emerge as valuable templates for the development of multi-stage antiplasmodial drug candidates.


Subject(s)
Antimalarials , Cinnamates , Malaria, Falciparum , Humans , Antimalarials/pharmacology , Antimalarials/therapeutic use , Primaquine/pharmacology , Disclosure , Plasmodium falciparum , Malaria, Falciparum/drug therapy , Plasmodium berghei
5.
Malar J ; 22(1): 162, 2023 May 20.
Article in English | MEDLINE | ID: mdl-37210520

ABSTRACT

BACKGROUND: Reducing the risk of recurrent Plasmodium vivax malaria is critical for malaria control and elimination. Primaquine (PQ) is the only widely available drug against P. vivax dormant liver stages, but is recommended as a 14-day regimen, which can undermine adherence to a complete course of treatment. METHODS: This is a mixed-methods study to assess socio-cultural factors influencing adherence to a 14-day PQ regimen in a 3-arm, treatment effectiveness trial in Papua, Indonesia. The qualitative strand, consisting of interviews and participant observation was triangulated with a quantitative strand in which trial participants were surveyed using a questionnaire. RESULTS: Trial participants differentiated between two types of malaria: tersiana and tropika, equivalent to P. vivax and Plasmodium falciparum infection, respectively. The perceived severity of both types was similar with 44.0% (267/607) perceiving tersiana vs. 45.1% (274/607) perceiving tropika as more severe. There was no perceived differentiation whether malaria episodes were due to a new infection or relapse; and 71.3% (433/607) acknowledged the possibility of recurrence. Participants were familiar with malaria symptoms and delaying health facility visit by 1-2 days was perceived to increase the likelihood of a positive test. Prior to health facility visits, symptoms were treated with leftover drugs kept at home (40.4%; 245/607) or bought over the counter (17.0%; 103/607). Malaria was considered to be cured with 'blue drugs' (referring to dihydroartemisinin-piperaquine). Conversely, 'brown drugs,' referring to PQ, were not considered malaria medication and instead were perceived as supplements. Adherence to malaria treatment was 71.2% (131/184), in the supervised arm, 56.9% (91/160) in the unsupervised arm and 62.4% (164/263) in the control arm; p = 0.019. Adherence was 47.5% (47/99) among highland Papuans, 51.7% (76/147) among lowland Papuans, and 72.9% (263/361) among non-Papuans; p < 0.001. CONCLUSION: Adherence to malaria treatment was a socio-culturally embedded process during which patients (re-)evaluated the characteristics of the medicines in relation to the course of the illness, their past experiences with illness, and the perceived benefits of the treatment. Structural barriers that hinder the process of patient adherence are crucial to consider in the development and rollout of effective malaria treatment policies.


Subject(s)
Antimalarials , Malaria, Vivax , Malaria , Humans , Malaria, Vivax/drug therapy , Malaria, Vivax/prevention & control , Antimalarials/therapeutic use , Antimalarials/pharmacology , Indonesia , Plasmodium vivax , Primaquine/therapeutic use , Primaquine/pharmacology , Malaria/drug therapy
6.
Malar J ; 22(1): 17, 2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36635642

ABSTRACT

BACKGROUND: Mass drug administration (MDA) with primaquine (PQ) is being considered for accelerating Plasmodium vivax elimination in remaining active foci. This study aimed to determine the acceptability of MDA with PQ in malaria endemic villages in a malarious setting in the South of Thailand undergoing MDA with PQ. METHODS: A cross-sectional mixed-methods approach was conducted in seven malaria endemic villages where MDA with PQ was implemented. The data were collected from community villagers and health workers using structured questionnaires, in-depth interviews, and focus group discussions. Descriptive statistics and logistic regression models were used for quantitative data analysis. Thematic analysis was applied for qualitative data. RESULTS: Among a total of 469 participants from the MDA villages, 293 participants were eligible for MDA with PQ and 79.86% (234) completed 14-days of PQ. The logistic regressions indicated that males (adjusted odds ratio: 2.52 [95% confidence interval: 1.33-4.81]) and those who are farmers (2.57 [1.12-5.90]) were most likely to participate in the MDA. Among 293 participants in the post-MDA study, 74.06% had originally agreed to participate in the MDA with PQ while 25.94% had originally reported not wanting to participate in the MDA. Of those who originally reported being willing to participate in the MDA, 71.23% followed through with participation in the first or second round. Conversely, 93.24% of those who originally reported not being willing to participate in the MDA did in fact participate in the MDA. Factors contributing to higher odds of agreeing to participate and following through with participation included being male (1.98 [1.06-3.69]) and correctly responding that malaria is preventable (2.32 [1.01-5.35]) with some differences by village. Five key themes emerged from the qualitative analyses: concern about side effects from taking PQ; disbelief that malaria could be eliminated in this setting; low overall concern about malaria infections; misunderstandings about malaria; and a general need to tailor public health efforts for this unique context. CONCLUSION: While the reported likelihood of participating in MDA was high in this setting, actual follow-through was relatively moderate, partially because of eligibility (roughly 71% of those in the follow-up survey who originally agreed to participate actually followed through with participation). One of the largest concerns among study participants was PQ-related side effects-and these concerns likely heavily influenced participant adherence to the MDA. The results of this study can be used to tailor future MDAs, or other public health interventions, in this and potentially other similar settings.


Subject(s)
Antimalarials , Drug-Related Side Effects and Adverse Reactions , Malaria, Vivax , Malaria , Humans , Male , Female , Primaquine/therapeutic use , Primaquine/pharmacology , Antimalarials/therapeutic use , Antimalarials/pharmacology , Plasmodium vivax , Mass Drug Administration , Thailand , Cross-Sectional Studies , Malaria/drug therapy , Malaria, Vivax/drug therapy , Malaria, Vivax/prevention & control
7.
Bioorg Med Chem ; 38: 116110, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33831695

ABSTRACT

In our search for new antibiotic adjuvants as a novel strategy to deal with the emergence of multi-drug resistant (MDR) bacteria, a series of succinylprimaquine-polyamine (SPQ-PA) conjugates and derivatives of a cationic amphiphilic nature have been prepared. Evaluation of these primaquine conjugates for intrinsic antimicrobial properties and the ability to restore the antibiotic activity of doxycycline identified two derivatives, SPQ-PA3-8-3 and SPQ-PA3-10-3 that exhibited intrinsic activity against the Gram-positive bacteria Staphylococcus aureus and the yeast Cryptococcus neoformans. None of the analogues were active against the Gram-negative bacterium Pseudomonas aeruginosa. However, in the presence of a sub-therapeutic amount of doxycycline (4.5 µM), both SPQ-PA3-4-3 and SPQ-PA3-10-3 compounds displayed potent antibiotic adjuvant properties against P. aeruginosa, with MIC's of 6.25 µM. A series of derivatives were prepared to investigate the structure-activity relationship that explored the influence of both a simplified aryl lipophilic substituent and variation of the length of the polyamine scaffold on observed intrinsic antimicrobial properties and the ability to potentiate the action of doxycycline against P. aeruginosa.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Anti-Bacterial Agents/pharmacology , Gram-Positive Bacteria/drug effects , Polyamines/pharmacology , Primaquine/pharmacology , Adjuvants, Pharmaceutic/chemical synthesis , Adjuvants, Pharmaceutic/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Polyamines/chemistry , Primaquine/chemical synthesis , Primaquine/chemistry , Structure-Activity Relationship
8.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884765

ABSTRACT

Triple-negative breast cancer (TNBC) cells overexpress the epidermal growth factor receptor (EGFR). Nuclear EGFR (nEGFR) drives resistance to anti-EGFR therapy and is correlated with poor survival in breast cancer. Inhibition of EGFR nuclear translocation may be a reasonable approach for the treatment of TNBC. The anti-malarial drugs chloroquine and primaquine have been shown to promote an anticancer effect. The aim of the present study was to investigate the effect and mechanism of chloroquine- and primaquine-induced apoptosis of breast cancer cells. We showed that primaquine, a malaria drug, inhibits the growth, migration, and colony formation of breast cancer cells in vitro, and inhibits tumor growth in vivo. Primaquine induces damage to early endosomes and inhibits the nuclear translocation of EGFR. Primaquine inhibits the interaction of Stat3 and nEGFR and reduces the transcript and protein levels of c-Myc. Moreover, primaquine and chloroquine induce the apoptosis of breast cancer cells through c-Myc/Bcl-2 downregulation, induce early endosome damage and reduce nEGFR levels, and induce apoptosis in breast cancer through nEGFR/Stat3-dependent c-Myc downregulation. Our study of primaquine and chloroquine provides a rationale for targeting EGFR signaling components in the treatment of breast cancer.


Subject(s)
Apoptosis/physiology , Primaquine/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , STAT3 Transcription Factor/metabolism , Triple Negative Breast Neoplasms/drug therapy , Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chloroquine/pharmacology , Down-Regulation , Drug Repositioning , Endosomes/metabolism , ErbB Receptors/metabolism , Humans , Protein Transport/drug effects , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/pathology
9.
Molecules ; 26(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34208832

ABSTRACT

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


Subject(s)
Antimalarials , Erythrocytes/parasitology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/growth & development , Primaquine , Tetraoxanes , Adult , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antimalarials/pharmacology , Erythrocytes/metabolism , Erythrocytes/pathology , Female , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/pathology , Male , Middle Aged , Primaquine/analogs & derivatives , Primaquine/chemical synthesis , Primaquine/chemistry , Primaquine/pharmacology , Tetraoxanes/chemical synthesis , Tetraoxanes/chemistry , Tetraoxanes/pharmacology
10.
Article in English | MEDLINE | ID: mdl-33077656

ABSTRACT

For a long while, 8-aminoquinoline compounds have been the only therapeutic agents against latent hepatic malaria parasites. These have poor activity against the blood-stage plasmodia causing acute malaria and must be used in conjunction with partner blood schizontocidal agents. We examined the impacts of one such agent, chloroquine, upon the activity of primaquine, an 8-aminoquinoline, against hepatic stages of Plasmodium cynomolgi, Plasmodium yoelii, Plasmodium berghei, and Plasmodium falciparum within several ex vivo systems-primary hepatocytes of Macaca fascicularis, primary human hepatocytes, and stably transformed human hepatocarcinoma cell line HepG2. Primaquine exposures to formed hepatic schizonts and hypnozoites of P. cynomolgi in primary simian hepatocytes exhibited similar 50% inhibitory concentration (IC50) values near 0.4 µM, whereas chloroquine in the same system exhibited no inhibitory activities. Combining chloroquine and primaquine in this system decreased the observed primaquine IC50 for all parasite forms in a chloroquine dose-dependent manner by an average of 18-fold. Chloroquine also decreased the primaquine IC50 against hepatic P. falciparum in primary human hepatocytes, P. berghei in simian primary hepatocytes, and P. yoelii in primary human hepatocytes. Chloroquine had no impact on primaquine IC50 against P. yoelii in HepG2 cells and, likewise, had no impact on the IC50 of atovaquone (hepatic schizontocide) against P. falciparum in human hepatocytes. We describe important sources of variability in the potentiation of primaquine activity by chloroquine in these systems. Chloroquine potentiated primaquine activity against hepatic forms of several plasmodia. We conclude that chloroquine specifically potentiated 8-aminoquinoline activities against active and dormant hepatic-stage plasmodia in normal primary hepatocytes but not in a hepatocarcinoma cell line.


Subject(s)
Antimalarials , Malaria , Plasmodium , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Chloroquine/pharmacology , Chloroquine/therapeutic use , Humans , Malaria/drug therapy , Primaquine/pharmacology , Primaquine/therapeutic use
11.
Nat Immunol ; 9(5): 558-66, 2008 May.
Article in English | MEDLINE | ID: mdl-18376402

ABSTRACT

Antiviral or antitumor immunity requires activation of cytotoxic CD8+ T cells by dendritic cells, which present viral or tumor antigens on major histocompatibility complex (MHC) class I molecules. The intracellular mechanisms facilitating MHC class I-restricted presentation of extracellular antigens ('cross-presentation') are unclear. Here we demonstrate that cross-presentation of soluble antigen occurred in an early endosomal compartment distinct from the endoplasmic reticulum where endogenous antigen is loaded onto MHC class I. Efficient cross-presentation required endotoxin-induced, Toll-like receptor 4- and signaling molecule MyD88-dependent relocation of the transporter associated with antigen processing, essential for loading of MHC class I, to early endosomes. Transport of cross-presented antigen from endosomes to the cell surface was inhibited by primaquine, which blocks endosomal trafficking. Thus, cross-presentation is spatially and mechanistically separated from endogenous MHC class I-restricted antigen presentation and is biased toward antigens containing microbial molecular patterns.


Subject(s)
Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/immunology , ATP-Binding Cassette Transporters/metabolism , Animals , Antigen Presentation , Antigens/immunology , Antigens/metabolism , Cross-Priming , Dendritic Cells/metabolism , Endosomes/metabolism , Histocompatibility Antigens Class I/metabolism , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Mannose Receptor , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Ovalbumin/immunology , Ovalbumin/metabolism , Primaquine/pharmacology , Protein Subunits/immunology , Protein Transport/drug effects , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Solubility , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 4/immunology
12.
J Autoimmun ; 114: 102505, 2020 11.
Article in English | MEDLINE | ID: mdl-32595012

ABSTRACT

Multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) are neuroinflammatory conditions where inflammatory CD4+ T cells play a major role. Forkhead box P3 (Foxp3)+ regulatory T (Treg) cells suppress inflammation and an increase in their numbers and activity is beneficial for MS and EAE. However, studies have shown that Treg cells can transdifferentiate to pathogenic Th17 cells under inflammatory conditions. Drugs that stimulate Treg cell induction and their resistance to inflammatory stimuli are necessary to develop effective therapies to treat MS. Here, we show that primaquine (PQ), an anti-malarial drug, suppresses EAE through the stimulation of Foxp3+ Treg cells. PQ-elicited Treg cells are refractory to inflammatory stimuli and suppress EAE. Additionally, PQ-elicited Foxp3+ Treg cells were more efficient in suppressing the proliferation of responder cells compared to PBS-elicited Treg cells. Although PQ does not directly induce Foxp3+ Treg cell differentiation from naïve T cells, it modulated dendritic cells (DCs) to induce Foxp3+ Treg cells in an indoleamine 2,3 dioxygenase (IDO)-dependent manner. Together, our results show that PQ elicits Foxp3+ Treg cells with a superior suppressive activity to reduce EAE. PQ has the potential as a safe and effective treatment for MS and other CNS autoimmune inflammatory diseases.


Subject(s)
Forkhead Transcription Factors/metabolism , Primaquine/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antimalarials/pharmacology , Autoimmunity , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Lymphocyte Activation/immunology , Mice , Multiple Sclerosis/drug therapy , Multiple Sclerosis/etiology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism
13.
J Infect Dis ; 220(3): 448-456, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30882150

ABSTRACT

BACKGROUND: Plasmodium vivax is the main cause of malaria in Nepal. Relapse patterns have not been characterized previously. METHODS: Patients with P. vivax malaria were randomized to receive chloroquine (CQ; 25 mg base/kg given over 3 days) alone or together with primaquine (PQ; 0.25 mg base/kg/day for 14 days) and followed intensively for 1 month, then at 1- to 2-month intervals for 1 year. Parasite isolates were genotyped. RESULTS: One hundred and one (49%) patients received CQ and 105 (51%) received CQ + PQ. In the CQ + PQ arm, there were 3 (4.1%) recurrences in the 73 patients who completed 1 year of follow-up compared with 22 of 78 (28.2%) in the CQ-only arm (risk ratio, 0.146 [95% confidence interval, .046-.467]; P < .0001). Microsatellite genotyping showed relatively high P. vivax genetic diversity (mean heterozygosity, 0.843 [range 0.570-0.989] with low multiplicity of infection (mean, 1.05) reflecting a low transmission preelimination setting. Of the 12 genetically homologous relapses, 5 (42%) occurred in a cluster after 9 months, indicating long latency. CONCLUSIONS: Although there may be emerging CQ resistance, the combination of CQ and the standard-dose 14-day PQ regimen is highly efficacious in providing radical cure of short- and long-latency P. vivax malaria in Nepal.


Subject(s)
Antimalarials/pharmacology , Malaria, Vivax/drug therapy , Malaria, Vivax/prevention & control , Plasmodium vivax/drug effects , Primaquine/pharmacology , Adolescent , Adult , Chloroquine/pharmacology , Chronic Disease/drug therapy , Chronic Disease/prevention & control , Drug Therapy, Combination/methods , Female , Follow-Up Studies , Genotype , Humans , Longitudinal Studies , Male , Middle Aged , Nepal , Prospective Studies , Recurrence , Young Adult
14.
Clin Infect Dis ; 69(8): 1436-1439, 2019 09 27.
Article in English | MEDLINE | ID: mdl-30753355

ABSTRACT

Gametocyte density and sex ratio can predict the proportion of mosquitoes that will become infected after feeding on blood of patients receiving nongametocytocidal drugs. Because primaquine and methylene blue sterilize gametocytes before affecting their density and sex ratio, mosquito feeding experiments are required to demonstrate their early transmission-blocking effects.


Subject(s)
Antimalarials/pharmacology , Culicidae/physiology , Malaria, Falciparum/prevention & control , Methylene Blue/pharmacology , Plasmodium falciparum/drug effects , Primaquine/pharmacology , Adolescent , Adult , Animals , Child , Child, Preschool , Culicidae/parasitology , Female , Humans , Malaria, Falciparum/parasitology , Male , Mali , Middle Aged , Sex Ratio , Sterilization , Young Adult
15.
Article in English | MEDLINE | ID: mdl-31383656

ABSTRACT

Single-dose primaquine (PQ) clears mature gametocytes and reduces the transmission of Plasmodium falciparum after artemisinin combination therapy. Genetic variation in CYP2D6, the gene producing the drug-metabolizing enzyme cytochrome P450 2D6 (CYP2D6), influences plasma concentrations of PQ and its metabolites and is associated with PQ treatment failure in Plasmodium vivax malaria. Using blood and saliva samples of varying quantity and quality from 8 clinical trials across Africa (n = 1,076), we were able to genotype CYP2D6 for 774 samples (72%). We determined whether genetic variation in CYP2D6 has implications for PQ efficacy in individuals with gametocytes at the time of PQ administration (n = 554) and for safety in glucose-6-phosphate dehydrogenase (G6PD)-deficient individuals treated with PQ (n = 110). Individuals with a genetically inferred CYP2D6 poor/intermediate metabolizer status had a higher gametocyte prevalence on day 7 or 10 after PQ than those with an extensive/ultrarapid CYP2D6 metabolizer status (odds ratio [OR] = 1.79 [95% confidence interval {CI}, 1.10, 2.90]; P = 0.018). The mean minimum hemoglobin concentrations during follow-up for G6PD-deficient individuals were 11.8 g/dl for CYP2D6 extensive/ultrarapid metabolizers and 12.1 g/dl for CYP2D6 poor/intermediate metabolizers (P = 0. 803). CYP2D6 genetically inferred metabolizer status was also not associated with anemia following PQ treatment (P = 0.331). We conclude that CYP2D6 poor/intermediate metabolizer status may be associated with prolonged gametocyte carriage after treatment with single-low-dose PQ but not with treatment safety.


Subject(s)
Antimalarials/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Glucosephosphate Dehydrogenase Deficiency/drug therapy , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Polymorphism, Genetic , Primaquine/pharmacokinetics , Adult , Africa , Antimalarials/blood , Antimalarials/pharmacology , Artemether, Lumefantrine Drug Combination/administration & dosage , Artemisinins/administration & dosage , Child , Cytochrome P-450 CYP2D6/deficiency , Drug Administration Schedule , Female , Gene Expression , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase Deficiency/blood , Glucosephosphate Dehydrogenase Deficiency/parasitology , Humans , Life Cycle Stages/drug effects , Life Cycle Stages/physiology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Male , Patient Safety , Plasmodium falciparum/physiology , Primaquine/blood , Primaquine/pharmacology , Quinolines/administration & dosage , Treatment Outcome
16.
Malar J ; 18(1): 201, 2019 Jun 19.
Article in English | MEDLINE | ID: mdl-31217011

ABSTRACT

BACKGROUND: The challenge in anti-malarial chemotherapy is based on the emergence of resistance to drugs and the search for medicines against all stages of the life cycle of Plasmodium spp. as a therapeutic target. Nowadays, many molecules with anti-malarial activity are reported. However, few studies about the cellular and molecular mechanisms to understand their mode of action have been explored. Recently, new primaquine-based hybrids as new molecules with potential multi-acting anti-malarial activity were reported and two hybrids of primaquine linked to quinoxaline 1,4-di-N-oxide (PQ-QdNO) were identified as the most active against erythrocytic, exoerythrocytic and sporogonic stages. METHODS: To further understand the anti-malarial mode of action (MA) of these hybrids, hepg2-CD81 were infected with Plasmodium yoelii 17XNL and treated with PQ-QdNO hybrids during 48 h. After were evaluated the production of ROS, the mitochondrial depolarization, the total glutathione content, the DNA damage and proteins related to oxidative stress and death cell. RESULTS: In a preliminary analysis as tissue schizonticidals, these hybrids showed a mode of action dependent on peroxides production, but independent of the activation of transcription factor p53, mitochondrial depolarization and arrest cell cycle. CONCLUSIONS: Primaquine-quinoxaline 1,4-di-N-oxide hybrids exert their antiplasmodial activity in the exoerythrocytic phase by generating high levels of oxidative stress which promotes the increase of total glutathione levels, through oxidation stress sensor protein DJ-1. In addition, the role of HIF1a in the mode of action of quinoxaline 1,4-di-N-oxide is independent of biological activity.


Subject(s)
Antimalarials/pharmacology , Plasmodium yoelii/drug effects , Primaquine/pharmacology , Quinoxalines/pharmacology , Drug Combinations , Erythrocytes/parasitology , Hep G2 Cells , Humans , Sporozoites/drug effects
17.
Malar J ; 18(1): 325, 2019 Sep 23.
Article in English | MEDLINE | ID: mdl-31547827

ABSTRACT

BACKGROUND: Activation of hypnozoites of vivax malaria causes multiple clinical relapses, which contribute to the Plasmodium vivax burden and continuing transmission. Artemisinin-based combination therapy (ACT) is effective against blood-stage P. vivax but requires co-administration with primaquine to achieve radical cure. The therapeutic efficacy of primaquine depends on the generation of a therapeutically active metabolite via cytochrome P450 2D6 (CYP2D6). Impaired CYP2D6 metabolism has been associated with primaquine treatment failure. This study investigated the association between impaired CYP2D6 genotypes, drug-exposure to the long-acting ACT component (schizonticidal drugs) and tolerance and efficacy. METHODS: Adult patients with acute vivax malaria were enrolled in a recently completed trial and treated with artesunate-mefloquine, chloroquine or artemether-lumefantrine. All received concomitant primaquine (0.5 mg/kg/day for 7-9 days). The association between efficacy and safety and drug exposure was explored using area-under-the-curve (AUC) and half-life (t1/2) estimates obtained by non-compartmental analysis of the long half-life drugs. Parasite recurrences by day 63 were categorized as related relapses or re-infections/unrelated hypnozoite activation by genotyping three microsatellite loci and two polymorphic loci of merozoite surface antigen-1. The CYP2D6 genotype was identified with Taqman assays by real-time PCR to 9 polymorphisms (8 SNPs and one deletion). Impaired CYP2D6 activity was inferred using the Activity Score System. RESULTS: Most recurrences in the ASMQ (67%), CQ (80%) and AL (85%) groups were considered related relapses. Eight of nine (88.9%) of the patients with impaired CYP2D6 activity relapsed with related parasite compared to 18/25 (72%) with normal activity (RR = 1.23, 0.88; 1.72, p = 0.40). There were no associations between the measured PK parameters and recurrence. Patients with longer chloroquine half-lives had more pruritus (RR = 1.09, 1.03; 1.14, p = 0.001). Higher CQ AUCs were associated with reduced falls in haemoglobin by day 14 (Coef - 0.02, - 0.005; - 0.03, p = 0.01). All regimens were well tolerated. CONCLUSION: Genotyping of P. vivax showed that activation of related (homologous) hypnozoites was the most frequent cause of recurrence. The high proportion of the impaired CYP2D6 activity among patients with recurrent infections suggests that slow primaquine metabolism might influence related relapse rates in Brazil among patients receiving primaquine for radical cure, although confirmatory studies are needed. There was no association between drug exposure of the long-acting ACT component (schizonticidal drugs) and risk of related relapse. ACT was well tolerated. These results provide further re-assurance about the safety and efficacy of ACT when combined with short course primaquine to treat uncomplicated malaria vivax in Brazil. Trial registration RBR-79s56s ( http://www.ensaiosclinicos.gov.br/rg/RBR-79s56s/ ).


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Chloroquine/pharmacology , Primaquine/pharmacology , Adult , Aged , Antimalarials/pharmacokinetics , Artemisinins/pharmacokinetics , Brazil , Chloroquine/pharmacokinetics , Drug Combinations , Female , Humans , Male , Middle Aged , Primaquine/pharmacokinetics , Young Adult
18.
Bioorg Med Chem Lett ; 29(19): 126614, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31431364

ABSTRACT

Primaquine homodimers, e.g. symmetric PQ-diamides of dicarboxylic acids containing 4 to 8 carbon atoms, were evaluated against Plasmodium berghei hepatic stages and P. falciparum blood stages, as well as against three cancer cell lines. Novel PQ-homodimers exerted much higher activity against hepatic stages, but less pronounced activity against blood stages in comparison to the parent drug. The submicromolar activity of succinic, fumaric and maleic derivatives against P. berghei was determined (IC50 values: 726.2, 198.1 and 358.4 nM, respectively). Our results indicated that the length and type of spacer between two PQ moieties highly modified the antiproliferative activities of PQ-homodimers. The general antiproliferative activity of the adipic and mesaconic derivatives against three cancer cell lines (MCF-7, HCT116, H 460) was observed (GI50 = 1.78-13.7 and 2.36-4.31 µM, respectively), but adipic derivative was less toxic to human embryonic kidney cells (HEK 293). High selectivity of fumaric and suberic derivatives against breast adenocarcinoma cell line MCF-7 was detected. These two compounds have shown no antiproliferative activity against other tumor cells and HEK 293.


Subject(s)
Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Malaria, Falciparum/drug therapy , Neoplasms/drug therapy , Plasmodium falciparum/drug effects , Primaquine/pharmacology , Antimalarials/chemistry , Antineoplastic Agents/chemistry , Humans , Malaria, Falciparum/pathology , Molecular Structure , Neoplasms/pathology , Primaquine/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
19.
Exp Parasitol ; 197: 93-102, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30562480

ABSTRACT

The intraerythrocytic malaria parasite digests haemoglobin to provide amino acids for metabolism and releases toxic haem that is sequestered into haemozoin, a non-toxic, insoluble, crystalline pigment. Following erythrocyte rupture, haemozoin is released into circulation and phagocytosed by monocytes. Phagocytosed haemozoin and antimalarial drugs have both been reported to modulate monocyte functions. This study determined the effects of therapeutic concentrations of seven antimalarial drugs; amodiaquine, artemisinin, chloroquine, doxycycline, primaquine, pyrimethamine and quinine, on the phagocytosis of ß-haematin (synthetic haemozoin) by two monocytic cell lines, J774A.1 and U937, and human peripheral blood mononuclear cells. A novel spectrophotometric method based on the absorbance (O.D 400 nm) of alkali/SDS treated monocytes containing ß-haematin was developed to complement counting phagocytosis with microscopy. The method has potential use for the large scale screening of monocyte phagocytic activity. Artemisinin, quinine, primaquine and pyrimethamine activated ß-haematin phagocytosis by 12% or more, whereas amodiaquine, chloroquine and doxycyline inhibited ß-haematin phagocytosis. In contrast, antimalarial drugs had minimal inhibitory effects on the phagocytosis of latex beads with only quinine resulting in more than 20% inhibition. Antimalarial drugs appear to alter monocyte phagocytic activity which has implications for the treatment, pathogenicity and adjunct therapies for malaria.


Subject(s)
Antimalarials/pharmacology , Hemeproteins/metabolism , Monocytes/drug effects , Phagocytosis/drug effects , Amodiaquine/pharmacology , Animals , Artemisinins/pharmacology , Cell Count , Cell Line , Chloroquine/pharmacology , Doxycycline/pharmacology , Electron Probe Microanalysis , Heme/analysis , Hemeproteins/biosynthesis , Hemeproteins/chemistry , Hemeproteins/ultrastructure , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Monocytes/enzymology , Monocytes/metabolism , Peroxidase/isolation & purification , Peroxidase/metabolism , Primaquine/pharmacology , Pyrimethamine/pharmacology , Quinine/pharmacology , Spectrophotometry , Temperature , U937 Cells
20.
Molecules ; 24(24)2019 Dec 12.
Article in English | MEDLINE | ID: mdl-31842498

ABSTRACT

Recently, we disclosed primaquine cell penetrating peptide conjugates that were more potent than parent primaquine against liver stage Plasmodium parasites and non-toxic to hepatocytes. The same strategy was now applied to the blood-stage antimalarial chloroquine, using a wide set of peptides, including TP10, a cell penetrating peptide with intrinsic antiplasmodial activity. Chloroquine-TP10 conjugates displaying higher antiplasmodial activity than the parent TP10 peptide were identified, at the cost of an increased hemolytic activity, which was further confirmed for their primaquine analogues. Fluorescence microscopy and flow cytometry suggest that these drug-peptide conjugates strongly bind, and likely destroy, erythrocyte membranes. Taken together, the results herein reported put forward that coupling antimalarial aminoquinolines to cell penetrating peptides delivers hemolytic conjugates. Hence, despite their widely reported advantages as carriers for many different types of cargo, from small drugs to biomacromolecules, cell penetrating peptides seem unsuitable for safe intracellular delivery of antimalarial aminoquinolines due to hemolysis issues. This highlights the relevance of paying attention to hemolytic effects of cell penetrating peptide-drug conjugates.


Subject(s)
Antimalarials , Cell-Penetrating Peptides , Chloroquine , Erythrocytes/parasitology , Plasmodium falciparum/growth & development , Primaquine , Recombinant Fusion Proteins , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antimalarials/pharmacology , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Chloroquine/chemistry , Chloroquine/pharmacology , Erythrocytes/metabolism , Humans , Primaquine/chemistry , Primaquine/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL