10 results on '"Plasmodium cynomolgi drug effects"'
Search Results
2. Azithromycin disrupts apicoplast biogenesis in replicating and dormant liver stages of the relapsing malaria parasites Plasmodium vivax and Plasmodium cynomolgi.
- Author
-
Amanzougaghene N, Tajeri S, Franetich JF, Ashraf K, Soulard V, Bigeard P, Guindo CO, Bouillier C, Lemaitre J, Relouzat F, Legrand R, Kocken CHM, Zeeman AM, Roobsoong W, Sattabongkot J, Yang Z, Snounou G, and Mazier D
- Subjects
- Animals, Hepatocytes parasitology, Hepatocytes drug effects, Humans, Organelle Biogenesis, Malaria, Vivax parasitology, Malaria, Vivax drug therapy, Mice, Malaria parasitology, Malaria drug therapy, Azithromycin pharmacology, Plasmodium vivax drug effects, Plasmodium cynomolgi drug effects, Antimalarials pharmacology, Liver parasitology, Liver drug effects, Apicoplasts drug effects
- Abstract
The control and elimination of malaria caused by Plasmodium vivax is hampered by the threat of relapsed infection resulting from the activation of dormant hepatic hypnozoites. Currently, only the 8-aminoquinolines, primaquine and tafenoquine, have been approved for the elimination of hypnozoites, although their use is hampered by potential toxicity. Therefore, an alternative radical curative drug that safely eliminates hypnozoites is a pressing need. This study assessed the potential hypnozoiticidal activity of the antibiotic azithromycin, which is thought to exert antimalarial activity by inhibiting prokaryote-like ribosomal translation within the apicoplast, an indispensable organelle. The results show that azithromycin inhibited apicoplast development during liver-stage schizogony in P. vivax and Plasmodium cynomolgi, leading to impaired parasite maturation. More importantly, this study found that azithromycin is likely to impair the hypnozoite's apicoplast, resulting in the loss of this organelle. Subsequently, using a recently developed long-term hepatocyte culture system, this study found that this loss likely induces a delay in the hypnozoite activation rate, and that those parasites that do proceed to schizogony display liver-stage arrest prior to differentiating into hepatic merozoites, thus potentially preventing relapse. Overall, this work provides evidence for the potential use of azithromycin for the radical cure of relapsing malaria, and identifies apicoplast functions as potential drug targets in quiescent hypnozoites., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. A New Thienopyrimidinone Chemotype Shows Multistage Activity against Plasmodium falciparum, Including Artemisinin-Resistant Parasites.
- Author
-
Bosson-Vanga H, Primas N, Franetich JF, Lavazec C, Gomez L, Ashraf K, Tefit M, Soulard V, Dereuddre-Bosquet N, Le Grand R, Donnette M, Mustière R, Amanzougaghene N, Tajeri S, Suzanne P, Malzert-Fréon A, Rault S, Vanelle P, Hutter S, Cohen A, Snounou G, Roques P, Azas N, Lagardère P, Lisowski V, Masurier N, Nguyen M, Paloque L, Benoit-Vical F, Verhaeghe P, and Mazier D
- Subjects
- Animals, Antimalarials chemistry, Artemisinins pharmacology, Cell Line, Tumor, Disease Models, Animal, Dogs, Drug Resistance physiology, Female, Hep G2 Cells, Humans, Liver parasitology, Macaca fascicularis, Madin Darby Canine Kidney Cells, Male, Mice, Mice, Inbred BALB C, Pyrimidinones chemistry, Antimalarials pharmacology, Malaria, Falciparum drug therapy, Plasmodium cynomolgi drug effects, Plasmodium falciparum drug effects, Plasmodium yoelii drug effects, Pyrimidinones pharmacology
- Abstract
Human malaria infection begins with a one-time asymptomatic liver stage followed by a cyclic symptomatic blood stage. For decades, the research for novel antimalarials focused on the high-throughput screening of molecules that only targeted the asexual blood stages. In a search for new effective compounds presenting a triple action against erythrocytic and liver stages in addition to the ability to block the transmission of the disease via the mosquito vector, 2-amino-thienopyrimidinone derivatives were synthesized and tested for their antimalarial activity. One molecule, named gamhepathiopine (denoted as "M1" herein), was active at submicromolar concentrations against both erythrocytic (50% effective concentration [EC
50 ] = 0.045 μM) and liver (EC50 = 0.45 μM) forms of Plasmodium falciparum. Furthermore, gamhepathiopine efficiently blocked the development of the sporogonic cycle in the mosquito vector by inhibiting the exflagellation step. Moreover, M1 was active against artemisinin-resistant forms (EC50 = 0.227 μM), especially at the quiescent stage. Nevertheless, in mice, M1 showed modest activity due to its rapid metabolization by P450 cytochromes into inactive derivatives, calling for the development of new parent compounds with improved metabolic stability and longer half-lives. These results highlight the thienopyrimidinone scaffold as a novel antiplasmodial chemotype of great interest to search for new drug candidates displaying multistage activity and an original mechanism of action with the potential to be used in combination therapies for malaria elimination in the context of artemisinin resistance. IMPORTANCE This work reports a new chemical structure that (i) displays activity against the human malaria parasite Plasmodium falciparum at 3 stages of the parasitic cycle (blood stage, hepatic stage, and sexual stages), (ii) remains active against parasites that are resistant to the first-line treatment recommended by the World Health Organization (WHO) for the treatment of severe malaria (artemisinins), and (iii) reduces transmission of the parasite to the mosquito vector in a mouse model. This new molecule family could open the way to the conception of novel antimalarial drugs with an original multistage mechanism of action to fight against Plasmodium drug resistance and block interhuman transmission of malaria.- Published
- 2021
- Full Text
- View/download PDF
4. Safety, Pharmacokinetics, and Activity of High-Dose Ivermectin and Chloroquine against the Liver Stage of Plasmodium cynomolgi Infection in Rhesus Macaques.
- Author
-
Vanachayangkul P, Im-Erbsin R, Tungtaeng A, Kodchakorn C, Roth A, Adams J, Chaisatit C, Saingam P, Sciotti RJ, Reichard GA, Nolan CK, Pybus BS, Black CC, Lugo-Roman LA, Wegner MD, Smith PL, Wojnarski M, Vesely BA, and Kobylinski KC
- Subjects
- Animals, Antimalarials blood, Antimalarials pharmacokinetics, Biological Availability, Chloroquine blood, Chloroquine pharmacokinetics, Drug Administration Schedule, Drug Combinations, Drug Synergism, Female, Hepatocytes drug effects, Hepatocytes parasitology, Ivermectin blood, Ivermectin pharmacokinetics, Liver parasitology, Macaca mulatta, Malaria parasitology, Male, Parasitemia drug therapy, Plasmodium cynomolgi growth & development, Plasmodium cynomolgi pathogenicity, Primary Cell Culture, Schizonts drug effects, Schizonts growth & development, Antimalarials pharmacology, Chloroquine pharmacology, Ivermectin pharmacology, Liver drug effects, Malaria drug therapy, Plasmodium cynomolgi drug effects
- Abstract
Previously, ivermectin (1 to 10 mg/kg of body weight) was shown to inhibit the liver-stage development of Plasmodium berghei in orally dosed mice. Here, ivermectin showed inhibition of the in vitro development of Plasmodium cynomolgi schizonts (50% inhibitory concentration [IC
50 ], 10.42 μM) and hypnozoites (IC50 , 29.24 μM) in primary macaque hepatocytes when administered as a high dose prophylactically but not when administered in radical cure mode. The safety, pharmacokinetics, and efficacy of oral ivermectin (0.3, 0.6, and 1.2 mg/kg) with and without chloroquine (10 mg/kg) administered for 7 consecutive days were evaluated for prophylaxis or radical cure of P. cynomolgi liver stages in rhesus macaques. No inhibition or delay to blood-stage P. cynomolgi parasitemia was observed at any ivermectin dose (0.3, 0.6, and 1.2 mg/kg). Ivermectin (0.6 and 1.2 mg/kg) and chloroquine (10 mg/kg) in combination were well-tolerated with no adverse events and no significant pharmacokinetic drug-drug interactions observed. Repeated daily ivermectin administration for 7 days did not inhibit ivermectin bioavailability. It was recently demonstrated that both ivermectin and chloroquine inhibit replication of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro Further ivermectin and chloroquine trials in humans are warranted to evaluate their role in Plasmodium vivax control and as adjunctive therapies against COVID-19 infections., (Copyright © 2020 Vanachayangkul et al.)- Published
- 2020
- Full Text
- View/download PDF
5. Tafenoquine: A Step toward Malaria Elimination.
- Author
-
Lu KY and Derbyshire ER
- Subjects
- Aminoquinolines adverse effects, Anemia, Hemolytic chemically induced, Animals, Antimalarials adverse effects, Cytochrome P-450 CYP2D6 genetics, Cytochrome P-450 CYP2D6 metabolism, Gene Knockdown Techniques, Glucosephosphate Dehydrogenase Deficiency metabolism, Haplorhini, Humans, Methemoglobinemia chemically induced, Mice, Plasmodium cynomolgi drug effects, Plasmodium vivax drug effects, Zebrafish, Aminoquinolines therapeutic use, Antimalarials therapeutic use, Malaria, Vivax drug therapy
- Abstract
There is a pressing need for compounds with broad-spectrum activity against malaria parasites at various life cycle stages to achieve malaria elimination. However, this goal cannot be accomplished without targeting the tenacious dormant liver-stage hypnozoite that causes multiple relapses after the first episode of illness. In the search for the magic bullet to radically cure Plasmodium vivax malaria, tafenoquine outperformed other candidate drugs and was approved by the U.S. Food and Drug Administration in 2018. Tafenoquine is an 8-aminoquinoline that inhibits multiple life stages of various Plasmodium species. Additionally, its much longer half-life allows for single-dose treatment, which will improve the compliance rate. Despite its approval and the long-time use of other 8-aminoquinolines, the mechanisms behind tafenoquine's activity and adverse effects are still largely unknown. In this Perspective, we discuss the plausible underlying mechanisms of tafenoquine's antiparasitic activity and highlight its role as a cellular stressor. We also discuss potential drug combinations and the development of next-generation 8-aminoquinolines to further improve the therapeutic index of tafenoquine for malaria treatment and prevention.
- Published
- 2020
- Full Text
- View/download PDF
6. Substituted Aminoacetamides as Novel Leads for Malaria Treatment.
- Author
-
Norcross NR, Wilson C, Baragaña B, Hallyburton I, Osuna-Cabello M, Norval S, Riley J, Fletcher D, Sinden R, Delves M, Ruecker A, Duffy S, Meister S, Antonova-Koch Y, Crespo B, de Cózar C, Sanz LM, Gamo FJ, Avery VM, Frearson JA, Gray DW, Fairlamb AH, Winzeler EA, Waterson D, Campbell SF, Willis PA, Read KD, and Gilbert IH
- Subjects
- Acetamides chemical synthesis, Acetamides pharmacokinetics, Animals, Antimalarials chemical synthesis, Antimalarials pharmacokinetics, Humans, Mice, Microsomes, Liver metabolism, Molecular Structure, Parasitic Sensitivity Tests, Plasmodium berghei drug effects, Plasmodium cynomolgi drug effects, Plasmodium falciparum drug effects, Structure-Activity Relationship, Acetamides pharmacology, Antimalarials pharmacology
- Abstract
Herein we describe the optimization of a phenotypic hit against Plasmodium falciparum based on an aminoacetamide scaffold. This led to N-(3-chloro-4-fluorophenyl)-2-methyl-2-{[4-methyl-3-(morpholinosulfonyl)phenyl]amino}propanamide (compound 28) with low-nanomolar activity against the intraerythrocytic stages of the malaria parasite, and which was found to be inactive in a mammalian cell counter-screen up to 25 μm. Inhibition of gametes in the dual gamete activation assay suggests that this family of compounds may also have transmission blocking capabilities. Whilst we were unable to optimize the aqueous solubility and microsomal stability to a point at which the aminoacetamides would be suitable for in vivo pharmacokinetic and efficacy studies, compound 28 displayed excellent antimalarial potency and selectivity; it could therefore serve as a suitable chemical tool for drug target identification., (© 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
7. Differential activity of methylene blue against erythrocytic and hepatic stages of Plasmodium.
- Author
-
Bosson-Vanga H, Franetich JF, Soulard V, Sossau D, Tefit M, Kane B, Vaillant JC, Borrmann S, Müller O, Dereuddre-Bosquet N, Le Grand R, Silvie O, and Mazier D
- Subjects
- Animals, Anopheles parasitology, Erythrocytes parasitology, Female, Liver parasitology, Mice parasitology, Mice, Inbred BALB C, Antimalarials pharmacology, Methylene Blue pharmacology, Plasmodium cynomolgi drug effects, Plasmodium falciparum drug effects, Plasmodium yoelii drug effects
- Abstract
Background: In the context of malaria elimination/eradication, drugs that are effective against the different developmental stages of the parasite are highly desirable. The oldest synthetic anti-malarial drug, the thiazine dye methylene blue (MB), is known for its activity against Plasmodium blood stages, including gametocytes. The aim of the present study was to investigate a possible effect of MB against malaria parasite liver stages., Methods: MB activity was investigated using both in vitro and in vivo models. In vitro assays consisted of testing MB activity on Plasmodium falciparum, Plasmodium cynomolgi and Plasmodium yoelii parasites in human, simian or murine primary hepatocytes, respectively. MB in vivo activity was evaluated using intravital imaging in BALB/c mice infected with a transgenic bioluminescent P. yoelii parasite line. The transmission-blocking activity of MB was also addressed using mosquitoes fed on MB-treated mice., Results: MB shows no activity on Plasmodium liver stages, including hypnozoites, in vitro in primary hepatocytes. In BALB/c mice, MB has moderate effect on P. yoelii hepatic development but is highly effective against blood stage growth. MB is active against gametocytes and abrogates parasite transmission from mice to mosquitoes., Conclusion: While confirming activity of MB against both sexual and asexual blood stages, the results indicate that MB has only little activity on the development of the hepatic stages of malaria parasites.
- Published
- 2018
- Full Text
- View/download PDF
8. Synthesis of primaquine glyco-conjugates as potential tissue schizontocidal antimalarial agents.
- Author
-
Azad CS, Saxena M, Siddiqui AJ, Bhardwaj J, Puri SK, Dutta GP, Anand N, and Saxena AK
- Subjects
- Animals, Antimalarials chemical synthesis, Antimalarials pharmacology, Female, Glycoconjugates chemical synthesis, Glycoconjugates chemistry, Glycoconjugates pharmacology, Glycoconjugates therapeutic use, Macaca mulatta, Malaria, Vivax drug therapy, Male, Mice, Plasmodium cynomolgi drug effects, Plasmodium vivax drug effects, Primaquine chemical synthesis, Primaquine pharmacology, Antimalarials chemistry, Antimalarials therapeutic use, Malaria drug therapy, Plasmodium drug effects, Primaquine analogs & derivatives, Primaquine therapeutic use
- Abstract
Primaquine (PQ) is the only drug used to prevent relapse of malaria due to P. vivax and P. ovale, by eradicating the dormant liver form of the parasite (hypnozoites). The side-effects associated with PQ limits is uses in treatment of malaria. To overcome the premature oxidative deamination and to increase the life span of drug in the biological system, the novel glyco-conjugates of PQ were synthesized by coupling of primaquine with hexoses in phosphate buffer. The saccharide part of the hybrid molecules thought to direct the drug to the liver, where hypnozoites resides. All the synthesized compounds were fully characterized and evaluated for their radical curative activities. The three compounds viz glucoside (15a), galactoside (15b) and mannoside (15c) with high activity were tested for their activity in rhesus monkeys where the most active compound 15b showed twofold activity (100% radical curative activity at 1.92 mmol/kg) than the standard drug PQ diphosphate (3.861 mmol/kg). It is proposed that results from these studies may be advantageous to develop a new potent tissue schizonticide antimalarial compound., (© 2017 John Wiley & Sons A/S.)
- Published
- 2017
- Full Text
- View/download PDF
9. Synthesis and Evaluation of Chirally Defined Side Chain Variants of 7-Chloro-4-Aminoquinoline To Overcome Drug Resistance in Malaria Chemotherapy.
- Author
-
Dola VR, Soni A, Agarwal P, Ahmad H, Raju KS, Rashid M, Wahajuddin M, Srivastava K, Haq W, Dwivedi AK, Puri SK, and Katti SB
- Subjects
- Administration, Oral, Aminoquinolines pharmacology, Animals, Antimalarials pharmacology, Chlorocebus aethiops, Chloroquine pharmacology, Drug Resistance drug effects, Erythrocytes drug effects, Erythrocytes parasitology, Heme antagonists & inhibitors, Heme metabolism, Hemin antagonists & inhibitors, Hemin biosynthesis, Inhibitory Concentration 50, Macaca mulatta, Malaria parasitology, Malaria, Falciparum drug therapy, Malaria, Falciparum parasitology, Parasitic Sensitivity Tests, Plasmodium cynomolgi growth & development, Plasmodium cynomolgi metabolism, Plasmodium falciparum growth & development, Plasmodium falciparum metabolism, Plasmodium yoelii growth & development, Plasmodium yoelii metabolism, Structure-Activity Relationship, Vero Cells, Aminoquinolines chemical synthesis, Antimalarials chemical synthesis, Malaria drug therapy, Plasmodium cynomolgi drug effects, Plasmodium falciparum drug effects, Plasmodium yoelii drug effects
- Abstract
A novel 4-aminoquinoline derivative [( S )-7-chloro- N -(4-methyl-1-(4-methylpiperazin-1-yl)pentan-2-yl)-quinolin-4-amine triphosphate] exhibiting curative activity against chloroquine-resistant malaria parasites has been identified for preclinical development as a blood schizonticidal agent. The lead molecule selected after detailed structure-activity relationship (SAR) studies has good solid-state properties and promising activity against in vitro and in vivo experimental malaria models. The in vitro absorption, distribution, metabolism, and excretion (ADME) parameters indicate a favorable drug-like profile., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
10. Pharmacokinetics and pharmacodynamics of (+)-primaquine and (-)-primaquine enantiomers in rhesus macaques (Macaca mulatta).
- Author
-
Saunders D, Vanachayangkul P, Imerbsin R, Khemawoot P, Siripokasupkul R, Tekwani BL, Sampath A, Nanayakkara NP, Ohrt C, Lanteri C, Gettyacamin M, Teja-Isavadharm P, and Walker L
- Subjects
- Animals, Antimalarials blood, Antimalarials chemistry, Antimalarials pharmacology, Disease Models, Animal, Drug Administration Schedule, Drug Therapy, Combination, Humans, Kidney drug effects, Liver drug effects, Macaca mulatta, Malaria blood, Malaria parasitology, Malaria, Vivax, Male, Methemoglobin metabolism, Oxidative Stress, Plasmodium cynomolgi growth & development, Plasmodium vivax, Primaquine blood, Primaquine chemistry, Primaquine pharmacology, Recurrence, Stereoisomerism, Antimalarials pharmacokinetics, Chloroquine pharmacology, Malaria drug therapy, Plasmodium cynomolgi drug effects, Primaquine pharmacokinetics
- Abstract
Primaquine (PQ) remains the sole available drug to prevent relapse of Plasmodium vivax malaria more than 60 years after licensure. While this drug was administered as a racemic mixture, prior studies suggested a pharmacodynamic advantage based on differential antirelapse activity and/or toxicities of its enantiomers. Oral primaquine enantiomers prepared using a novel, easily scalable method were given for 7 days to healthy rhesus macaques in a dose-rising fashion to evaluate their effects on the blood, liver, and kidneys. The enantiomers were then administered to Plasmodium cynomolgi-infected rhesus macaques at doses of 1.3 and 0.6 mg/kg of body weight/day in combination with chloroquine. The (-)-PQ enantiomer had higher clearance and apparent volume of distribution than did (+)-PQ and was more extensively converted to the carboxy metabolite. There is evidence for differential oxidative stress with a concentration-dependent rise in methemoglobin (MetHgb) with increasing doses of (+)-PQ greater than that seen for (-)-PQ. There was a marked, reversible hepatotoxicity in 2 of 3 animals dosed with (-)-PQ at 4.5 mg/kg. (-)-PQ in combination with chloroquine was successful in preventing P. cynomolgi disease relapse at doses of 0.6 and 1.3 mg/kg/day, while 1 of 2 animals receiving (+)-PQ at 0.6 mg/kg/day relapsed. While (-)-PQ was also associated with hepatotoxicity at higher doses as seen previously, this has not been identified as a clinical concern in humans during >60 years of use. Limited evidence for increased MetHgb generation with the (+) form in the rhesus macaque model suggests that it may be possible to improve the therapeutic window for hematologic toxicity in the clinic by separating primaquine into its enantiomers., (Copyright © 2014, American Society for Microbiology. All Rights Reserved.)
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.