Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Lancet Infect Dis ; 22(6): 867-878, 2022 06.
Article in English | MEDLINE | ID: mdl-35276064

ABSTRACT

BACKGROUND: Late treatment failures after artemisinin-based combination therapies (ACTs) for falciparum malaria have increased in the Greater Mekong subregion in southeast Asia. Addition of amodiaquine to artemether-lumefantrine could provide an efficacious treatment for multidrug-resistant infections. METHODS: We conducted an open-label, randomised trial at five hospitals or health centres in three locations (western Cambodia, eastern Cambodia, and Vietnam). Eligible participants were male and female patients aged 2-65 years with uncomplicated Plasmodium falciparum malaria. Patients were randomly allocated (1:1 in blocks of eight to 12) to either artemether-lumefantrine alone (dosed according to WHO guidelines) or artemether-lumefantrine plus amodiaquine (10 mg base per kg/day), both given orally as six doses over 3 days. All received a single dose of primaquine (0·25 mg/kg) 24 h after the start of study treatment to limit transmission of the parasite. Parasites were genotyped, identifying artemisinin resistance. The primary outcome was Kaplan-Meier 42-day PCR-corrected efficacy against recrudescence of the original parasite, assessed by intent-to-treat. Safety was a secondary outcome. This completed trial is registered at ClinicalTrials.gov (NCT03355664). FINDINGS: Between March 18, 2018, and Jan 30, 2020, 310 patients received randomly allocated treatment; 154 received artemether-lumefantrine alone and 156 received artemether-lumefantrine plus amodiaquine. Parasites from 305 of these patients were genotyped. 42-day PCR-corrected treatment efficacy was noted in 151 (97%, 95% CI 92-99) of 156 patients with artemether-lumefantrine plus amodiaquine versus 146 (95%, 89-97) of 154 patients with artemether-lumefantrine alone; hazard ratio (HR) for recrudescence 0·6 (95% CI 0·2-1·9, p=0·38). Of the 13 recrudescences, 12 were in 174 (57%) of 305 infections with pfkelch13 mutations indicating artemisinin resistance, for which 42-day efficacy was noted in 89 (96%) of 93 infections with artemether-lumefantrine plus amodiaquine versus 73 (90%) of 81 infections with artemether-lumefantrine alone; HR for recrudescence 0·44 (95% CI 0·14-1·40, p=0·17). Artemether-lumefantrine plus amodiaquine was generally well tolerated, but the number of mild (grade 1-2) adverse events, mainly gastrointestinal, was greater in this group compared with artemether-lumefantrine alone (vomiting, 12 [8%] with artemether-lumefantrine plus amodiaquine vs three [2%] with artemether-lumefantrine alone, p=0·03; and nausea, 11 [7%] with artemether-lumefantrine plus amodiaquine vs three [2%] with artemether-lumefantrine alone, p=0·05). Early vomiting within 1 h of treatment, requiring retreatment, occurred in no patients of 154 with artemether-lumefantrine alone versus five (3%) of 156 with artemether-lumefantrine plus amodiaquine, p=0·06. Bradycardia (≤54 beats/min) of any grade was noted in 59 (38%) of 154 patients with artemether-lumefantrine alone and 95 (61%) of 156 with artemether-lumefantrine plus amodiaquine, p=0·0001. INTERPRETATION: Artemether-lumefantrine plus amodiaquine provides an alternative to artemether-lumefantrine alone as first-line treatment for multidrug-resistant P falciparum malaria in the Greater Mekong subregion, and could prolong the therapeutic lifetime of artemether-lumefantrine in malaria-endemic populations. FUNDING: Bill & Melinda Gates Foundation, Wellcome Trust.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Malaria , Amodiaquine/therapeutic use , Antimalarials/therapeutic use , Artemether/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Artemisinins/therapeutic use , Drug Combinations , Ethanolamines/therapeutic use , Female , Fluorenes/therapeutic use , Humans , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Male , Plasmodium falciparum , Recurrence , Vomiting
2.
Wellcome Open Res ; 6: 142, 2021.
Article in English | MEDLINE | ID: mdl-34381874

ABSTRACT

Higher academic institutions in the UK need to drive improvements in equity, diversity, and inclusion (EDI) through sustainable practical interventions. A broad view of inclusivity is based on an intersectional approach that considers race, geographical location, caring responsibilities, disability, neurodiversity, religion, and LGBTQIA+ identities. We describe the establishment of a diverse stakeholder group to develop practical grass-roots recommendations through which improvements can be advanced. We have developed a manifesto for change, comprising six domains through which academic institutions can drive progress through setting short, medium, and long-term priorities. Interventions will yield rewards in recruitment and retention of a diverse talent pool, leading to enhanced impact and output.

3.
Article in English | MEDLINE | ID: mdl-33139275

ABSTRACT

Resistance to artemisinin-based combination therapy (ACT) in the Plasmodium falciparum parasite is threatening to reverse recent gains in reducing global deaths from malaria. While resistance manifests as delayed parasite clearance in patients, the phenotype can only spread geographically via the sexual stages and mosquito transmission. In addition to their asexual killing properties, artemisinin and its derivatives sterilize sexual male gametocytes. Whether resistant parasites overcome this sterilizing effect has not, however, been fully tested. Here, we analyzed P. falciparum clinical isolates from the Greater Mekong Subregion, each demonstrating delayed clinical clearance and known resistance-associated polymorphisms in the Kelch13 (PfK13var) gene. As well as demonstrating reduced asexual sensitivity to drug, certain PfK13var isolates demonstrated a marked reduction in sensitivity to artemisinin in an in vitro male gamete formation assay. Importantly, this same reduction in sensitivity was observed when the most resistant isolate was tested directly in mosquito feeds. These results indicate that, under artemisinin drug pressure, while sensitive parasites are blocked, resistant parasites continue transmission. This selective advantage for resistance transmission could favor acquisition of additional host-specificity or polymorphisms affecting partner drug sensitivity in mixed infections. Favored resistance transmission under ACT coverage could have profound implications for the spread of multidrug-resistant malaria beyond Southeast Asia.


Subject(s)
Antimalarials , Artemisinins , Culicidae , Malaria, Falciparum , Parasites , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Asia, Southeastern , Drug Resistance/genetics , Humans , Malaria, Falciparum/drug therapy , Male , Plasmodium falciparum/genetics
4.
Front Microbiol ; 10: 2134, 2019.
Article in English | MEDLINE | ID: mdl-31572339

ABSTRACT

BACKGROUND: Despite recent successes at controlling malaria, progress has stalled with an estimated 219 million cases and 435,000 deaths in 2017 alone. Combined with emerging resistance to front line antimalarial therapies in Southeast Asia, there is an urgent need for new treatment options and novel approaches to halt the spread of malaria. Plasmodium, the parasite responsible for malaria propagates through mosquito transmission. This imposes an acute bottleneck on the parasite population and transmission-blocking interventions exploiting this vulnerability are recognized as vital for malaria elimination. METHODS: 13,533 small molecules with known activity against Plasmodium falciparum asexual parasites were screened for additional transmission-blocking activity in an ex vivo Plasmodium berghei ookinete development assay. Active molecules were then counterscreened in dose response against HepG2 cells to determine their activity/cytotoxicity window and selected non-toxic representative molecules were fully profiled in a range of transmission and mosquito infection assays. Furthermore, the entire dataset was compared to other published screens of the same molecules against P. falciparum gametocytes and female gametogenesis. RESULTS: 437 molecules inhibited P. berghei ookinete formation with an IC50 < 10 µM. of which 273 showed >10-fold parasite selectivity compared to activity against HepG2 cells. Active molecules grouped into 49 chemical clusters of three or more molecules, with 25 doublets and 94 singletons. Six molecules representing six major chemical scaffolds confirmed their transmission-blocking activity against P. falciparum male and female gametocytes and inhibited P. berghei oocyst formation in the standard membrane feeding assay at 1 µM. When screening data in the P. berghei development ookinete assay was compared to published screens of the same library in assays against P. falciparum gametocytes and female gametogenesis, it was established that each assay identified distinct, but partially overlapping subsets of transmission-blocking molecules. However, selected molecules unique to each assay show transmission-blocking activity in mosquito transmission assays. CONCLUSION: The P. berghei ookinete development assay is an excellent high throughput assay for efficiently identifying antimalarial molecules targeting early mosquito stage parasite development. Currently no high throughput transmission-blocking assay is capable of identifying all transmission-blocking molecules.

5.
PLoS One ; 14(7): e0219983, 2019.
Article in English | MEDLINE | ID: mdl-31318964

ABSTRACT

BACKGROUND: The Pint of Science festival is the biggest annual international science festival. In May 2017, we coordinated the first Pint of Science festival in Thailand and reported our initial reflections. Building on this work, we set out to evaluate more systematically events conducted in 2018. METHODS: In 2018, we conducted Pint of Science events at four different locations in Bangkok. Overall, there were 18 talks held over six event-days in 2018. We administered 180 self-reported questionnaires as well as conducted 11 semi-structured interviews and a focus group discussion with audience members and speakers. RESULTS: Of the 180 questionnaires handed out, 125 attendees completed the questionnaire. The majority of attendees came because they were interested in science (68.0%), to learn something new (46.4%) and to enjoy themselves (44.8%). Our qualitative results confirm the quantitative findings. In addition, speakers viewed that they benefited by improving their communication skills and having the opportunity to network with scientists and non-scientists. Speakers also mentioned that such events were a good means to engage with the public, can improve the visibility of their work and potentially attract more funding. To improve the Pint of Science activities, audience members suggested to include a more diverse range of topics, more collaborations with other local research institutions and to hold the event at larger venues. CONCLUSIONS: We conclude that Pint of Science was well received in Bangkok with recommendations to improve minor issues related to practicalities and logistics.


Subject(s)
Holidays , Science , Social Behavior , Adult , Female , Humans , Male , Middle Aged , Qualitative Research , Science/education , Science/methods , Surveys and Questionnaires , Thailand , Young Adult
6.
ChemMedChem ; 14(14): 1329-1335, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31188540

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.


Subject(s)
Acetamides/pharmacology , Antimalarials/pharmacology , 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
7.
Nat Commun ; 9(1): 3805, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228275

ABSTRACT

Spread of parasite resistance to artemisinin threatens current frontline antimalarial therapies, highlighting the need for new drugs with alternative modes of action. Since only 0.2-1% of asexual parasites differentiate into sexual, transmission-competent forms, targeting this natural bottleneck provides a tangible route to interrupt disease transmission and mitigate resistance selection. Here we present a high-throughput screen of gametogenesis against a ~70,000 compound diversity library, identifying seventeen drug-like molecules that target transmission. Hit molecules possess varied activity profiles including male-specific, dual acting male-female and dual-asexual-sexual, with one promising N-((4-hydroxychroman-4-yl)methyl)-sulphonamide scaffold found to have sub-micromolar activity in vitro and in vivo efficacy. Development of leads with modes of action focussed on the sexual stages of malaria parasite development provide a previously unexplored base from which future therapeutics can be developed, capable of preventing parasite transmission through the population.


Subject(s)
Antimalarials/analysis , Drug Evaluation, Preclinical , High-Throughput Screening Assays/methods , Malaria/parasitology , Malaria/transmission , Parasites/physiology , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Feeding Behavior , Female , Gametogenesis/drug effects , Hep G2 Cells , Humans , Male , Mice , Parasites/drug effects , Phenotype , Reproducibility of Results , Structure-Activity Relationship
8.
Nat Microbiol ; 2(10): 1403-1414, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28808258

ABSTRACT

Antimalarial compounds with dual therapeutic and transmission-blocking activity are desired as high-value partners for combination therapies. Here, we report the identification and characterization of hexahydroquinolines (HHQs) that show low nanomolar potency against both pathogenic and transmissible intra-erythrocytic forms of the malaria parasite Plasmodium falciparum. This activity translates into potent transmission-blocking potential, as shown by in vitro male gamete formation assays and reduced oocyst infection and prevalence in Anopheles mosquitoes. In vivo studies illustrated the ability of lead HHQs to suppress Plasmodium berghei blood-stage parasite proliferation. Resistance selection studies, confirmed by CRISPR-Cas9-based gene editing, identified the digestive vacuole membrane-spanning transporter PfMDR1 (P. falciparum multidrug resistance gene-1) as a determinant of parasite resistance to HHQs. Haemoglobin and haem fractionation assays suggest a mode of action that results in reduced haemozoin levels and might involve inhibition of host haemoglobin uptake into intra-erythrocytic parasites. Furthermore, parasites resistant to HHQs displayed increased susceptibility to several first-line antimalarial drugs, including lumefantrine, confirming that HHQs have a different mode of action to other antimalarials drugs for which PfMDR1 is known to confer resistance. This work evokes therapeutic strategies that combine opposing selective pressures on this parasite transporter as an approach to countering the emergence and transmission of multidrug-resistant P. falciparum malaria.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Malaria/drug therapy , Plasmodium berghei/drug effects , Quinolines/pharmacology , Amino Acid Sequence , Animals , Anopheles , CRISPR-Cas Systems/genetics , DNA, Protozoan/genetics , DNA, Protozoan/metabolism , Drug Combinations , Drug Resistance , Endocytosis/drug effects , Ethanolamines/pharmacology , Fluorenes/pharmacology , Gene Editing , HEK293 Cells , Heme , Hemoglobins/drug effects , High-Throughput Screening Assays , Humans , Lumefantrine , Malaria/transmission , Malaria, Falciparum/blood , Malaria, Falciparum/transmission , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/drug effects , Multidrug Resistance-Associated Proteins/genetics , Mutation , Oocysts/drug effects , Plasmodium berghei/pathogenicity , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Quinolines/chemistry
9.
Nat Commun ; 8: 15159, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28537265

ABSTRACT

K13 gene mutations are a primary marker of artemisinin resistance in Plasmodium falciparum malaria that threatens the long-term clinical utility of artemisinin-based combination therapies, the cornerstone of modern day malaria treatment. Here we describe a multinational drug discovery programme that has delivered a synthetic tetraoxane-based molecule, E209, which meets key requirements of the Medicines for Malaria Venture drug candidate profiles. E209 has potent nanomolar inhibitory activity against multiple strains of P. falciparum and P. vivax in vitro, is efficacious against P. falciparum in in vivo rodent models, produces parasite reduction ratios equivalent to dihydroartemisinin and has pharmacokinetic and pharmacodynamic characteristics compatible with a single-dose cure. In vitro studies with transgenic parasites expressing variant forms of K13 show no cross-resistance with the C580Y mutation, the primary variant observed in Southeast Asia. E209 is a superior next generation endoperoxide with combined pharmacokinetic and pharmacodynamic features that overcome the liabilities of artemisinin derivatives.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Drug Resistance/drug effects , Plasmodium falciparum/drug effects , Plasmodium vivax/drug effects , Protozoan Proteins/metabolism , Tetraoxanes/chemistry , Tetraoxanes/pharmacology , Animals , Antimalarials/chemistry , Dogs , Dose-Response Relationship, Drug , Drug Resistance/genetics , Erythrocytes/parasitology , Female , Half-Life , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Plasmodium falciparum/genetics , Plasmodium vivax/genetics , Rats , Rats, Sprague-Dawley , Tetraoxanes/pharmacokinetics , Transgenes
10.
Nat Commun ; 8: 15160, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28513586

ABSTRACT

Plasmodium falciparum stage V gametocytes are responsible for parasite transmission, and drugs targeting this stage are needed to support malaria elimination. We here screen the Tres Cantos Antimalarial Set (TCAMS) using the previously developed P. falciparum female gametocyte activation assay (Pf FGAA), which assesses stage V female gametocyte viability and functionality using Pfs25 expression. We identify over 400 compounds with activities <2 µM, chemically classified into 57 clusters and 33 singletons. Up to 68% of the hits are chemotypes described for the first time as late-stage gametocyte-targeting molecules. In addition, the biological profile of 90 compounds representing the chemical diversity is assessed. We confirm in vitro transmission-blocking activity of four of the six selected molecules belonging to three distinct scaffold clusters. Overall, this TCAMS gametocyte screen provides 276 promising antimalarial molecules with dual asexual/sexual activity, representing starting points for target identification and candidate selection.


Subject(s)
Antimalarials/pharmacology , Drug Evaluation, Preclinical , Germ Cells/metabolism , Adenosine Triphosphate/metabolism , Animals , Antimalarials/chemistry , Antimalarials/pharmacokinetics , Antimalarials/therapeutic use , Disease Models, Animal , Female , Flagella/metabolism , Hep G2 Cells , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Reproducibility of Results
11.
Sci Transl Med ; 9(387)2017 04 26.
Article in English | MEDLINE | ID: mdl-28446690

ABSTRACT

As part of the global effort toward malaria eradication, phenotypic whole-cell screening revealed the 2-aminopyridine class of small molecules as a good starting point to develop new antimalarial drugs. Stemming from this series, we found that the derivative, MMV390048, lacked cross-resistance with current drugs used to treat malaria. This compound was efficacious against all Plasmodium life cycle stages, apart from late hypnozoites in the liver. Efficacy was shown in the humanized Plasmodium falciparum mouse model, and modest reductions in mouse-to-mouse transmission were achieved in the Plasmodium berghei mouse model. Experiments in monkeys revealed the ability of MMV390048 to be used for full chemoprotection. Although MMV390048 was not able to eliminate liver hypnozoites, it delayed relapse in a Plasmodium cynomolgi monkey model. Both genomic and chemoproteomic studies identified a kinase of the Plasmodium parasite, phosphatidylinositol 4-kinase, as the molecular target of MMV390048. The ability of MMV390048 to block all life cycle stages of the malaria parasite suggests that this compound should be further developed and may contribute to malaria control and eradication as part of a single-dose combination treatment.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Aminopyridines/therapeutic use , Antimalarials/therapeutic use , Sulfones/therapeutic use , Aminopyridines/pharmacology , Animals , Antimalarials/pharmacology , Female , Malaria/drug therapy , Malaria/enzymology , Male , Mice , Mice, SCID , Parasitic Sensitivity Tests , Plasmodium/drug effects , Plasmodium/pathogenicity , Sulfones/pharmacology
12.
Sci Commun ; 39(6): 810-820, 2017 Dec.
Article in English | MEDLINE | ID: mdl-30369782

ABSTRACT

The Pint of Science Festival is the largest annual international science festival. So far the event has been held simultaneously in Europe, North America, South America, Africa, and Australia but not in Asia. Pint of Science Thailand was held for the first time this year, in Thailand's capital, Bangkok. This article briefly discusses some of the successes, challenges, and lessons learnt associated with running the first Pint of Science event in Asia, a culture very different to the Western Hemisphere cities that have currently hosted Pint of Science events.

13.
ACS Cent Sci ; 2(10): 687-701, 2016 Oct 26.
Article in English | MEDLINE | ID: mdl-27800551

ABSTRACT

The development of new antimalarial compounds remains a pivotal part of the strategy for malaria elimination. Recent large-scale phenotypic screens have provided a wealth of potential starting points for hit-to-lead campaigns. One such public set is explored, employing an open source research mechanism in which all data and ideas were shared in real time, anyone was able to participate, and patents were not sought. One chemical subseries was found to exhibit oral activity but contained a labile ester that could not be replaced without loss of activity, and the original hit exhibited remarkable sensitivity to minor structural change. A second subseries displayed high potency, including activity within gametocyte and liver stage assays, but at the cost of low solubility. As an open source research project, unexplored avenues are clearly identified and may be explored further by the community; new findings may be cumulatively added to the present work.

14.
PLoS Med ; 13(10): e1002138, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27701420

ABSTRACT

BACKGROUND: Artemisinin resistance observed in Southeast Asia threatens the continued use of artemisinin-based combination therapy in endemic countries. Additionally, the diversity of chemical mode of action in the global portfolio of marketed antimalarials is extremely limited. Addressing the urgent need for the development of new antimalarials, a chemical class of potent antimalarial compounds with a novel mode of action was recently identified. Herein, the preclinical characterization of one of these compounds, ACT-451840, conducted in partnership with academic and industrial groups is presented. METHOD AND FINDINGS: The properties of ACT-451840 are described, including its spectrum of activities against multiple life cycle stages of the human malaria parasite Plasmodium falciparum (asexual and sexual) and Plasmodium vivax (asexual) as well as oral in vivo efficacies in two murine malaria models that permit infection with the human and the rodent parasites P. falciparum and Plasmodium berghei, respectively. In vitro, ACT-451840 showed a 50% inhibition concentration of 0.4 nM (standard deviation [SD]: ± 0.0 nM) against the drug-sensitive P. falciparum NF54 strain. The 90% effective doses in the in vivo efficacy models were 3.7 mg/kg against P. falciparum (95% confidence interval: 3.3-4.9 mg/kg) and 13 mg/kg against P. berghei (95% confidence interval: 11-16 mg/kg). ACT-451840 potently prevented male gamete formation from the gametocyte stage with a 50% inhibition concentration of 5.89 nM (SD: ± 1.80 nM) and dose-dependently blocked oocyst development in the mosquito with a 50% inhibitory concentration of 30 nM (range: 23-39). The compound's preclinical safety profile is presented and is in line with the published results of the first-in-man study in healthy male participants, in whom ACT-451840 was well tolerated. Pharmacokinetic/pharmacodynamic (PK/PD) modeling was applied using efficacy in the murine models (defined either as antimalarial activity or as survival) in relation to area under the concentration versus time curve (AUC), maximum observed plasma concentration (Cmax), and time above a threshold concentration. The determination of the dose-efficacy relationship of ACT-451840 under curative conditions in rodent malaria models allowed prediction of the human efficacious exposure. CONCLUSION: The dual activity of ACT-451840 against asexual and sexual stages of P. falciparum and the activity on P. vivax have the potential to meet the specific profile of a target compound that could replace the fast-acting artemisinin component and harbor additional gametocytocidal activity and, thereby, transmission-blocking properties. The fast parasite reduction ratio (PRR) and gametocytocidal effect of ACT-451840 were recently also confirmed in a clinical proof-of-concept (POC) study.


Subject(s)
Acrylamides/pharmacology , Antimalarials/pharmacology , Piperazines/pharmacology , Plasmodium falciparum/drug effects , Plasmodium vivax/drug effects , Acrylamides/pharmacokinetics , Animals , Antimalarials/pharmacokinetics , Artemisinins/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred NOD , Microbial Sensitivity Tests , Multidrug Resistance-Associated Proteins/metabolism , Piperazines/pharmacokinetics , Plasmodium berghei/drug effects
15.
Nat Protoc ; 11(9): 1668-80, 2016 09.
Article in English | MEDLINE | ID: mdl-27560172

ABSTRACT

The prevention of parasite transmission from the human host to the mosquito has been recognized as a vital tool for malaria eradication campaigns. However, transmission-blocking antimalarial drug and/or vaccine discovery and development is currently hampered by the expense and difficulty of producing mature Plasmodium falciparum gametocytes in vitro-the parasite stage responsible for mosquito infection. Current protocols for P. falciparum gametocyte culture usually require complex parasite synchronization and addition of stimulating and/or inhibitory factors, and they may not have demonstrated the essential property of mosquito infectivity. This protocol details all the steps required for reliable P. falciparum gametocyte production and highlights common factors that influence culture success. The protocol can be completed in 15 d, and particular emphasis is placed upon operating a gametocyte culture facility on a continuous cycle. In addition, we show how functionally viable gametocytes can be used to evaluate transmission-blocking drugs both in a field setting and at high throughput (HTP) for drug discovery.


Subject(s)
Cell Culture Techniques/methods , Germ Cells/cytology , Plasmodium falciparum/cytology , Humans
17.
Sci Transl Med ; 7(296): 296ra111, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26180101

ABSTRACT

Malaria is one of the most significant causes of childhood mortality, but disease control efforts are threatened by resistance of the Plasmodium parasite to current therapies. Continued progress in combating malaria requires development of new, easy to administer drug combinations with broad-ranging activity against all manifestations of the disease. DSM265, a triazolopyrimidine-based inhibitor of the pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase (DHODH), is the first DHODH inhibitor to reach clinical development for treatment of malaria. We describe studies profiling the biological activity, pharmacological and pharmacokinetic properties, and safety of DSM265, which supported its advancement to human trials. DSM265 is highly selective toward DHODH of the malaria parasite Plasmodium, efficacious against both blood and liver stages of P. falciparum, and active against drug-resistant parasite isolates. Favorable pharmacokinetic properties of DSM265 are predicted to provide therapeutic concentrations for more than 8 days after a single oral dose in the range of 200 to 400 mg. DSM265 was well tolerated in repeat-dose and cardiovascular safety studies in mice and dogs, was not mutagenic, and was inactive against panels of human enzymes/receptors. The excellent safety profile, blood- and liver-stage activity, and predicted long half-life in humans position DSM265 as a new potential drug combination partner for either single-dose treatment or once-weekly chemoprevention. DSM265 has advantages over current treatment options that are dosed daily or are inactive against the parasite liver stage.


Subject(s)
Antimalarials/chemistry , Enzyme Inhibitors/chemistry , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Pyrimidines/chemistry , Triazoles/chemistry , Administration, Oral , Animals , Antimalarials/pharmacokinetics , Area Under Curve , Caco-2 Cells , Crystallography, X-Ray , Dihydroorotate Dehydrogenase , Dogs , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Haplorhini , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Plasmodium falciparum , Pyrimidines/pharmacokinetics , Rabbits , Substrate Specificity , Triazoles/pharmacokinetics
18.
Nature ; 522(7556): 315-20, 2015 06 18.
Article in English | MEDLINE | ID: mdl-26085270

ABSTRACT

There is an urgent need for new drugs to treat malaria, with broad therapeutic potential and novel modes of action, to widen the scope of treatment and to overcome emerging drug resistance. Here we describe the discovery of DDD107498, a compound with a potent and novel spectrum of antimalarial activity against multiple life-cycle stages of the Plasmodium parasite, with good pharmacokinetic properties and an acceptable safety profile. DDD107498 demonstrates potential to address a variety of clinical needs, including single-dose treatment, transmission blocking and chemoprotection. DDD107498 was developed from a screening programme against blood-stage malaria parasites; its molecular target has been identified as translation elongation factor 2 (eEF2), which is responsible for the GTP-dependent translocation of the ribosome along messenger RNA, and is essential for protein synthesis. This discovery of eEF2 as a viable antimalarial drug target opens up new possibilities for drug discovery.


Subject(s)
Antimalarials/pharmacology , Gene Expression Regulation/drug effects , Malaria/parasitology , Plasmodium/drug effects , Plasmodium/metabolism , Protein Biosynthesis/drug effects , Quinolines/pharmacology , Animals , Antimalarials/administration & dosage , Antimalarials/adverse effects , Antimalarials/pharmacokinetics , Drug Discovery , Female , Life Cycle Stages/drug effects , Liver/drug effects , Liver/parasitology , Malaria/drug therapy , Male , Models, Molecular , Peptide Elongation Factor 2/antagonists & inhibitors , Peptide Elongation Factor 2/metabolism , Plasmodium/genetics , Plasmodium/growth & development , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Plasmodium vivax/drug effects , Plasmodium vivax/metabolism , Quinolines/administration & dosage , Quinolines/chemistry , Quinolines/pharmacokinetics
19.
Antimicrob Agents Chemother ; 59(6): 3298-305, 2015.
Article in English | MEDLINE | ID: mdl-25801574

ABSTRACT

In response to a call for the global eradication of malaria, drug discovery has recently been extended to identify compounds that prevent the onward transmission of the parasite, which is mediated by Plasmodium falciparum stage V gametocytes. Lately, metabolic activity has been used in vitro as a surrogate for gametocyte viability; however, as gametocytes remain relatively quiescent at this stage, their ability to undergo onward development (gamete formation) may be a better measure of their functional viability. During gamete formation, female gametocytes undergo profound morphological changes and express translationally repressed mRNA. By assessing female gamete cell surface expression of one such repressed protein, Pfs25, as the readout for female gametocyte functional viability, we developed an imaging-based high-throughput screening (HTS) assay to identify transmission-blocking compounds. This assay, designated the P. falciparum female gametocyte activation assay (FGAA), was scaled up to a high-throughput format (Z' factor, 0.7 ± 0.1) and subsequently validated using a selection of 50 known antimalarials from diverse chemical families. Only a few of these agents showed submicromolar 50% inhibitory concentrations in the assay: thiostrepton, methylene blue, and some endoperoxides. To determine the best conditions for HTS, a robustness test was performed with a selection of the GlaxoSmithKline Tres Cantos Antimalarial Set (TCAMS) and the final screening conditions for this library were determined to be a 2 µM concentration and 48 h of incubation with gametocytes. The P. falciparum FGAA has been proven to be a robust HTS assay faithful to Plasmodium transmission-stage cell biology, and it is an innovative useful tool for antimalarial drug discovery which aims to identify new molecules with transmission-blocking potential.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Animals , Female , High-Throughput Screening Assays , Inhibitory Concentration 50 , Methylene Blue/pharmacology , Plasmodium falciparum/genetics , RNA, Messenger/genetics , Thiostrepton/pharmacology
20.
Antimicrob Agents Chemother ; 59(2): 950-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25421480

ABSTRACT

Current antimalarials are under continuous threat due to the relentless development of drug resistance by malaria parasites. We previously reported promising in vitro parasite-killing activity with the histone methyltransferase inhibitor BIX-01294 and its analogue TM2-115. Here, we further characterize these diaminoquinazolines for in vitro and in vivo efficacy and pharmacokinetic properties to prioritize and direct compound development. BIX-01294 and TM2-115 displayed potent in vitro activity, with 50% inhibitory concentrations (IC50s) of <50 nM against drug-sensitive laboratory strains and multidrug-resistant field isolates, including artemisinin-refractory Plasmodium falciparum isolates. Activities against ex vivo clinical isolates of both P. falciparum and Plasmodium vivax were similar, with potencies of 300 to 400 nM. Sexual-stage gametocyte inhibition occurs at micromolar levels; however, mature gametocyte progression to gamete formation is inhibited at submicromolar concentrations. Parasite reduction ratio analysis confirms a high asexual-stage rate of killing. Both compounds examined displayed oral efficacy in in vivo mouse models of Plasmodium berghei and P. falciparum infection. The discovery of a rapid and broadly acting antimalarial compound class targeting blood stage infection, including transmission stage parasites, and effective against multiple malaria-causing species reveals the diaminoquinazoline scaffold to be a very promising lead for development into greatly needed novel therapies to control malaria.


Subject(s)
Antimalarials/therapeutic use , Azepines/therapeutic use , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Malaria/drug therapy , Quinazolines/therapeutic use , Animals , Antimalarials/chemistry , Azepines/chemistry , Female , Hep G2 Cells , Histone Methyltransferases , Humans , Malaria, Falciparum/drug therapy , Mice , Mice, SCID , Plasmodium berghei/drug effects , Plasmodium berghei/pathogenicity , Plasmodium falciparum/drug effects , Plasmodium falciparum/pathogenicity , Quinazolines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL