Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Publication year range
1.
J Infect Dis ; 225(6): 1062-1069, 2022 03 15.
Article in English | MEDLINE | ID: mdl-32479608

ABSTRACT

BACKGROUND: Interventions that effectively target Plasmodium vivax are critical for the future control and elimination of malaria. We conducted a P. vivax volunteer infection study to characterize the antimalarial activity of artefenomel, a new drug candidate. METHODS: Eight healthy, malaria-naive participants were intravenously inoculated with blood-stage P. vivax and subsequently received a single oral 200-mg dose of artefenomel. Blood samples were collected to monitor the development and clearance of parasitemia, and plasma artefenomel concentration. Mosquito feeding assays were conducted before artefenomel dosing to investigate parasite transmissibility. RESULTS: Initial parasite clearance occurred in all participants after artefenomel administration (log10 parasite reduction ratio over 48 hours, 1.67; parasite clearance half-life, 8.67 hours). Recrudescence occurred in 7 participants 11-14 days after dosing. A minimum inhibitory concentration of 0.62 ng/mL and minimum parasiticidal concentration that achieves 90% of maximum effect of 0.83 ng/mL were estimated, and a single 300-mg dose was predicted to clear 109 parasites per milliliter with 95% certainty. Gametocytemia developed in all participants and was cleared 4-8 days after dosing. At peak gametocytemia, 75% of participants were infectious to mosquitoes. CONCLUSIONS: The in vivo antimalarial activity of artefenomel supports its further clinical development as a treatment for P. vivax malaria. CLINICAL TRIALS REGISTRATION: NCT02573857.


Subject(s)
Antimalarials , Culicidae , Folic Acid Antagonists , Malaria, Falciparum , Malaria, Vivax , Parasites , Adamantane/analogs & derivatives , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Folic Acid Antagonists/pharmacology , Humans , Malaria, Falciparum/parasitology , Malaria, Vivax/drug therapy , Peroxides , Plasmodium falciparum , Plasmodium vivax
2.
Antimicrob Agents Chemother ; 66(7): e0011422, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35727057

ABSTRACT

The rate at which parasitemia declines in a host after treatment with an antimalarial drug is a major metric for assessment of antimalarial drug activity in preclinical models and in early clinical trials. However, this metric does not distinguish between viable and nonviable parasites. Thus, enumeration of parasites may result in underestimation of drug activity for some compounds, potentially confounding its use as a metric for assessing antimalarial activity in vivo. Here, we report a study of the effect of artesunate on Plasmodium falciparum viability in humans and in mice. We first measured the drug effect in mice by estimating the decrease in parasite viability after treatment using two independent approaches to estimate viability. We demonstrate that, as previously reported in humans, parasite viability declines much faster after artesunate treatment than does the decline in parasitemia (termed parasite clearance). We also observed that artesunate kills parasites faster at higher concentrations, which is not discernible from the traditional parasite clearance curve and that each subsequent dose of artesunate maintains its killing effect. Furthermore, based on measures of parasite viability, we could accurately predict the in vivo recrudescence of infection. Finally, using pharmacometrics modeling, we show that the apparent differences in the antimalarial activity of artesunate in mice and humans are partly explained by differences in host removal of dead parasites in the two hosts. However, these differences, along with different pharmacokinetic profiles, do not fully account for the differences in activity. (This study has been registered with the Australian New Zealand Clinical Trials Registry under identifier ACTRN12617001394336.).


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Parasites , Animals , Antimalarials/pharmacokinetics , Antimalarials/therapeutic use , Artemisinins/pharmacokinetics , Artemisinins/therapeutic use , Artesunate/pharmacology , Artesunate/therapeutic use , Australia , Humans , Malaria, Falciparum/drug therapy , Mice , Parasitemia/drug therapy , Parasitemia/parasitology , Plasmodium falciparum
3.
Antimicrob Agents Chemother ; 66(1): e0158421, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34694880

ABSTRACT

Despite repeated malaria infection, individuals living in areas where malaria is endemic remain vulnerable to reinfection. The Janus kinase (JAK1/2) inhibitor ruxolitinib could potentially disrupt the parasite-induced dysfunctional immune response when administered with antimalarial therapy. This randomized, single-blind, placebo-controlled, single-center phase 1 trial investigated the safety, tolerability, and pharmacokinetic and pharmacodynamic profile of ruxolitinib and the approved antimalarial artemether-lumefantrine in combination. Ruxolitinib pharmacodynamics were assessed by inhibition of phosphorylation of signal transducer and activator of transcription 3 (pSTAT3). Eight healthy male and female participants ages 18 to 55 years were randomized to either ruxolitinib (20 mg) (n = 6) or placebo (n = 2) administered 2 h after artemether-lumefantrine (80/480 mg) twice daily for 3 days. Mild adverse events occurred in six participants (four ruxolitinib; two placebo). The combination of artemether-lumefantrine and ruxolitinib was well tolerated, with adverse events and pharmacokinetics consistent with the known profiles of both drugs. The incidence of adverse events and artemether, dihydroartemisinin (the major active metabolite of artemether), and lumefantrine exposure were not affected by ruxolitinib coadministration. Ruxolitinib coadministration resulted in a 3-fold-greater pSTAT3 inhibition compared to placebo (geometric mean ratio = 3.01 [90% confidence interval = 2.14 to 4.24]), with a direct and predictable relationship between ruxolitinib plasma concentrations and %pSTAT3 inhibition. This study supports the investigation of the combination of artemether-lumefantrine and ruxolitinib in healthy volunteers infected with Plasmodium falciparum malaria. (This study has been registered at ClinicalTrials.gov under registration no. NCT04456634.).


Subject(s)
Antimalarials , Malaria, Falciparum , Adolescent , Adult , Antimalarials/adverse effects , Artemether/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Drug Combinations , Ethanolamines/therapeutic use , Female , Fluorenes/therapeutic use , Humans , Lumefantrine/therapeutic use , Malaria, Falciparum/drug therapy , Male , Middle Aged , Nitriles , Pyrazoles , Pyrimidines , Single-Blind Method , Young Adult
4.
Annu Rev Pharmacol Toxicol ; 58: 567-582, 2018 01 06.
Article in English | MEDLINE | ID: mdl-28992431

ABSTRACT

Malaria is a critical public health problem resulting in substantial morbidity and mortality, particularly in developing countries. Owing to the development of resistance toward current therapies, novel approaches to accelerate the development efforts of new malaria therapeutics are urgently needed. There have been significant advancements in the development of in vitro and in vivo experiments that generate data used to inform decisions about the potential merit of new compounds. A comprehensive disease-drug model capable of integrating discrete data from different preclinical and clinical components would be a valuable tool across all stages of drug development. This could have an enormous impact on the otherwise slow and resource-intensive process of traditional clinical drug development.


Subject(s)
Antimalarials/pharmacology , Antimalarials/therapeutic use , Malaria/drug therapy , Animals , Drug Development/methods , Humans
5.
Article in English | MEDLINE | ID: mdl-33526486

ABSTRACT

Ensuring continued success against malaria depends on a pipeline of new antimalarials. Antimalarial drug development utilizes preclinical murine and experimental human malaria infection studies to evaluate drug efficacy. A sequential approach is typically adapted, with results from each stage informing the design of the next stage of development. The validity of this approach depends on confidence that results from murine malarial studies predict the outcome of clinical trials in humans. Parasite clearance rates following treatment are key parameters of drug efficacy. To investigate the validity of forward predictions, we developed a suite of mathematical models to capture parasite growth and drug clearance along the drug development pathway and estimated parasite clearance rates. When comparing the three infection experiments, we identified different relationships of parasite clearance with dose and different maximum parasite clearance rates. In Plasmodium berghei-NMRI mouse infections, we estimated a maximum parasite clearance rate of 0.2 (1/h); in Plasmodium falciparum-SCID mouse infections, 0.05 (1/h); and in human volunteer infection studies with P. falciparum, we found a maximum parasite clearance rate of 0.12 (1/h) and 0.18 (1/h) after treatment with OZ439 and MMV048, respectively. Sensitivity analysis revealed that host-parasite driven processes account for up to 25% of variance in parasite clearance for medium-high doses of antimalarials. Although there are limitations in translating parasite clearance rates across these experiments, they provide insight into characterizing key parameters of drug action and dose response and assist in decision-making regarding dosage for further drug development.


Subject(s)
Antimalarials , Malaria, Falciparum , Parasites , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Malaria, Falciparum/drug therapy , Mice , Mice, SCID , Plasmodium berghei , Plasmodium falciparum
6.
Clin Infect Dis ; 71(10): e657-e664, 2020 12 17.
Article in English | MEDLINE | ID: mdl-32239164

ABSTRACT

BACKGROUND: MMV390048 is the first Plasmodium phosphatidylinositol 4-kinase inhibitor to reach clinical development as a new antimalarial. We aimed to characterize the safety, pharmacokinetics, and antimalarial activity of a tablet formulation of MMV390048. METHODS: A 2-part, phase 1 trial was conducted in healthy adults. Part 1 was a double-blind, randomized, placebo-controlled, single ascending dose study consisting of 3 cohorts (40, 80, 120 mg MMV390048). Part 2 was an open-label volunteer infection study using the Plasmodium falciparum induced blood-stage malaria model consisting of 2 cohorts (40 mg and 80 mg MMV390048). RESULTS: Twenty four subjects were enrolled in part 1 (n = 8 per cohort, randomized 3:1 MMV390048:placebo) and 15 subjects were enrolled in part 2 (40 mg [n = 7] and 80 mg [n = 8] cohorts). One subject was withdrawn from part 2 (80 mg cohort) before dosing and was not included in analyses. No serious or severe adverse events were attributed to MMV390048. The rate of parasite clearance was greater in subjects administered 80 mg compared to those administered 40 mg (clearance half-life 5.5 hours [95% confidence interval {CI}, 5.2-6.0 hours] vs 6.4 hours [95% CI, 6.0-6.9 hours]; P = .005). Pharmacokinetic/pharmacodynamic modeling estimated a minimum inhibitory concentration of 83 ng/mL and a minimal parasiticidal concentration that would achieve 90% of the maximum effect of 238 ng/mL, and predicted that a single 120-mg dose would achieve an adequate clinical and parasitological response with 92% certainty. CONCLUSIONS: The safety, pharmacokinetics, and pharmacodynamics of MMV390048 support its further development as a partner drug of a single-dose combination therapy for malaria. CLINICAL TRIALS REGISTRATION: NCT02783820 (part 1); NCT02783833 (part 2).


Subject(s)
Antimalarials/therapeutic use , Malaria, Falciparum , 1-Phosphatidylinositol 4-Kinase , Adult , Aminopyridines , Antimalarials/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Malaria, Falciparum/drug therapy , Plasmodium , Sulfones , Volunteers
7.
PLoS Med ; 17(8): e1003203, 2020 08.
Article in English | MEDLINE | ID: mdl-32822347

ABSTRACT

BACKGROUND: Artemisinin resistance is threatening malaria control. We aimed to develop and test a human model of artemisinin-resistant (ART-R) Plasmodium falciparum to evaluate the efficacy of drugs against ART-R malaria. METHODS AND FINDINGS: We conducted 2 sequential phase 1, single-centre, open-label clinical trials at Q-Pharm, Brisbane, Australia, using the induced blood-stage malaria (IBSM) model, whereby healthy participants are intravenously inoculated with blood-stage parasites. In a pilot study, participants were inoculated (Day 0) with approximately 2,800 viable P. falciparum ART-R parasites. In a comparative study, participants were randomised to receive approximately 2,800 viable P. falciparum ART-R (Day 0) or artemisinin-sensitive (ART-S) parasites (Day 1). In both studies, participants were administered a single approximately 2 mg/kg oral dose of artesunate (AS; Day 9). Primary outcomes were safety, ART-R parasite infectivity, and parasite clearance. In the pilot study, 2 participants were enrolled between April 27, 2017, and September 12, 2017, and included in final analyses (males n = 2 [100%], mean age = 26 years [range, 23-28 years]). In the comparative study, 25 participants were enrolled between October 26, 2017, and October 18, 2018, of whom 22 were inoculated and included in final analyses (ART-R infected participants: males n = 7 [53.8%], median age = 22 years [range, 18-40 years]; ART-S infected participants: males n = 5 [55.6%], median age = 28 years [range, 22-35 years]). In both studies, all participants inoculated with ART-R parasites became parasitaemic. A total of 36 adverse events were reported in the pilot study and 277 in the comparative study. Common adverse events in both studies included headache, pyrexia, myalgia, nausea, and chills; none were serious. Seven participants experienced transient severe falls in white cell counts and/or elevations in liver transaminase levels which were considered related to malaria. Additionally, 2 participants developed ventricular extrasystoles that were attributed to unmasking of a predisposition to benign fever-induced tachyarrhythmia. In the comparative study, parasite clearance half-life after AS was significantly longer for ART-R infected participants (n = 13, 6.5 hours; 95% confidence interval [CI] 6.3-6.7 hours) compared with ART-S infected participants (n = 9, 3.2 hours; 95% CI 3.0-3.3 hours; p < 0.001). The main limitation of this study was that the ART-R and ART-S parasite strains did not share the same genetic background. CONCLUSIONS: We developed the first (to our knowledge) human model of ART-R malaria. The delayed clearance profile of ART-R parasites after AS aligns with field study observations. Although based on a relatively small sample size, results indicate that this model can be safely used to assess new drugs against ART-R P. falciparum. TRIAL REGISTRATION: The studies were registered with the Australian New Zealand Clinical Trials Registry: ACTRN12617000244303 (https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=372357) and ACTRN12617001394336 (https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=373637).


Subject(s)
Anti-Infective Agents/therapeutic use , Antimalarials/therapeutic use , Artemisinins/therapeutic use , Malaria, Falciparum/blood , Malaria, Falciparum/drug therapy , Plasmodium falciparum/metabolism , Adolescent , Adult , Animals , Anti-Infective Agents/adverse effects , Anti-Infective Agents/pharmacology , Antimalarials/adverse effects , Antimalarials/pharmacology , Artemisinins/adverse effects , Artemisinins/pharmacology , Artesunate/adverse effects , Artesunate/pharmacology , Artesunate/therapeutic use , Australia/epidemiology , Female , Headache/chemically induced , Healthy Volunteers , Humans , Malaria, Falciparum/epidemiology , Male , Nausea/chemically induced , Parasites/metabolism , Pilot Projects , Young Adult
8.
Article in English | MEDLINE | ID: mdl-30858218

ABSTRACT

DSM265 is a novel antimalarial drug in clinical development that acts as a selective inhibitor of Plasmodium dihydroorotate dehydrogenase. In a previous phase 1b study, a single 150-mg dose of DSM265 showed partial efficacy against experimentally induced blood-stage Plasmodium falciparum malaria (IBSM). Pharmacokinetic/pharmacodynamic modeling predicted a human efficacious dose of 340 mg. The primary objectives of the current study were to determine the safety and efficacy of a single oral 400-mg dose of DSM265 against P. falciparum in the IBSM model. Eight healthy participants were inoculated intravenously with 2,800 parasites and treated with DSM265 7 days later. Unexpectedly, one participant did not develop parasitemia during the study. All other participants developed parasitemia, with the complete clearance of asexual parasites occurring following DSM265 treatment. All seven subjects also became gametocytemic. The secondary objectives were to investigate the gametocytocidal and transmission-blocking activity of a second 400-mg dose of DSM265, which was administered 23 days after inoculation. Gametocytes were not cleared by the second dose of DSM265, and transmission-blocking activity could not be determined due to low gametocyte densities. Three DSM265-related adverse events occurred, including a cutaneous rash in one subject on the day of the second DSM265 dose. The results obtained in this study support the prediction of the efficacious dose of DSM265 and provide further evidence that DSM265 is generally safe and well tolerated. In addition, this study confirms preclinical data indicating that DSM265 permits the development and maturation of gametocytes and does not clear mature circulating gametocytes. (This study has been registered at ClinicalTrials.gov under identifier NCT02573857.).


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/physiology , Pyrimidines/pharmacology , Triazoles/pharmacology , Animals , Anopheles , Antimalarials/administration & dosage , Antimalarials/adverse effects , Antimalarials/pharmacokinetics , Female , Half-Life , Healthy Volunteers , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Male , Mosquito Vectors , Parasitemia/drug therapy , Parasitemia/parasitology , Plasmodium falciparum/drug effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Triazoles/administration & dosage , Triazoles/adverse effects , Triazoles/pharmacokinetics , Young Adult
9.
Article in English | MEDLINE | ID: mdl-31685476

ABSTRACT

Artefenomel and DSM265 are two new compounds that have been shown to be well tolerated and effective when administered as monotherapy malaria treatment. This study aimed to determine the safety, pharmacokinetics, and pharmacodynamics of artefenomel and DSM265 administered in combination to healthy subjects in a volunteer infection study using the Plasmodium falciparum-induced blood-stage malaria model. Thirteen subjects were inoculated with parasite-infected erythrocytes on day 0 and received a single oral dose of artefenomel and DSM265 on day 7. Cohort 1 (n = 8) received 200 mg artefenomel plus 100 mg DSM265, and cohort 2 (n = 5) received 200 mg artefenomel plus 50 mg DSM265. Blood samples were collected to measure parasitemia, gametocytemia, and artefenomel-DSM265 plasma concentrations. There were no treatment-related adverse events. The pharmacokinetic profiles of artefenomel and DSM265 were similar to those of the compounds when administered as monotherapy, suggesting no pharmacokinetic interactions. A reduction in parasitemia occurred in all subjects following treatment (log10 parasite reduction ratios over 48 h [PRR48] of 2.80 for cohort 1 and 2.71 for cohort 2; parasite clearance half-lives of 5.17 h for cohort 1 and 5.33 h for cohort 2). Recrudescence occurred in 5/8 subjects in cohort 1 between days 19 and 28 and in 5/5 subjects in cohort 2 between days 15 and 22. Low-level gametocytemia (1 to 330 female gametocytes/ml) was detected in all subjects from day 14. The results of this single-dosing combination study support the further clinical development of the use of artefenomel and DSM265 in combination as a treatment for falciparum malaria. (This study has been registered at ClinicalTrials.gov under identifier NCT02389348.).


Subject(s)
Adamantane/analogs & derivatives , Antimalarials/administration & dosage , Malaria, Falciparum/drug therapy , Parasitemia/drug therapy , Peroxides/administration & dosage , Pyrimidines/administration & dosage , Triazoles/administration & dosage , Adamantane/administration & dosage , Adamantane/pharmacokinetics , Administration, Oral , Adult , Antimalarials/pharmacokinetics , Drug Combinations , Female , Healthy Volunteers , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Male , Middle Aged , Parasitemia/metabolism , Parasitemia/parasitology , Peroxides/pharmacokinetics , Plasmodium falciparum/drug effects , Pyrimidines/pharmacokinetics , Triazoles/pharmacokinetics , Young Adult
10.
J Infect Dis ; 217(5): 693-702, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29216395

ABSTRACT

Background: DSM265 is a selective inhibitor of Plasmodium dihydroorotate dehydrogenase that fully protected against controlled human malarial infection (CHMI) by direct venous inoculation of Plasmodium falciparum sporozoites when administered 1 day before challenge and provided partial protection when administered 7 days before challenge. Methods: A double-blinded, randomized, placebo-controlled trial was performed to assess safety, tolerability, pharmacokinetics, and efficacy of 1 oral dose of 400 mg of DSM265 before CHMI. Three cohorts were studied, with DSM265 administered 3 or 7 days before direct venous inoculation of sporozoites or 7 days before 5 bites from infected mosquitoes. Results: DSM265-related adverse events consisted of mild-to-moderate headache and gastrointestinal symptoms. DSM265 concentrations were consistent with pharmacokinetic models (mean area under the curve extrapolated to infinity, 1707 µg*h/mL). Placebo-treated participants became positive by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and were treated 7-10 days after CHMI. Among DSM265-treated subjects, 2 of 6 in each cohort were sterilely protected. DSM265-treated recipients had longer times to development of parasitemia than placebo-treated participants (P < .004). Conclusions: This was the first CHMI study of a novel antimalarial compound to compare direct venous inoculation of sporozoites and mosquito bites. Times to qRT-PCR positivity and treatment were comparable for both routes. DSM265 given 3 or 7 days before CHMI was safe and well tolerated but sterilely protected only one third of participants.


Subject(s)
Antimalarials/administration & dosage , Chemoprevention/methods , Malaria, Falciparum/prevention & control , Pyrimidines/administration & dosage , Triazoles/administration & dosage , Adolescent , Adult , Animals , Antimalarials/adverse effects , Antimalarials/pharmacokinetics , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Male , Middle Aged , Parasitemia/prevention & control , Placebos/administration & dosage , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Real-Time Polymerase Chain Reaction , Treatment Outcome , Triazoles/adverse effects , Triazoles/pharmacokinetics , Young Adult
11.
Malar J ; 17(1): 402, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30384848

ABSTRACT

Over the last 15 years, the majority of malaria drug discovery and development efforts have focused on new molecules and regimens to treat patients with uncomplicated or severe disease. In addition, a number of new molecular scaffolds have been discovered which block the replication of the parasite in the liver, offering the possibility of new tools for oral prophylaxis or chemoprotection, potentially with once-weekly dosing. However, an intervention which requires less frequent administration than this would be a key tool for the control and elimination of malaria. Recent progress in HIV drug discovery has shown that small molecules can be formulated for injections as native molecules or pro-drugs which provide protection for at least 2 months. Advances in antibody engineering offer an alternative approach whereby a single injection could potentially provide protection for several months. Building on earlier profiles for uncomplicated and severe malaria, a target product profile is proposed here for an injectable medicine providing long-term protection from this disease. As with all of such profiles, factors such as efficacy, cost, safety and tolerability are key, but with the changing disease landscape in Africa, new clinical and regulatory approaches are required to develop prophylactic/chemoprotective medicines. An overall framework for these approaches is suggested here.


Subject(s)
Antimalarials , Drug Development , Drug Discovery , Injections, Intravenous , Malaria/prevention & control , Humans
12.
Malar J ; 16(1): 26, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28086874

ABSTRACT

A decade of discovery and development of new anti-malarial medicines has led to a renewed focus on malaria elimination and eradication. Changes in the way new anti-malarial drugs are discovered and developed have led to a dramatic increase in the number and diversity of new molecules presently in pre-clinical and early clinical development. The twin challenges faced can be summarized by multi-drug resistant malaria from the Greater Mekong Sub-region, and the need to provide simplified medicines. This review lists changes in anti-malarial target candidate and target product profiles over the last 4 years. As well as new medicines to treat disease and prevent transmission, there has been increased focus on the longer term goal of finding new medicines for chemoprotection, potentially with long-acting molecules, or parenteral formulations. Other gaps in the malaria armamentarium, such as drugs to treat severe malaria and endectocides (that kill mosquitoes which feed on people who have taken the drug), are defined here. Ultimately the elimination of malaria requires medicines that are safe and well-tolerated to be used in vulnerable populations: in pregnancy, especially the first trimester, and in those suffering from malnutrition or co-infection with other pathogens. These updates reflect the maturing of an understanding of the key challenges in producing the next generation of medicines to control, eliminate and ultimately eradicate malaria.


Subject(s)
Antimalarials/isolation & purification , Antimalarials/pharmacology , Drug Discovery/trends , Malaria/drug therapy , Malaria/prevention & control , Clinical Trials as Topic , Drug Evaluation, Preclinical/trends , Humans
13.
J Infect Dis ; 214(5): 753-61, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27252475

ABSTRACT

BACKGROUND: The emergence of drug-resistant malaria highlights the need for new agents. A desired characteristic of candidate antimalarials is rapid killing of parasites. This is typically measured by the rate of exponential clearance of parasitemia following treatment. However, this clearance rate excludes the highly variable lag phase, when the parasitemia level may increase, remain constant, or decrease. Understanding factors determining this lag phase is important for drug development. METHODS: We assessed the kinetics of parasitemia in 112 volunteers infected with blood-stage Plasmodium falciparum and treated with 8 different antimalarials. The parasitemia level was measured by quantitative polymerase chain reaction. We analyzed the relationship between the timing of treatment in the parasite growth cycle, and whether the parasitemia level rose or fell in the first 12 or 24 hours after treatment. RESULTS: The timing of treatment in the parasite life cycle predicted whether subjects experienced rises or falls in parasitemia level after treatment. Antimalarials were unable to prevent rises in the parasitemia level in the first 12 hours. However, in the first 24 hours after treatment, fast-acting but not slow-acting drugs reduced the parasitemia level independent of when treatment was administered. CONCLUSIONS: The highly variable lag phase depends on the speed of action of an antimalarial and when in the periodic growth cycle it is administered.


Subject(s)
Antimalarials/administration & dosage , Malaria, Falciparum/drug therapy , Parasitemia/drug therapy , Plasmodium falciparum/isolation & purification , Animals , Cohort Studies , Humans , Malaria, Falciparum/parasitology , Parasitemia/parasitology , Plasmodium falciparum/drug effects , Real-Time Polymerase Chain Reaction , Time Factors , Treatment Outcome , Volunteers
14.
J Infect Dis ; 214(1): 105-13, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27056954

ABSTRACT

BACKGROUND: Piperaquine, coformulated with dihydroartemisinin, is a component of a widely used artemisinin combination therapy. There is a paucity of data on its antimalarial activity as a single agent. Such data, if available, would inform selection of new coformulations. METHODS: We undertook a study in healthy subjects, using the induced blood stage malaria (IBSM) model to test the antimalarial activity of single doses of piperaquine (960, 640, and 480 mg) in 3 cohorts. In a pilot study in the third cohort, gametocyte clearance following administration of 15 mg, or 45 mg or no primaquine was investigated. RESULTS: Parasite clearance over the 48-hour period after piperaquine administration was more rapid in the 960 mg cohort, compared with the 640 mg cohort (parasite reduction ratio, 2951 [95% confidence interval {CI}, 1520-5728] vs 586 [95% CI, 351-978]; P < .001). All 24 subjects developed gametocytemia as determined by pfs25 transcripts. Clearance of pfs25 was significantly faster in those receiving primaquine than in those not receiving primaquine (P < .001). CONCLUSIONS: Piperaquine possesses rapid parasite-clearing activity, but monotherapy is followed by the appearance of gametocytemia, which could facilitate the spread of malaria. This new information should be taken into account when developing future antimalarial coformulations. CLINICAL TRIALS REGISTRATION: ACTRN12613000565741.


Subject(s)
Antimalarials/adverse effects , Antimalarials/therapeutic use , Gametogenesis/drug effects , Malaria, Falciparum/drug therapy , Parasitemia/drug therapy , Plasmodium falciparum/isolation & purification , Quinolines/therapeutic use , Adolescent , Adult , Australia , Cohort Studies , Female , Humans , Male , Middle Aged , Pilot Projects , Plasmodium falciparum/drug effects , Young Adult
15.
Antimicrob Agents Chemother ; 60(6): 3669-75, 2016 06.
Article in English | MEDLINE | ID: mdl-27044554

ABSTRACT

Effective progression of candidate antimalarials is dependent on optimal dosing in clinical studies, which is determined by a sound understanding of pharmacokinetics and pharmacodynamics (PK/PD). Recently, two important translational models for antimalarials have been developed: the NOD/SCID/IL2Rγ(-/-) (NSG) model, whereby mice are engrafted with noninfected and Plasmodium falciparum-infected human erythrocytes, and the induced blood-stage malaria (IBSM) model in human volunteers. The antimalarial mefloquine was used to directly measure the PK/PD in both models, which were compared to previously published trial data for malaria patients. The clinical part was a single-center, controlled study using a blood-stage Plasmodium falciparum challenge inoculum in volunteers to characterize the effectiveness of mefloquine against early malaria. The study was conducted in three cohorts (n = 8 each) using different doses of mefloquine. The characteristic delay in onset of action of about 24 h was seen in both NSG and IBSM systems. In vivo 50% inhibitory concentrations (IC50s) were estimated at 2.0 µg/ml and 1.8 µg/ml in the NSG and IBSM models, respectively, aligning with 1.8 µg/ml reported previously for patients. In the IBSM model, the parasite reduction ratios were 157 and 195 for the 10- and 15-mg/kg doses, within the range of previously reported clinical data for patients but significantly lower than observed in the mouse model. Linking mouse and human challenge models to clinical trial data can accelerate the accrual of critical data on antimalarial drug activity. Such data can guide large clinical trials required for development of urgently needed novel antimalarial combinations. (This trial was registered at the Australian New Zealand Clinical Trials Registry [http://anzctr.org.au] under registration number ACTRN12612000323820.).


Subject(s)
Antimalarials/pharmacokinetics , Malaria, Falciparum/drug therapy , Mefloquine/pharmacokinetics , Plasmodium falciparum/drug effects , Adult , Animals , Antimalarials/blood , Antimalarials/pharmacology , Cohort Studies , Disease Models, Animal , Drug Administration Schedule , Drug Dosage Calculations , Erythrocytes/drug effects , Erythrocytes/parasitology , Female , Healthy Volunteers , Humans , Inhibitory Concentration 50 , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Male , Mefloquine/blood , Mefloquine/pharmacology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Plasmodium falciparum/growth & development
16.
J Antimicrob Chemother ; 71(9): 2620-7, 2016 09.
Article in English | MEDLINE | ID: mdl-27272721

ABSTRACT

OBJECTIVES: OZ439, or artefenomel, is an investigational synthetic ozonide antimalarial with similar potency, but a significantly improved pharmacokinetic profile, compared with artemisinins. We wished to measure key pharmacokinetic and pharmacodynamic parameters and the pharmacokinetic/pharmacodynamic relationship of artefenomel in humans to guide the drug's further development as combination therapy in patients. PATIENTS AND METHODS: We tested artefenomel in the human induced blood-stage malaria (IBSM) model. Plasmodium infection was monitored by quantitative PCR (qPCR) and upon reaching 1000 parasites/mL single doses of 100, 200 and 500 mg of artefenomel were administered orally with evaluation of drug exposure and parasitaemia until rescue treatment after 16 days or earlier, if required. RESULTS: A single 100 mg dose had only a transient effect, while the 200 mg dose resulted in a significant reduction in parasitaemia before early recrudescence. At the highest (500 mg) dose, initial clearance of parasites below the limit of detection of qPCR was observed, with a 48 h parasite reduction ratio (PRR48) >10 000 and a parasite clearance half-life of 3.6 h (95% CI 3.4-3.8 h). However, at this dose, recrudescence was seen in four of eight subjects 6-10 days after treatment. Pharmacokinetic/pharmacodynamic modelling predicted an MIC of 4.1 ng/mL. CONCLUSIONS: These results confirm the antimalarial potential of artefenomel for use in a single-exposure combination therapy. The observations from this study support and will assist further clinical development of artefenomel.


Subject(s)
Adamantane/analogs & derivatives , Antimalarials/pharmacology , Antimalarials/pharmacokinetics , Malaria, Falciparum/drug therapy , Peroxides/pharmacology , Peroxides/pharmacokinetics , Plasmodium falciparum/drug effects , Adamantane/administration & dosage , Adamantane/pharmacokinetics , Adamantane/pharmacology , Administration, Oral , Adolescent , Adult , Antimalarials/administration & dosage , Cohort Studies , Female , Healthy Volunteers , Humans , Male , Microbial Sensitivity Tests , Parasite Load , Peroxides/administration & dosage , Real-Time Polymerase Chain Reaction , Treatment Outcome , Young Adult
17.
Malar J ; 15: 469, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27624471

ABSTRACT

BACKGROUND: Ferroquine (SSR97193) is a candidate anti-malarial currently undergoing clinical trials for malaria. To better understand its pharmacokinetic (PK) and pharmacodynamic (PD) parameters the compound was tested in the experimentally induced blood stage malaria infection model in volunteers. METHODS: Male and non-pregnant female aged 18-50 years were screened for this phase II, controlled, single-centre clinical trial. Subjects were inoculated with ~1800 viable Plasmodium falciparum 3D7A-infected human erythrocytes, and treated with a single-dose of 800 mg ferroquine. Blood samples were taken at defined time-points to measure PK and PD parameters. The blood concentration of ferroquine and its active metabolite, SSR97213, were measured on dry blood spot samples by ultra-performance liquid chromatography with tandem mass spectrometry (LC-MS/MS). Parasitaemia and emergence of gametocytes were monitored by quantitative PCR. Safety was determined by recording adverse events and monitoring clinical laboratory assessments during the course of the study. RESULTS: Eight subjects were enrolled into the study, inoculated with infected erythrocytes and treated with 800 mg ferroquine. Ferroquine was rapidly absorbed with maximal exposure after 4-8 and 4-12 h exposure for SSR97213. Non-compartmental PK analysis resulted in estimates for half-lives of 10.9 and 23.8 days for ferroquine and SSR97213, respectively. Parasite clearance as reported by parasite reduction ratio was 162.9 (95 % CI 141-188) corresponding to a parasite clearance half-life of 6.5 h (95 % CI: 6.4-6.7 h). PK/PD modelling resulted in a predicted minimal parasiticidal concentration of 20 ng/mL, and the single dosing tested in this study was predicted to maintain an exposure above this threshold for 454 h (37.8 days). Although ferroquine was overall well tolerated, transient elevated transaminase levels were observed in three subjects. Paracetamol was the only concomitant treatment among the two out of these three subjects that may have played a role in the elevated transaminases levels. No clinically significant ECG abnormalities were observed. CONCLUSIONS: The parameters and PK/PD model derived from this study pave the way to the further rational development of ferroquine as an anti-malarial partner drug. The safety of ferroquine has to be further explored in controlled human trials. Trial registration anzctr.org.au (registration number: ACTRN12613001040752), registered 18/09/2013.


Subject(s)
Aminoquinolines/administration & dosage , Antimalarials/administration & dosage , Ferrous Compounds/administration & dosage , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Adolescent , Adult , Aminoquinolines/pharmacokinetics , Aminoquinolines/pharmacology , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Blood/parasitology , Blood Chemical Analysis , Chromatography, Liquid , Female , Ferrous Compounds/pharmacokinetics , Ferrous Compounds/pharmacology , Healthy Volunteers , Humans , Male , Metallocenes , Middle Aged , Parasite Load , Plasmodium falciparum/isolation & purification , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry , Treatment Outcome , Young Adult
18.
Br J Clin Pharmacol ; 82(2): 412-21, 2016 08.
Article in English | MEDLINE | ID: mdl-27062080

ABSTRACT

AIMS: The aim of this study was to use data from an experimental induced blood stage malaria clinical trial to characterize the antimalarial activity of the new compound Actelion-451840 using pharmacokinetic/pharmacodynamic (PK/PD) modelling. Then, using simulations from the model, the dose and dosing regimen necessary to achieve cure of infection were derived. METHODS: Eight healthy male subjects were infected with blood stage P. falciparum. After 7 days, a single dose of 500 mg of Actelion-451840 was administered under fed conditions. Parasite and drug concentrations were sampled frequently. Parasite growth and the relation to drug exposure were estimated using PK/PD modelling. Simulations were then undertaken to derive estimates of the likelihood of achieving cure in different scenarios. RESULTS: Actelion-451840 was safe and well tolerated. Single dose treatment markedly reduced the level of P. falciparum parasitaemia, with a weighted average parasite reduction rate of 73.6 (95% CI 56.1, 96.5) and parasite clearance half-life of 7.7 h (95% CI 7.3, 8.3). A two compartment PK/PD model with a steep concentration-kill effect predicted maximum effect with a sustained concentration of 10-15 ng ml(-1) and cure achieved in 90% of subjects with six once daily doses of 300 mg once daily. CONCLUSIONS: Actelion-451840 shows clinical efficacy against P. falciparum infections. The PK/PD model developed from a single proof-of-concept study with eight healthy subjects enabled prediction of therapeutic effects, with cure rates with seven daily doses predicted to be equivalent to artesunate monotherapy. Larger doses or more frequent dosing are not predicted to achieve more rapid cure.


Subject(s)
Acrylamides/administration & dosage , Antimalarials/administration & dosage , Malaria, Falciparum/drug therapy , Models, Biological , Piperazines/administration & dosage , Acrylamides/pharmacokinetics , Acrylamides/pharmacology , Adult , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Half-Life , Humans , Male , Parasitemia/drug therapy , Piperazines/pharmacokinetics , Piperazines/pharmacology , Treatment Outcome , Young Adult
19.
Antimicrob Agents Chemother ; 59(10): 6117-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26195523

ABSTRACT

The 4-aminoquinoline naphthoquine (NQ) and the thiazine dye methylene blue (MB) have potent in vitro efficacies against Plasmodium falciparum, but susceptibility data for P. vivax are limited. The species- and stage-specific ex vivo activities of NQ and MB were assessed using a modified schizont maturation assay on clinical field isolates from Papua, Indonesia, where multidrug-resistant P. falciparum and P. vivax are prevalent. Both compounds were highly active against P. falciparum (median [range] 50% inhibitory concentration [IC50]: NQ, 8.0 nM [2.6 to 71.8 nM]; and MB, 1.6 nM [0.2 to 7.0 nM]) and P. vivax (NQ, 7.8 nM [1.5 to 34.2 nM]; and MB, 1.2 nM [0.4 to 4.3 nM]). Stage-specific drug susceptibility assays revealed significantly greater IC50s in parasites exposed at the trophozoite stage than at the ring stage for NQ in P. falciparum (26.5 versus 5.1 nM, P = 0.021) and P. vivax (341.6 versus 6.5 nM, P = 0.021) and for MB in P. vivax (10.1 versus 1.6 nM, P = 0.010). The excellent ex vivo activities of NQ and MB against both P. falciparum and P. vivax highlight their potential utility for the treatment of multidrug-resistant malaria in areas where both species are endemic.


Subject(s)
Antimalarials/pharmacology , Methylene Blue/pharmacology , Plasmodium falciparum/drug effects , Plasmodium vivax/drug effects
20.
Lancet ; 383(9922): 1049-58, 2014 Mar 22.
Article in English | MEDLINE | ID: mdl-24360369

ABSTRACT

BACKGROUND: Clinical effectiveness of previous regimens to treat Plasmodium vivax infection have been hampered by compliance. We aimed to assess the dose-response, safety, and tolerability of single-dose tafenoquine plus 3-day chloroquine for P vivax malaria radical cure. METHODS: In this double-blind, randomised, dose-ranging phase 2b study, men and women (aged ≥16 years) with microscopically confirmed P vivax monoinfection (parasite density >100 to <100,000 per µL blood) were enrolled from community health centres and hospitals across seven sites in Brazil, Peru, India, and Thailand. Patients with glucose-6-phosphate dehydrogenase enzyme activity of less than 70% were excluded. Eligible patients received chloroquine (days 1-3) and were randomly assigned (1:1:1:1:1:1) by a computer-generated randomisation schedule to receive single-dose tafenoquine 50 mg, 100 mg, 300 mg, or 600 mg, primaquine 15 mg for 14 days, or chloroquine alone. Randomisation was stratified by baseline parasite count (≤7500 and >7500 per µL blood). The primary efficacy endpoint was relapse-free efficacy at 6 months from initial dose (ie, clearance of initial infection without subsequent microscopically confirmed infection), analysed by intention to treat. This study is registered with ClinicalTrials.gov, number NCT01376167. FINDINGS: Between Sept 19, 2011, and March 25, 2013, 329 patients were randomly assigned to a treatment group (chloroquine plus tafenoquine 50 mg [n=55], 100 mg [n=57], 300 mg [n=57], 600 mg [n=56]; or to chloroquine plus primaquine [n=50]; or chloroquine alone [n=54]). Relapse-free efficacy at 6 months was 57·7% (95% CI 43-70) with tafenoquine 50 mg, 54·1% (40-66) with tafenoquine 100 mg, 89·2% (77-95) with tafenoquine 300 mg, 91·9% (80-97) with tafenoquine 600 mg, 77·3% (63-87) with primaquine, and 37·5% (23-52) with chloroquine alone. Tafenoquine 300 mg and 600 mg had better efficacy than chloroquine alone (treatment differences 51·7% [95% CI 35-69], p<0·0001, with tafenoquine 300 mg and 54·5% [38-71], p<0·0001, with tafenoquine 600 mg), as did primaquine (treatment difference 39·9% [21-59], p=0·0004). Adverse events were similar between treatments. 29 serious adverse events occurred in 26 (8%) of 329 patients; QT prolongation was the most common serious adverse event (11 [3%] of 329), occurring in five (2%) of 225 patients receiving tafenoquine, four (8%) of 50 patients receiving primaquine, and two (4%) of 54 patients receiving chloroquine alone, with no evidence of an additional effect on QT of chloroquine plus tafenoquine coadministration. INTERPRETATION: Single-dose tafenoquine 300 mg coadministered with chloroquine for P vivax malaria relapse prevention was more efficacious than chloroquine alone, with a similar safety profile. As a result, it has been selected for further clinical assessment in phase 3. FUNDING: GlaxoSmithKline, Medicines for Malaria Venture.


Subject(s)
Aminoquinolines/administration & dosage , Antimalarials/administration & dosage , Chloroquine/therapeutic use , Malaria, Vivax/drug therapy , Adolescent , Adult , Aged , Brazil , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination , Female , Humans , India , Malaria, Vivax/prevention & control , Male , Middle Aged , Peru , Primaquine/therapeutic use , Secondary Prevention , Thailand , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL