Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Molecules ; 29(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38731597

ABSTRACT

Fibrosis is a ubiquitous pathology, and prior studies have indicated that various artemisinin (ART) derivatives (including artesunate (AS), artemether (AM), and dihydroartemisinin (DHA)) can reduce fibrosis in vitro and in vivo. The medicinal plant Artemisia annua L. is the natural source of ART and is widely used, especially in underdeveloped countries, to treat a variety of diseases including malaria. A. afra contains no ART but is also antimalarial. Using human dermal fibroblasts (CRL-2097), we compared the effects of A. annua and A. afra tea infusions, ART, AS, AM, DHA, and a liver metabolite of ART, deoxyART (dART), on fibroblast viability and expression of key fibrotic marker genes after 1 and 4 days of treatment. AS, DHA, and Artemisia teas reduced fibroblast viability 4 d post-treatment in up to 80% of their respective controls. After 4 d of treatment, AS DHA and Artemisia teas downregulated ACTA2 up to 10 fold while ART had no significant effect, and AM increased viability by 10%. MMP1 and MMP3 were upregulated by AS, 17.5 and 32.6 fold, respectively, and by DHA, 8 and 51.8 fold, respectively. ART had no effect, but A. annua and A. afra teas increased MMP3 5 and 16-fold, respectively. Although A. afra tea increased COL3A1 5 fold, MMP1 decreased >7 fold with no change in either transcript by A. annua tea. Although A. annua contains ART, it had a significantly greater anti-fibrotic effect than ART alone but was less effective than A. afra. Immunofluorescent staining for smooth-muscle α-actin (α-SMA) correlated well with the transcriptional responses of drug-treated fibroblasts. Together, proliferation, qPCR, and immunofluorescence results show that treatment with ART, AS, DHA, and the two Artemisia teas yield differing responses, including those related to fibrosis, in human dermal fibroblasts, with evidence also of remodeling of fibrotic ECM.


Subject(s)
Artemisia , Artemisinins , Fibroblasts , Fibrosis , Humans , Artemisinins/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Artemisia/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Survival/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/genetics , Actins/metabolism , Actins/genetics , Artesunate/pharmacology , Gene Expression Regulation/drug effects , Artemether/pharmacology , Skin/drug effects , Skin/metabolism , Skin/pathology
2.
Acta Trop ; 254: 107196, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38521124

ABSTRACT

BACKGROUND: The drug of choice for the treatment of opisthorchiasis caused by trematodes Opisthorchis viverrini and O. felineus is praziquantel (PZQ), but there is a constant search for new anthelmintics, including those of plant origin. Positive results on the use of artemisinin derivatives against O. viverrini opisthorchiasis have been shown previously, but the effect of these compounds on O. felineus has not been studied. Therefore, here, a comparative analysis of anthelmintic properties of artemisinin derivatives (artesunate [AS], artemether [AM], and dihydroartemisinin [DHA]) was carried out in vitro in relation to PZQ. Experiments were performed on newly excysted metacercariae (NEMs) and adult flukes of O. felineus. RESULTS: Dose- and time-dependent effects of artemisinin derivatives and of PZQ were assessed in terms of motility and mortality of both NEMs and adult flukes. The most pronounced anthelmintic action was exerted by DHA, whose half-maximal inhibitory concentrations (IC50) of 1.9 (NEMs) and 2.02 µg/mL (adult flukes) were lower than those of PZQ (0.56 and 0.25 µg/mL, respectively). In contrast to PZQ, the effects of DHA and AS were similar when we compared the two developmental stages of O. felineus (NEMs and adult flukes). In addition, AM, AS, and especially DHA at doses of 100 µg/mL disrupted tegument integrity in adult flukes, which was not observed with PZQ. CONCLUSIONS: Artemisinin derivatives (AS, AM, and DHA) have good anthelmintic efficacy against the trematode O. felineus, and the action of these substances is comparable to (and sometimes better than) the effects of PZQ.


Subject(s)
Anthelmintics , Artemisinins , Opisthorchis , Animals , Artemisinins/pharmacology , Opisthorchis/drug effects , Anthelmintics/pharmacology , Inhibitory Concentration 50 , Praziquantel/pharmacology , Survival Analysis , Artemether/pharmacology , Artesunate/pharmacology , Dose-Response Relationship, Drug
3.
Lancet Infect Dis ; 24(2): 172-183, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37748496

ABSTRACT

BACKGROUND: Primaquine is used to eliminate Plasmodium vivax hypnozoites, but its optimal dosing regimen remains unclear. We undertook a systematic review and individual patient data meta-analysis to investigate the efficacy and tolerability of different primaquine dosing regimens to prevent P vivax recurrence. METHODS: For this systematic review and individual patient data meta-analysis, we searched MEDLINE, Web of Science, Embase, and Cochrane Central for prospective clinical studies of uncomplicated P vivax from endemic countries published between Jan 1, 2000, and June 8, 2023. We included studies if they had active follow-up of at least 28 days, and if they included a treatment group with daily primaquine given over multiple days, where primaquine was commenced within 7 days of schizontocidal treatment and was given alone or coadministered with chloroquine or one of four artemisinin-based combination therapies (ie, artemether-lumefantrine, artesunate-mefloquine, artesunate-amodiaquine, or dihydroartemisinin-piperaquine). We excluded studies if they were on prevention, prophylaxis, or patients with severe malaria, or if data were extracted retrospectively from medical records outside of a planned trial. For the meta-analysis, we contacted the investigators of eligible trials to request individual patient data and we then pooled data that were made available by Aug 23, 2021. We assessed the effects of total dose and duration of primaquine regimens on the rate of first P vivax recurrence between day 7 and day 180 by Cox's proportional hazards regression (efficacy analysis). The effect of primaquine daily dose on gastrointestinal symptoms on days 5-7 was assessed by modified Poisson regression (tolerability analysis). The study was registered with PROSPERO, CRD42019154470. FINDINGS: Of 226 identified studies, 23 studies with patient-level data from 6879 patients from 16 countries were included in the efficacy analysis. At day 180, the risk of recurrence was 51·0% (95% CI 48·2-53·9) in 1470 patients treated without primaquine, 19·3% (16·9-21·9) in 2569 patients treated with a low total dose of primaquine (approximately 3·5 mg/kg), and 8·1% (7·0-9·4) in 2811 patients treated with a high total dose of primaquine (approximately 7 mg/kg), regardless of primaquine treatment duration. Compared with treatment without primaquine, the rate of P vivax recurrence was lower after treatment with low-dose primaquine (adjusted hazard ratio 0·21, 95% CI 0·17-0·27; p<0·0001) and high-dose primaquine (0·10, 0·08-0·12; p<0·0001). High-dose primaquine had greater efficacy than low-dose primaquine in regions with high and low relapse periodicity (ie, the time from initial infection to vivax relapse). 16 studies with patient-level data from 5609 patients from ten countries were included in the tolerability analysis. Gastrointestinal symptoms on days 5-7 were reported by 4·0% (95% CI 0·0-8·7) of 893 patients treated without primaquine, 6·2% (0·5-12·0) of 737 patients treated with a low daily dose of primaquine (approximately 0·25 mg/kg per day), 5·9% (1·8-10·1) of 1123 patients treated with an intermediate daily dose (approximately 0·5 mg/kg per day) and 10·9% (5·7-16·1) of 1178 patients treated with a high daily dose (approximately 1 mg/kg per day). 20 of 23 studies included in the efficacy analysis and 15 of 16 in the tolerability analysis had a low or unclear risk of bias. INTERPRETATION: Increasing the total dose of primaquine from 3·5 mg/kg to 7 mg/kg can reduce P vivax recurrences by more than 50% in most endemic regions, with a small associated increase in gastrointestinal symptoms. FUNDING: Australian National Health and Medical Research Council, Bill & Melinda Gates Foundation, and Medicines for Malaria Venture.


Subject(s)
Antimalarials , Malaria, Vivax , Malaria , Humans , Primaquine/therapeutic use , Antimalarials/adverse effects , Plasmodium vivax , Artesunate/therapeutic use , Prospective Studies , Retrospective Studies , Artemether/pharmacology , Artemether/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Australia , Malaria, Vivax/drug therapy , Malaria, Vivax/prevention & control , Malaria, Vivax/epidemiology , Malaria/drug therapy , Recurrence
4.
Clin Infect Dis ; 78(2): 445-452, 2024 02 17.
Article in English | MEDLINE | ID: mdl-38019958

ABSTRACT

BACKGROUND: Recent cases of clinical failure in malaria patients in the United Kingdom (UK) treated with artemether-lumefantrine have implications for malaria chemotherapy worldwide. METHODS: Parasites were isolated from an index case of confirmed Plasmodium falciparum treatment failure after standard treatment, and from comparable travel-acquired UK malaria cases. Drug susceptibility in vitro and genotypes at 6 resistance-associated loci were determined for all parasite isolates and compared with clinical outcomes for each parasite donor. RESULTS: A traveler, who returned to the UK from Uganda in 2022 with Plasmodium falciparum malaria, twice failed treatment with full courses of artemether-lumefantrine. Parasites from the patient exhibited significantly reduced susceptibility to artemisinin (ring-stage survival, 17.3% [95% confidence interval {CI}, 13.6%-21.1%]; P < .0001) and lumefantrine (effective concentration preventing 50% of growth = 259.4 nM [95% CI, 130.6-388.2 nM]; P = .001). Parasite genotyping identified an allele of pfk13 encoding both the A675V variant in the Pfk13 propeller domain and a novel L145V nonpropeller variant. In vitro susceptibility testing of 6 other P. falciparum lines of Ugandan origin identified reduced susceptibility to artemisinin and lumefantrine in 1 additional line, also from a 2022 treatment failure case. These parasites did not harbor a pfk13 propeller domain variant but rather the novel nonpropeller variant T349I. Variant alleles of pfubp1, pfap2mu, and pfcoronin were also identified among the 7 parasite lines. CONCLUSIONS: We confirm, in a documented case of artemether-lumefantrine treatment failure imported from Uganda, the presence of pfk13 mutations encoding L145V and A675V. Parasites with reduced susceptibility to both artemisinin and lumefantrine may be emerging in Uganda.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Malaria , Humans , Lumefantrine/pharmacology , Lumefantrine/therapeutic use , Plasmodium falciparum , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemether, Lumefantrine Drug Combination/pharmacology , Artemether, Lumefantrine Drug Combination/therapeutic use , Uganda , Drug Resistance , Artemether/pharmacology , Artemether/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Treatment Failure , United Kingdom , Protozoan Proteins/genetics
5.
J Travel Med ; 31(3)2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38157311

ABSTRACT

BACKGROUND: Failure of artemisinin-based combination therapy is increasingly reported in patients with Plasmodium falciparum malaria in sub-Saharan Africa. We aimed to describe the clinical and genomic characteristics of recent cases of P. falciparum malaria failing artemether-lumefantrine in Belgium. METHODS: Travel-related cases of malaria confirmed at the national reference laboratory of the Institute of Tropical Medicine, Antwerp, Belgium, were reviewed. All cases for which attending clinicians reported persistence (beyond Day 3 post-treatment initiation, i.e. early failure) or recrudescence (from Day 7 to 42, i.e. late failure) of P. falciparum parasites despite adequate drug intake were analysed. Both initial and persistent/recurrent samples were submitted to next generation sequencing to investigate resistance-conferring mutations. RESULTS: From July 2022 to June 2023, eight P. falciparum cases of failure with artemether-lumefantrine therapy were reported (early failure = 1; late failure = 7). All travellers were returning from sub-Saharan Africa, most (6/8) after a trip to visit friends and relatives. PfKelch13 (PF3D7_1343700) mutations associated with resistance to artemisinin were found in two travellers returning from East Africa, including the validated marker R561H in the patient with early failure and the candidate marker A675V in a patient with late failure. Additional mutations were detected that could contribute to decreased susceptibility to artemisinin in another three cases, lumefantrine in six cases and proguanil in all eight participants. Various regimens were used to treat the persistent/recrudescent cases, with favourable outcome. CONCLUSION: Within a 12-month period, we investigated eight travellers returning from sub-Saharan Africa with P. falciparum malaria and in whom artemether-lumefantrine failure was documented. Mutations conferring resistance to antimalarials were found in all analysed blood samples, especially against lumefantrine and proguanil, but also artemisinin. There is a pressing need for systematic genomic surveillance of resistance to antimalarials in international travellers with P. falciparum malaria, especially those experiencing treatment failure.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Malaria , Humans , Antimalarials/pharmacology , Artemether/pharmacology , Artemether, Lumefantrine Drug Combination/pharmacology , Artemisinins/pharmacology , Belgium , Drug Combinations , Genomics , Lumefantrine/pharmacology , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Malaria, Falciparum/epidemiology , Plasmodium falciparum/genetics , Proguanil/pharmacology , Travel , Travel-Related Illness
6.
Nutrients ; 15(23)2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38068719

ABSTRACT

Intestinal inflammation is a key determinant of intestinal and systemic health, and when our intestines are damaged, there is disruption of the intestinal barrier, which in turn induces a systemic inflammatory response. However, the etiology and pathogenesis of inflammatory diseases of the intestine are still not fully understood. Artemether (ART), one of the artemisinin derivatives, has been widely used to treat malaria. Nevertheless, the effect of ART on intestinal inflammation remains unclear. The present study intended to elucidate the potential mechanism of ART in diet-induced intestinal injury. A high-fat and high-fructose (HFHF) diet-induced mouse model of intestinal injury was constructed, and the mice were treated with ART to examine their role in intestinal injury. RT-qPCR, Western blotting, immunohistochemical staining, and 16S rRNA gene sequencing were used to investigate the anti-intestinal inflammation effect and mechanism of ART. The results indicated that ART intervention may significantly ameliorate the intestinal flora imbalance caused by the HFHF diet and alleviate intestinal barrier function disorders and inflammatory responses by raising the expression of tight junction proteins ZO-1 and occludin and decreasing the expression of pro-inflammatory factors TNF-α and IL-1ß. Moreover, ART intervention restrained HFHF-induced activation of the TLR4/NF-κB p65 pathway in colon tissue, which may be concerned with the potential protective effect of ART on intestinal inflammation. ART might provide new insights into further explaining the mechanism of action of other metabolic diseases caused by intestinal disorders.


Subject(s)
Gastrointestinal Microbiome , Mice , Animals , Artemether/pharmacology , Fructose/adverse effects , RNA, Ribosomal, 16S , Diet , Inflammation/drug therapy , Inflammation/pathology , Mice, Inbred C57BL , Diet, High-Fat/adverse effects
7.
Lancet ; 402(10417): 2101-2110, 2023 12 02.
Article in English | MEDLINE | ID: mdl-37979594

ABSTRACT

BACKGROUND: In areas co-endemic for Plasmodium vivax and Plasmodium falciparum there is an increased risk of P vivax parasitaemia following P falciparum malaria. Radical cure is currently only recommended for patients presenting with P vivax malaria. Expanding the indication for radical cure to patients presenting with P falciparum malaria could reduce their risk of subsequent P vivax parasitaemia. METHODS: We did a multicentre, open-label, superiority randomised controlled trial in five health clinics in Bangladesh, Indonesia, and Ethiopia. In Bangladesh and Indonesia, patients were excluded if they were younger than 1 year, whereas in Ethiopia patients were excluded if they were younger than 18 years. Patients with uncomplicated P falciparum monoinfection who had fever or a history of fever in the 48 h preceding clinic visit were eligible for enrolment and were required to have a glucose-6-dehydrogenase (G6PD) activity of 70% or greater. Patients received blood schizontocidal treatment (artemether-lumefantrine in Ethiopia and Bangladesh and dihydroartemisinin-piperaquine in Indonesia) and were randomly assigned (1:1) to receive either high-dose short-course oral primaquine (intervention arm; total dose 7 mg/kg over 7 days) or standard care (standard care arm; single dose oral primaquine of 0·25 mg/kg). Random assignment was done by an independent statistician in blocks of eight by use of sealed envelopes. All randomly assigned and eligible patients were included in the primary and safety analyses. The per-protocol analysis excluded those who did not complete treatment or had substantial protocol violations. The primary endpoint was the incidence risk of P vivax parasitaemia on day 63. This trial is registered at ClinicalTrials.gov, NCT03916003. FINDINGS: Between Aug 18, 2019, and March 14, 2022, a total of 500 patients were enrolled and randomly assigned, and 495 eligible patients were included in the intention-to-treat analysis (246 intervention and 249 control). The incidence risk of P vivax parasitaemia at day 63 was 11·0% (95% CI 7·5-15·9) in the standard care arm compared with 2·5% (1·0-5·9) in the intervention arm (hazard ratio 0·20, 95% CI 0·08-0·51; p=0·0009). The effect size differed with blood schizontocidal treatment and site. Routine symptom reporting on day 2 and day 7 were similar between groups. In the first 42 days, there were a total of four primaquine-related adverse events reported in the standard care arm and 26 in the intervention arm; 132 (92%) of all 143 adverse events were mild. There were two serious adverse events in the intervention arm, which were considered unrelated to the study drug. None of the patients developed severe anaemia (defined as haemoglobin <5 g/dL). INTERPRETATION: In patients with a G6PD activity of 70% or greater, high-dose short-course primaquine was safe and relatively well tolerated and reduced the risk of subsequent P vivax parasitaemia within 63 days by five fold. Universal radical cure therefore potentially offers substantial clinical, public health, and operational benefits, but these benefits will vary with endemic setting. FUNDING: Australian Academy of Science Regional Collaborations Program, Bill & Melinda Gates Foundation, and National Health and Medical Research Council.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria, Vivax , Malaria , Humans , Primaquine/adverse effects , Antimalarials/adverse effects , Plasmodium vivax , Artemether/pharmacology , Artemether/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Australia , Malaria, Falciparum/drug therapy , Malaria, Falciparum/epidemiology , Malaria, Vivax/drug therapy , Malaria, Vivax/epidemiology , Malaria/drug therapy , Plasmodium falciparum , Parasitemia/drug therapy , Parasitemia/epidemiology
8.
Int J Antimicrob Agents ; 62(6): 107012, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37865152

ABSTRACT

BACKGROUND: The clinical use of artemisinin-based combination therapies is threatened by increasing failure rates due to the emergence and spread of multiple drug resistance genes in most human Plasmodium strains. The aim of this study was to generate artemether-resistant (AMR) parasites from Plasmodium berghei ANKA (AMS), and determine their fitness cost. METHODS: Artemether resistance was generated by increasing drug pressure doses gradually for 9 months. Effective doses (ED50 and ED90) were determined using the 4-day suppressive test, and the indices of resistance (I) at 50% and 90% (I50 and I90) were determined using the ratio of either ED50 or ED90 of AMR to AMS, respectively. The stability of the AMR parasites was evaluated by: five drug-free passages (5DFPs), 3 months of cryopreservation (CP), and drug-free serial passages (DFSPs) for 4 months. Analysis of variance was used to compare differences in growth rates between AMR and AMS with 95% confidence intervals. RESULTS: ED50 and ED90 of AMS were 0.61 and 3.43 mg/kg/day respectively. I50 and I90 after 20 cycles of artemether selection pressure were 19.67 and 21.45, respectively; 5DFP values were 39.16 and 15.27, respectively; 3-month CP values were 29.36 and 10.79, respectively; and DFSP values were 31.34 and 12.29, respectively. The mean parasitaemia value of AMR (24.70% ± 3.60) relative to AMS (37.66% ± 3.68) at Day 7 post infection after DFSPs revealed a fitness cost of 34.41%. CONCLUSION: A moderately stable AMRP. berghei line was generated. Known and unknown mutations may be involved in modulating artemether resistance, and therefore molecular investigations are recommended.


Subject(s)
Antimalarials , Malaria , Parasites , Animals , Humans , Artemether/pharmacology , Artemether/therapeutic use , Antimalarials/pharmacology , Antimalarials/therapeutic use , Malaria/drug therapy , Plasmodium berghei/genetics , Plasmodium falciparum , Drug Resistance
9.
Mikrobiyol Bul ; 57(3): 498-505, 2023 Jul.
Article in Turkish | MEDLINE | ID: mdl-37462313

ABSTRACT

Malaria is a serious, contagious infection caused by single-celled parasites. About 200 species of Plasmodium have been described that can cause infection in vertebrates. Five different species of Plasmodium are known to cause infection in humans to date. Infection with more than one type of pathogen is called coinfection. This type of infections can be caused by different species of the same genus, as well as by different species. Malaria coinfections are mostly caused by the combination of Plasmodium vivax and Plasmodium falciparum. In this study, a case of malaria admitted to the hospital and diagnosed was presented. Thin smear blood preparations were prepared from the peripheral blood of a 54 year-old Republic of Türkiye citizen male patient who applied to the emergency department with fever and chills. The preparations were stained with Giemsa and examined under a microscope with a x 100 objective, and trophozoite and gametocyte forms belonging to Plasmodium genus were determined. As a result of probe-based quantitative real-time polymerase chain reaction (qRt-PCR) study with primers specific to Plasmodium vivax, Plasmodium malariae, Plasmodium falciparum, Plasmodium ovale and Plasmodium knowlesi for definitive species identification, co-infection of P.vivax, P.falciparum, P.ovale and P.knowlesi was detected in the patient. In addition, it was proved that our patient was infected with four different species by conventional PCR study in which five species were studied and then by DNA sequence analysis. On the fourth day of artemether-lumefantrine treatment, the patient's fever response was observed and the trophozoite forms disappeared from the third day in the daily peripheral smear follow-up. Since P.vivax and P.ovale species were also detected after species determination by molecular methods, primaquine 1 x 30 mg tablet was added to the existing drugs for the treatment of hypnozoite forms of the parasite. In recent years, there has been an increase in malaria imported cases, especially after visits to African countries. Such rare cases of malaria coinfection may be encountered during visits to geographies located at the intersection of endemic regions. According to the data of the World Health Organization, maximum attention should be paid to the prevention and prophylaxis protocols from vectors, especially in travels to countries with the highest mortality and morbidity. In co-infection cases similar to our patient, for tertian malaria and tertiary ovale malaria, hypnozoid therapy should not be overlooked. When the insecticide-resistant vectors and drug-resistant Plasmodium strains encountered in recent years are evaluated as a whole, there is a need to develop more effective strategies in the fight against malaria. In addition to microscopic examination, which is accepted as the gold standard, we believe that evaluating molecular studies together in diagnosis is extremely important for the treatment process when hypnozoite periods are considered.


Subject(s)
Antimalarials , Coinfection , Malaria, Vivax , Malaria , Plasmodium , Animals , Male , Humans , Middle Aged , Coinfection/drug therapy , Coinfection/parasitology , Antimalarials/therapeutic use , Antimalarials/pharmacology , Cameroon , Artemether/pharmacology , Artemether/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Malaria/complications , Malaria/diagnosis , Malaria/drug therapy , Malaria, Vivax/diagnosis , Plasmodium/genetics , Plasmodium falciparum/genetics , Plasmodium vivax/genetics , Real-Time Polymerase Chain Reaction
10.
Lancet Infect Dis ; 23(9): 1051-1061, 2023 09.
Article in English | MEDLINE | ID: mdl-37327809

ABSTRACT

BACKGROUND: Emergence of drug resistance demands novel antimalarial drugs with new mechanisms of action. We aimed to identify effective and well tolerated doses of ganaplacide plus lumefantrine solid dispersion formulation (SDF) in patients with uncomplicated Plasmodium falciparum malaria. METHODS: This open-label, multicentre, parallel-group, randomised, controlled, phase 2 trial was conducted at 13 research clinics and general hospitals in ten African and Asian countries. Patients had microscopically-confirmed uncomplicated P falciparum malaria (>1000 and <150 000 parasites per µL). Part A identified the optimal dose regimens in adults and adolescents (aged ≥12 years) and in part B, the selected doses were assessed in children (≥2 years and <12 years). In part A, patients were randomly assigned to one of seven groups (once a day ganaplacide 400 mg plus lumefantrine-SDF 960 mg for 1, 2, or 3 days; ganaplacide 800 mg plus lumefantrine-SDF 960 mg as a single dose; once a day ganaplacide 200 mg plus lumefantrine-SDF 480 mg for 3 days; once a day ganaplacide 400 mg plus lumefantrine-SDF 480 mg for 3 days; or twice a day artemether plus lumefantrine for 3 days [control]), with stratification by country (2:2:2:2:2:2:1) using randomisation blocks of 13. In part B, patients were randomly assigned to one of four groups (once a day ganaplacide 400 mg plus lumefantrine-SDF 960 mg for 1, 2, or 3 days, or twice a day artemether plus lumefantrine for 3 days) with stratification by country and age (2 to <6 years and 6 to <12 years; 2:2:2:1) using randomisation blocks of seven. The primary efficacy endpoint was PCR-corrected adequate clinical and parasitological response at day 29, analysed in the per protocol set. The null hypothesis was that the response was 80% or lower, rejected when the lower limit of two-sided 95% CI was higher than 80%. This study is registered with EudraCT (2020-003284-25) and ClinicalTrials.gov (NCT03167242). FINDINGS: Between Aug 2, 2017, and May 17, 2021, 1220 patients were screened and of those, 12 were included in the run-in cohort, 337 in part A, and 175 in part B. In part A, 337 adult or adolescent patients were randomly assigned, 326 completed the study, and 305 were included in the per protocol set. The lower limit of the 95% CI for PCR-corrected adequate clinical and parasitological response on day 29 was more than 80% for all treatment regimens in part A (46 of 50 patients [92%, 95% CI 81-98] with 1 day, 47 of 48 [98%, 89-100] with 2 days, and 42 of 43 [98%, 88-100] with 3 days of ganaplacide 400 mg plus lumefantrine-SDF 960 mg; 45 of 48 [94%, 83-99] with ganaplacide 800 mg plus lumefantrine-SDF 960 mg for 1 day; 47 of 47 [100%, 93-100] with ganaplacide 200 mg plus lumefantrine-SDF 480 mg for 3 days; 44 of 44 [100%, 92-100] with ganaplacide 400 mg plus lumefantrine-SDF 480 mg for 3 days; and 25 of 25 [100%, 86-100] with artemether plus lumefantrine). In part B, 351 children were screened, 175 randomly assigned (ganaplacide 400 mg plus lumefantrine-SDF 960 mg once a day for 1, 2, or 3 days), and 171 completed the study. Only the 3-day regimen met the prespecified primary endpoint in paediatric patients (38 of 40 patients [95%, 95% CI 83-99] vs 21 of 22 [96%, 77-100] with artemether plus lumefantrine). The most common adverse events were headache (in seven [14%] of 51 to 15 [28%] of 54 in the ganaplacide plus lumefantrine-SDF groups and five [19%] of 27 in the artemether plus lumefantrine group) in part A, and malaria (in 12 [27%] of 45 to 23 [44%] of 52 in the ganaplacide plus lumefantrine-SDF groups and 12 [50%] of 24 in the artemether plus lumefantrine group) in part B. No patients died during the study. INTERPRETATION: Ganaplacide plus lumefantrine-SDF was effective and well tolerated in patients, especially adults and adolescents, with uncomplicated P falciparum malaria. Ganaplacide 400 mg plus lumefantrine-SDF 960 mg once daily for 3 days was identified as the optimal treatment regimen for adults, adolescents, and children. This combination is being evaluated further in a phase 2 trial (NCT04546633). FUNDING: Novartis and Medicines for Malaria Venture.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Malaria , Adult , Adolescent , Child , Humans , Lumefantrine/pharmacology , Lumefantrine/therapeutic use , Fluorenes/therapeutic use , Fluorenes/pharmacology , Ethanolamines/therapeutic use , Ethanolamines/pharmacology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Artemether/pharmacology , Artemether/therapeutic use , Malaria/drug therapy , Drug Combinations , Plasmodium falciparum , Treatment Outcome
11.
Mol Med Rep ; 28(2)2023 Aug.
Article in English | MEDLINE | ID: mdl-37387406

ABSTRACT

Adriamycin is a widely used and effective antitumor drug; however, its application is limited by various side effects, including irreversible cardiotoxicity. The central role of cardiac atrophy in Adriamycin­induced cardiotoxicity has been revealed; however, the underlying mechanism of this process remains unclear. Artemether is a well­known Chinese herbal medicine, and its pharmacological action is related to the regulation of mitochondrial function and redox status. The present study determined the effects of artemether on Adriamycin­induced cardiotoxicity and investigated the underlying mechanisms. After mouse model establishment and artemether intervention, experimental methods including pathological staining, immunohistochemistry, immunofluorescence, immunoblotting, ELISA and reverse transcription­quantitative PCR were used to evaluate the therapeutic effect. The results demonstrated that artemether prevented Adriamycin­induced cardiac atrophy and recovered the intercombination of connexin 43 and N­cadherin at the intercalated discs. Artemether also regulated the autophagy pathway and restored the unbalanced ratio of Bax and Bcl­2 in myocardial cells. In addition, the increased serum H2O2 levels after Adriamycin exposure were significantly decreased by artemether, and the mitochondrial alterations and redox imbalance in myocardial cells were also improved to varying degrees. In summary, the findings of the present study provide reliable evidence that artemether could ameliorate cardiac atrophy induced by Adriamycin. This therapeutic approach may be translated to the clinic for preventing drug­induced heart diseases.


Subject(s)
Cardiotoxicity , Doxorubicin , Animals , Mice , Doxorubicin/adverse effects , Hydrogen Peroxide , Myocytes, Cardiac , Artemether/pharmacology , Atrophy
12.
Parasitol Res ; 122(8): 1841-1850, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37256314

ABSTRACT

This study investigated the effects of co-administration of a commercial juice rich in vitamin C (Vit C) on the antimalarial efficacy of artemether-lumefantrine (AL) in Plasmodium berghei-infected mice. Fifty Balb/c mice were infected with Plasmodium berghei NK65 strain from a donor mouse. Parasitemia was established after 72 h. Animals were grouped into 6 (n = 10) and treated daily for 3 days with normal saline, chloroquine, artemether-lumefantrine (AL), AL plus 50% commercial juice (CJ), and AL plus 50% Vit C supplementation in drinks ad libitum, respectively. Body weight, parasitemia levels, and mean survival time were determined. Tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), nitrite, malondialdehyde, reduced glutathione (GSH), catalase, and superoxide dismutase (SOD) were determined in the serum and liver tissues. Spleen histopathological changes were determined by H&E staining. Parasitemia was cleared by administration of AL and was not affected by Vit C and CJ supplementation. Vit C significantly prevented body weight reduction in AL-treated mice. CJ and Vit C supplementation to AL-treated mice significantly improved survival proportion compared with AL alone animals. Vit C and CJ supplementation significantly improved reduction of TNF-α, IL-6, and malondialdehyde, and increased GSH, CAT, and SOD in AL-treated mice. Spleen cell degeneration and presence of malaria pigment were reduced in AL-treated animals. The results suggest that ad libitum co-administration of commercial juice and vitamin C with artemether-lumefantrine does not impair its antimalarial efficacy but rather improved antioxidant and anti-inflammatory effects in mice.


Subject(s)
Antimalarials , Malaria , Animals , Mice , Antimalarials/therapeutic use , Antimalarials/pharmacology , Artemether, Lumefantrine Drug Combination/pharmacology , Artemether, Lumefantrine Drug Combination/therapeutic use , Plasmodium berghei , Artemether/pharmacology , Artemether/therapeutic use , Malaria/drug therapy , Malaria/pathology , Ascorbic Acid/pharmacology , Parasitemia/drug therapy , Interleukin-6 , Tumor Necrosis Factor-alpha , Superoxide Dismutase , Malondialdehyde
13.
Clin Infect Dis ; 76(11): 1919-1927, 2023 06 08.
Article in English | MEDLINE | ID: mdl-36795050

ABSTRACT

BACKGROUND: The long-acting 8-aminoquinoline tafenoquine may be a good candidate for mass drug administration if it exhibits sufficient blood-stage antimalarial activity at doses low enough to be tolerated by glucose 6-phosphate dehydrogenase (G6PD)-deficient individuals. METHODS: Healthy adults with normal levels of G6PD were inoculated with Plasmodium falciparum 3D7-infected erythrocytes on day 0. Different single oral doses of tafenoquine were administered on day 8. Parasitemia and concentrations of tafenoquine and the 5,6-orthoquinone metabolite in plasma/whole blood/urine were measured and standard safety assessments performed. Curative artemether-lumefantrine therapy was administered if parasite regrowth occurred, or on day 48 ± 2. Outcomes were parasite clearance kinetics, pharmacokinetic and pharmacokinetic/pharmacodynamic (PK/PD) parameters from modelling, and dose simulations in a theoretical endemic population. RESULTS: Twelve participants were inoculated and administered 200 mg (n = 3), 300 mg (n = 4), 400 mg (n = 2), or 600 mg (n = 3) tafenoquine. The parasite clearance half-life with 400 mg or 600 mg (5.4 hours and 4.2 hours, respectively) was faster than with 200 mg or 300 mg (11.8 hours and 9.6 hours, respectively). Parasite regrowth occurred after dosing with 200 mg (3/3 participants) and 300 mg (3/4 participants) but not after 400 mg or 600 mg. Simulations using the PK/PD model predicted that 460 mg and 540 mg would clear parasitaemia by a factor of 106 and 109, respectively, in a 60-kg adult. CONCLUSIONS: Although a single dose of tafenoquine exhibits potent P. falciparum blood-stage antimalarial activity, the estimated doses to effectively clear asexual parasitemia will require prior screening to exclude G6PD deficiency. Clinical Trials Registration. Australian and New Zealand Clinical Trials Registry (ACTRN12620000995976).


Subject(s)
Antimalarials , Malaria, Falciparum , Adult , Humans , Antimalarials/adverse effects , Plasmodium falciparum , Healthy Volunteers , Parasitemia/drug therapy , Artemether/pharmacology , Artemether/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Australia , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology
14.
J Nanobiotechnology ; 21(1): 15, 2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36647056

ABSTRACT

BACKGROUND: Malaria remains a serious threat to global public health. With poor efficacies of vaccines and the emergence of drug resistance, novel strategies to control malaria are urgently needed. RESULTS: We developed erythrocyte membrane-camouflaged nanoparticles loaded with artemether based on the growth characteristics of Plasmodium. The nanoparticles could capture the merozoites to inhibit them from repeatedly infecting normal erythrocytes, owing to the interactions between merozoites and heparin-like molecules on the erythrocyte membrane. Modification with a phosphatidylserine-targeting peptide (CLIPPKF) improved the drug accumulation in infected red blood cells (iRBCs) from the externalized phosphatidylserine induced by Plasmodium infection. In Plasmodium berghei ANKA strain (pbANKA)-infected C57BL/6 mice, the nanoparticles significantly attenuated Plasmodium-induced inflammation, apoptosis, and anemia. We observed reduced weight variation and prolonged survival time in pbANKA-challenged mice, and the nanoparticles showed good biocompatibility and negligible cytotoxicity. CONCLUSION: Erythrocyte membrane-camouflaged nanoparticles loaded with artemether were shown to provide safe and effective protection against Plasmodium infection.


Subject(s)
Malaria , Merozoites , Animals , Mice , Erythrocyte Membrane , Phosphatidylserines , Biomimetics , Mice, Inbred C57BL , Malaria/drug therapy , Malaria/prevention & control , Erythrocytes , Artemether/pharmacology , Plasmodium berghei , Plasmodium falciparum
15.
Mycotoxin Res ; 39(1): 67-80, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36701108

ABSTRACT

Populations in malaria endemic areas are frequently exposed to mycotoxin-contaminated diets. The possible toxicological outcome of co-exposure to dietary aflatoxin B1 (AFB1) and artemisinin-based combination therapy warrants investigation to ascertain amplification or attenuation of cellular injury. Here, we investigated the neurobehavioral and biochemical responses associated with co-exposure to anti-malarial drug coartem, an artemether-lumefantrine combination (5 mg/kg body weight, twice a day and 3 days per week) and AFB1 (35 and 70 µg/kg body weight) in rats. Motor deficits, locomotor incompetence, and anxiogenic-like behavior induced by low AFB1 dose were significantly (p < 0.05) assuaged by coartem but failed to rescue these behavioral abnormalities in high AFB1-dosed group. Coartem administration did not alter exploratory deficits typified by reduced track plot densities and greater heat map intensity in high AFB1-dosed animals. Furthermore, the reduction in cerebral and cerebellar acetylcholinesterase activity, anti-oxidant enzyme activities, and glutathione and thiol levels were markedly assuaged by coartem administration in low AFB1 group but not in high AFB1-dosed animals. The significant attenuation of cerebral and cerebellar oxidative stress indices namely reactive oxygen and nitrogen species, xanthine oxidase activity, and lipid peroxidation by coartem administration was evident in low AFB1 group but not high AFB1 dose. Although coartem administration abated nitric oxide level, activities of myeloperoxidase, caspase-9, and caspase-3 in animals exposed to both doses of AFB1, these indices were significantly higher than the control. Coartem administration ameliorated histopathological and mophometrical changes due to low AFB1 exposure but not in high AFB1 exposure. In conclusion, contrary to AFB1 alone, behavioral and biochemical responses were not altered in animals singly exposed to coartem. Co-exposure to coartem and AFB1 elicited no additional risk but partially lessened neurotoxicity associated with AFB1 exposure.


Subject(s)
Antimalarials , Artemisinins , Rats , Animals , Aflatoxin B1/toxicity , Aflatoxin B1/metabolism , Pharmaceutical Preparations/metabolism , Antimalarials/metabolism , Antimalarials/pharmacology , Artemether, Lumefantrine Drug Combination/metabolism , Artemether, Lumefantrine Drug Combination/pharmacology , Acetylcholinesterase/metabolism , Acetylcholinesterase/pharmacology , Artemether/metabolism , Artemether/pharmacology , Oxidative Stress , Artemisinins/metabolism , Artemisinins/pharmacology , Body Weight , Liver/metabolism
16.
Clin Infect Dis ; 76(3): 506-512, 2023 02 08.
Article in English | MEDLINE | ID: mdl-35731843

ABSTRACT

BACKGROUND: Blocking the transmission of parasites from humans to mosquitoes is a key component of malaria control. Tafenoquine exhibits activity against all stages of the malaria parasite and may have utility as a transmission blocking agent. We aimed to characterize the transmission blocking activity of low-dose tafenoquine. METHODS: Healthy adults were inoculated with Plasmodium falciparum 3D7-infected erythrocytes on day 0. Piperaquine was administered on days 9 and 11 to clear asexual parasitemia while allowing gametocyte development. A single 50-mg oral dose of tafenoquine was administered on day 25. Transmission was determined by enriched membrane feeding assays predose and at 1, 4, and 7 days postdose. Artemether-lumefantrine was administered following the final assay. Outcomes were the reduction in mosquito infection and gametocytemia after tafenoquine and safety parameters. RESULTS: Six participants were enrolled, and all were infective to mosquitoes before tafenoquine, with a median 86% (range, 22-98) of mosquitoes positive for oocysts and 57% (range, 4-92) positive for sporozoites. By day 4 after tafenoquine, the oocyst and sporozoite positivity rate had reduced by a median 35% (interquartile range [IQR]: 16-46) and 52% (IQR: 40-62), respectively, and by day 7, 81% (IQR 36-92) and 77% (IQR 52-98), respectively. The decline in gametocyte density after tafenoquine was not significant. No significant participant safety concerns were identified. CONCLUSIONS: Low-dose tafenoquine (50 mg) reduces P. falciparum transmission to mosquitoes, with a delay in effect.


Subject(s)
Anopheles , Antimalarials , Malaria, Falciparum , Malaria , Adult , Animals , Humans , Plasmodium falciparum , Antimalarials/adverse effects , Healthy Volunteers , Artemether/pharmacology , Artemether, Lumefantrine Drug Combination , Malaria, Falciparum/prevention & control , Sporozoites , Anopheles/parasitology
17.
Drug Dev Res ; 84(2): 156-171, 2023 04.
Article in English | MEDLINE | ID: mdl-36541217

ABSTRACT

Artemether (ATM) is a natural antimalarial drug that can also regulate glucose and lipid metabolism. However, little is known regarding its pharmacological action in metabolic dysfunction-associated fatty liver disease (MAFLD), and the underlying mechanisms remain undetermined. The aim of this study was to explore the therapeutic effects of ATM against hepatic steatosis and the possible mechanisms. ATM significantly decreased blood glucose levels, improved glucose tolerance, reduced inflammatory response, and alleviated hepatic steatosis in the ob/ob mouse model as well as the high-fat diet-fed mice. ATM also inhibited lipid accumulation in murine hepatocytes in vitro. Using RNA sequencing, miR-34a-5p and peroxisome proliferator-activated receptor-α (PPARα) were identified as important regulators during ATM treatment. ATM administration downregulated miR-34a-5p expression and miR-34a-5p abrogated the inhibitory effects of ATM on PO (palmitate + oleate)-induced lipid accumulation as well as triglycerides levels in murine hepatocytes. Furthermore, the expression of PPARα, a target gene of miR-34a-5p, was upregulated by ATM and PPARα inhibitor MK-886 abolished the positive effect of ATM. Consequently, PPARα agonist fenofibrate reversed the decreased mitochondrial fatty acid ß-oxidation induced by miR-34a-5p mimics after ATM treatment, thereby leading to attenuation of intracellular lipid accumulation. Taken together, ATM is a promising therapeutic agent against MAFLD that reduces lipid deposition by suppressing miR-34a-5p and upregulating PPARα.


Subject(s)
MicroRNAs , Non-alcoholic Fatty Liver Disease , Mice , Animals , PPAR alpha/genetics , PPAR alpha/metabolism , Artemether/pharmacology , Artemether/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Lipid Metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Lipids , Glucose/metabolism , Liver , Mice, Inbred C57BL
18.
J Pharm Pharmacol ; 74(12): 1758-1764, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36206181

ABSTRACT

OBJECTIVES: The antimalarial drug artemether is suggested to effect pancreatic islet cell transdifferentiation, presumably through activation γ-aminobutyric acid receptors, but this biological action is contested. METHODS: We have investigated changes in α-cell lineage in response to 10-days treatment with artemether (100 mg/kg oral, once daily) on a background of ß-cell stress induced by multiple low-dose streptozotocin (STZ) injection in GluCreERT2; ROSA26-eYFP transgenic mice. KEY FINDINGS: Artemether intervention did not affect the actions of STZ on body weight, food and fluid intake or blood glucose. Circulating insulin and glucagon were reduced by STZ treatment, with a corresponding decline in pancreatic insulin content, which were not altered by artemether. The detrimental changes to pancreatic islet morphology induced by STZ were also evident in artemether-treated mice. Tracing of α-cell lineage, through co-staining for glucagon and yellow fluorescent protein (YFP), revealed a significant decrease of the proportion of glucagon+YFP- cells in STZ-diabetic mice, which was reversed by artemether. However, artemether had no effect on transdifferentiation of α-cells into ß-cells and failed to augment the number of bi-hormonal, insulin+glucagon+, islet cells. CONCLUSIONS: Our observations confirm that artemisinin derivatives do not impart meaningful benefits on islet cell lineage transition events or pancreatic islet morphology.


Subject(s)
Diabetes Mellitus, Experimental , Insulin-Secreting Cells , Islets of Langerhans , Mice , Animals , Insulin/metabolism , Glucagon/metabolism , Glucagon/pharmacology , Cell Transdifferentiation , Diabetes Mellitus, Experimental/metabolism , Artemether/pharmacology , Artemether/metabolism , Artemether/therapeutic use , Blood Glucose , Streptozocin/pharmacology
19.
Nat Commun ; 13(1): 6353, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289202

ABSTRACT

Artemisinin partial resistance may facilitate selection of Plasmodium falciparum resistant to combination therapy partner drugs. We evaluated 99 P. falciparum isolates collected in 2021 from northern Uganda, where resistance-associated PfK13 C469Y and A675V mutations have emerged, and eastern Uganda, where these mutations are uncommon. With the ex vivo ring survival assay, isolates with the 469Y mutation (median survival 7.3% for mutant, 2.5% mixed, and 1.4% wild type) and/or mutations in Pfcoronin or falcipain-2a, had significantly greater survival; all isolates with survival >5% had mutations in at least one of these proteins. With ex vivo growth inhibition assays, susceptibility to lumefantrine (median IC50 14.6 vs. 6.9 nM, p < 0.0001) and dihydroartemisinin (2.3 vs. 1.5 nM, p = 0.003) was decreased in northern vs. eastern Uganda; 14/49 northern vs. 0/38 eastern isolates had lumefantrine IC50 > 20 nM (p = 0.0002). Targeted sequencing of 819 isolates from 2015-21 identified multiple polymorphisms associated with altered drug susceptibility, notably PfK13 469Y with decreased susceptibility to lumefantrine (p = 6 × 10-8) and PfCRT mutations with chloroquine resistance (p = 1 × 10-20). Our results raise concern regarding activity of artemether-lumefantrine, the first-line antimalarial in Uganda.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Humans , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Antimalarials/pharmacology , Antimalarials/therapeutic use , Lumefantrine/pharmacology , Lumefantrine/therapeutic use , Artemether, Lumefantrine Drug Combination/pharmacology , Artemether, Lumefantrine Drug Combination/therapeutic use , Uganda , Malaria, Falciparum/drug therapy , Drug Resistance/genetics , Artemether/pharmacology , Artemether/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Chloroquine/pharmacology , Drug Combinations , Protozoan Proteins/metabolism
20.
Lancet Microbe ; 3(9): e701-e710, 2022 09.
Article in English | MEDLINE | ID: mdl-35931099

ABSTRACT

BACKGROUND: Artemisinin-resistant genotypes of Plasmodium falciparum have now emerged a minimum of six times on three continents despite recommendations that all artemisinins be deployed as artemisinin combination therapies (ACTs). Widespread resistance to the non-artemisinin partner drugs in ACTs has the potential to limit the clinical and resistance benefits provided by combination therapy. We aimed to model and evaluate the long-term effects of high levels of partner-drug resistance on the early emergence of artemisinin-resistant genotypes. METHODS: Using a consensus modelling approach, we used three individual-based mathematical models of Plasmodium falciparum transmission to evaluate the effects of pre-existing partner-drug resistance and ACT deployment on the evolution of artemisinin resistance. Each model simulates 100 000 individuals in a particular transmission setting (malaria prevalence of 1%, 5%, 10%, or 20%) with a daily time step that updates individuals' infection status, treatment status, immunity, genotype-specific parasite densities, and clinical state. We modelled varying access to antimalarial drugs if febrile (coverage of 20%, 40%, or 60%) with one primary ACT used as first-line therapy: dihydroartemisinin-piperaquine (DHA-PPQ), artesunate-amodiaquine (ASAQ), or artemether-lumefantrine (AL). The primary outcome was time until 0·25 580Y allele frequency for artemisinin resistance (the establishment time). FINDINGS: Higher frequencies of pre-existing partner-drug resistant genotypes lead to earlier establishment of artemisinin resistance. Across all models, a 10-fold increase in the frequency of partner-drug resistance genotypes on average corresponded to loss of artemisinin efficacy 2-12 years earlier. Most reductions in time to artemisinin resistance establishment were observed after an increase in frequency of the partner-drug resistance genotype from 0·0 to 0·10. INTERPRETATION: Partner-drug resistance in ACTs facilitates the early emergence of artemisinin resistance and is a major public health concern. Higher-grade partner-drug resistance has the largest effect, with piperaquine resistance accelerating the early emergence of artemisinin-resistant alleles the most. Continued investment in molecular surveillance of partner-drug resistant genotypes to guide choice of first-line ACT is paramount. FUNDING: Schmidt Science Fellowship in partnership with the Rhodes Trust; Bill & Melinda Gates Foundation; Wellcome Trust.


Subject(s)
Antimalarials , Malaria, Falciparum , Antimalarials/pharmacology , Artemether/pharmacology , Artemether, Lumefantrine Drug Combination/pharmacology , Consensus , Drug Resistance/genetics , Humans , Malaria, Falciparum/drug therapy , Plasmodium falciparum/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...