Skip to main content
Erschienen in: Malaria Journal 1/2019

Open Access 01.12.2019 | Research

A comprehensive analysis of drug resistance molecular markers and Plasmodium falciparum genetic diversity in two malaria endemic sites in Mali

verfasst von: Seidina A. S. Diakité, Karim Traoré, Ibrahim Sanogo, Taane G. Clark, Susana Campino, Modibo Sangaré, Djeneba Dabitao, Antoine Dara, Drissa S. Konaté, Fousseyni Doucouré, Amadou Cissé, Bourama Keita, Mory Doumbouya, Merepen A. Guindo, Mahamoudou B. Toure, Nafomon Sogoba, Seydou Doumbia, Gordon A. Awandare, Mahamadou Diakité

Erschienen in: Malaria Journal | Ausgabe 1/2019

Abstract

Background

Drug resistance is one of the greatest challenges of malaria control programme in Mali. Recent advances in next-generation sequencing (NGS) technologies provide new and effective ways of tracking drug-resistant malaria parasites in Africa. The diversity and the prevalence of Plasmodium falciparum drug-resistance molecular markers were assessed in Dangassa and Nioro-du-Sahel in Mali, two sites with distinct malaria transmission patterns. Dangassa has an intense seasonal malaria transmission, whereas Nioro-du-Sahel has an unstable and short seasonal malaria transmission.

Methods

Up to 270 dried blood spot samples (214 in Dangassa and 56 in Nioro-du-Sahel) were collected from P. falciparum positive patients in 2016. Samples were analysed on the Agena MassARRAY® iPLEX platform. Specific codons were targeted in Pfcrt, Pfmdr1, Pfdhfr, and Pfdhps, Pfarps10, Pfferredoxin, Pfexonuclease and Pfmdr2 genes. The Sanger’s 101-SNPs-barcode method was used to assess the genetic diversity of P. falciparum and to determine the parasite species.

Results

The Pfcrt_76T chloroquine-resistance genotype was found at a rate of 64.4% in Dangassa and 45.2% in Nioro-du-Sahel (p = 0.025). The Pfdhfr_51I-59R-108N pyrimethamine-resistance genotype was 14.1% and 19.6%, respectively in Dangassa and Nioro-du-Sahel. Mutations in the Pfdhps_S436-A437-K540-A581-613A sulfadoxine-resistance gene was significantly more prevalent in Dangassa as compared to Nioro-du-Sahel (p = 0.035). Up to 17.8% of the isolates from Dangassa vs 7% from Nioro-du-Sahel harboured at least two codon substitutions in this haplotype. The amodiaquine-resistance Pfmdr1_N86Y mutation was identified in only three samples (two in Dangassa and one in Nioro-du-Sahel). The lumefantrine-reduced susceptibility Pfmdr1_Y184F mutation was found in 39.9% and 48.2% of samples in Dangassa and Nioro-du-Sahel, respectively. One piperaquine-resistance Exo_E415G mutation was found in Dangassa, while no artemisinin resistance genetic-background were identified. A high P. falciparum diversity was observed, but no clear genetic aggregation was found at either study sites. Higher multiplicity of infection was observed in Dangassa with both COIL (p = 0.04) and Real McCOIL (p = 0.02) methods relative to Nioro-du-Sahel.

Conclusions

This study reveals high prevalence of chloroquine and pyrimethamine-resistance markers as well as high codon substitution rate in the sulfadoxine-resistance gene. High genetic diversity of P. falciparum was observed. These observations suggest that the use of artemisinins is relevant in both Dangassa and Nioro-du-Sahel.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12936-019-2986-5.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
USTTB
University of Sciences, Technics and Technologies of Bamako
WACCBIP
West African Centre of Cell Biology of Infectious Pathogens
NGS
next-generation sequencing
PfCRT
Plasmodium falciparum chloroquine resistance transporter
PfDHFR
Plasmodium falciparum dihydrofolate-reductase
PfDHPS
Plasmodium falciparum dihydropteroate-synthetase
PfARPS10
Plasmodium falciparum Apicoplast Ribosomal Protein S10
PfFd
Plasmodium falciparum Ferredoxin
PfExo
Plasmodium falciparum exonuclease
PfMDR
Plamodium falciparum Multidrug resistance
COIL
complexity of infection likelihood
McCOIL
Method for the concurrent estimation of complexity of infection likelihood
NMCP
National Malaria Control Programme
ACT
artemisinin-based combination therapy
Artemisinins
artemisinin and its derivative drugs
WHO
World Health Organization
SNP
single nucleotid polymorphism
GWAS
Genome-Wide Association Study
PGB
parasite genetic background
LLINs
long lasting insecticide-impregnated nets
IPTp
intermittent preventive treatment of pregnant women
SP
sulfadoxine + pyrimethamine
RDT
rapid diagnostic test
HRP2
histidine rich-protein 2
pLDH
Plasmodium lactate dehydrogenage
i.m
intra muscular
WAG
whole genome amplification

Background

Despite numerous advances in malaria control strategies, the disease still kills countless children worldwide, mainly in sub-Saharan African countries. The World Health Organization (WHO) reported no significant progress in reducing global malaria burden during the period from 2015 to 2017 [1]. In Mali, as well as in the rest of the world, resistance to anti-malarial drugs is one of the greatest challenges of the National Malaria Control Programme (NMCP) [2, 3]. High prevalence of resistance to chloroquine and sulfadoxine-pyrimethamine (SP) led the Malian NMCP to switch to artemisinin- based combination therapy (ACT) in the 2000s, as recommended by the WHO [4]. Artemisinin and its derivative drugs (Artemisinins) are still very effective in Africa [5, 6]. Resistance to these drugs have been reported in East-Asia [7, 8]. One of the rationales of ACT use is to maintain long-term efficacy of artemisinins to P. falciparum [4]. To achieve this objective, the choice of the artemisinin partner drugs is important and should be made based on their effectiveness on circulating local parasite isolates. Consequently, it appears important to monitor the emergence and spread of the anti-malarial drug resistance in different geographical and endemic areas in order to inform the choice of the anti-malarial molecules to be associated with artemisinins in ACT [9, 10]. Also, assessing the genetic diversity of P. falciparum populations in different regions may allow to tract malaria parasites circulating across different geographic areas.
Molecular markers of drug-resistance are very useful in identifying drug-resistant P. falciparum. They are used in epidemiological surveillance of drug-resistance including their emergence and spread monitoring. Molecular markers have been described for many of the common anti-malarial drug and are constituted of either single nucleotide substitution (SNP) or concatenated SNP in genes involved in parasite interaction with drugs. Evidence of association between several alleles in P. falciparum multi-drug resistance (Pfmdr) gene and chloroquine-resistance was reported in 1990 [11]. However, the K76T mutation in P. falciparum chloroquine resistance transporter gene (Pfcrt_K76T) mostly associated with the Pfcrt_72C-73V-74I-75E-76T haplotype in Africa is known as the strongest chloroquine-resistance marker in P. falciparum [1215]. Resistance to pyrimethamine has also been associated with numbers of mutations combination in the P. falciparum dihydrofolate-reductase (Pfdhfr) gene, such as the Pfdhfr_51I-59R-164L [16] and Pfdhfr_51R-59N-108I genotypes [17, 18]. Also various combinations of substitutions at the codons 436, 437, 540, 581 and 613 of the P. falciparum dihydropteroate-synthetase (Pfdhps) gene may confer resistance to sulfadoxine [19]. The greater the number of substitution accumulation is in the gene, the more the parasite gets resistant to the sulfadoxine. The Pfdhps_437G-540E genotype was associated with substantially decreased susceptibility to sulfadoxine. In vitro experiments have shown that N86Y mutation in the P. falciparum multidrug resistance-1 (Pfmdr1) gene (Pfmdr1_86Y) increases the fifty percent inhibitory concentrations (IC50) of chloroquine and amodiaquine [20, 21].
Mutations in multidrug resistance protein (pfmdr1) have been associated with various parasite response to mefloquine and lumefantrine [22]. Reduced susceptibility to lumefantrine has been associated with the Pfmdr1_Y184F mutation [2325]. In a recent genome-wide association study (GWAS), the E415G mutation, a SNP in P. falciparum exonuclease gene (Pfexo_E415G)) was associated with ex vivo piperaquine IC50 of parasite isolates from Cambodia [26]. The co-inheritance of several mutations in P. falciparum Apicoplast Ribosomal Protein S10 (Pfarps10) gene [Pfarps10_127M-128Y/H], the P. falciparum ferredoxin (PfFd) gene [Pffd_ 193Y], the PfCRT gene: [Pfcrt_326S-356T], and the PfMDR2 [Pfmdr2_T484I] were shown to constitute a genetic background that allows for the emergence of mutations in the P. falciparum kelch 13 gene [27], a known artemisinin resistance locus [2830]. This haplotype is referred to as artemisinin-resistant parasite genetic background (Art-resistant PGB).
All thought previous studies has provided insights into some anti-malarial drug resistance markers in the south of Mali [31, 32], the risk of P. falciparum infection is variable from south (with intense and long transmission) to north (with unstable and short transmission) in Mali. This variability is related to environmental and climatic conditions. The level of transmission may also impact the malaria parasite genetic diversity and the spread of resistance to anti-malarial drugs. In fact, the higher the malaria transmission is, the more diverse the parasite population is, the more likely the parasite gets resistant to anti-malarial drugs.
In this study, both the genetic diversity of P. falciparum and the distribution of drug resistance markers were assessed in two distinct eco-climatic areas in Mali.

Methods

Study sites

The epidemiology, entomology and the impact of malaria vary widely across the Malian territory [33, 34]. The study was carried out in Dangassa (Lat = 12.150,925, Long = − 8.206974) and Nioro-du-Sahel (Lat = 15.224674, Long = − 9.583888). Dangassa is located in south savannah grassland of Mali along the right side of Niger River and has year-round access to water (Niger River). Malaria transmission is seasonal and mainly occurs from June to December. However, dry season transmission may occurs because of the Niger River. Indeed, mosquito breeding occurs primarily in microhabitats, such as the footprints of cattle and the receding river in the dry season, which leaves natural pools as well as man-made pools from river gold mining. Nioro-du-Sahel is located in Sahelian region of Mali near the Islamic Republic of Mauritania. Malaria transmission is unstable and occurs from July to September. Anopheles gambiae breeding sites are strictly rain-dependent. Anopheles gambiae (about 95%) and Anopheles arabiensis (about 5%) are the predominant malaria vectors [35] in the two sites. Population migration occurs in the two localities mainly for gold mining for Dangassa and for herding or for trading for Nioro-du-Sahel. In both localities, malaria control strategies rely on the use of long-lasting insecticide-impregnated nets (LLINs), ACT for treatment and sulfadoxine-pyrimethamine (SP) for intermittent preventive treatment of pregnant women (IPTp). In addition, the population may have access to other malaria drugs in the local market in the two localities.

Study design and sample collection

The study has been set up by the research team in collaboration with the local health staff for the entire malaria transmission season (June to December 2016). A medical team was installed at the health centre of the village. Population of all age group was encouraged to attend the local health centre if febrile, where they were examined by a study physician. Malaria cases were diagnosed by a rapid diagnostic test (RDT) [CareStart™: Malaria HRP2/pLDH(Pf/PAN) Combo] and/or microscopy. A four (4) ml venous blood was obtained from each TDR positive patient. A dried blood spot (DBS) was made by dropping approximatively 50 μl of the venous blood on a filter paper. The DNA was extract from the DBS and used for P. falciparum genomic and host genetic studies. Confirmed malaria cases were classified as mild or severe according to the World Health Organization (WHO) definitions [36], and treated according to the Malian NMCP guidelines (oral ACT for uncomplicated malaria, intramuscular (i.m) administration of artemether and/or quinine for severe malaria).

DNA extraction, sample processing, and genotyping

The dried blood spots on filter paper were sent to the Wellcome Sanger Institute for processing. DNA extraction was carried out using the Qiagen DNA Investigator Kit (No. 56504, Qiagen, Crawley, UK) for high-throughput robotic processing. DNA was eluted in 100 μl TE buffer and stored at − 20 °C for later use. Extracted DNA underwent whole genome amplification (WGA) by primer-extension pre-amplification [37] or selective WGA [38] prior to genotyping. Genotyping was performed according to manufacturer’s instruction on the Agena MassARRAY® iPLEX platform (Agena Bioscience, Hamburg, Germany). This system is able to accurately genotype large numbers of samples for multiple SNPs simultaneously. It previously been used in P. falciparum for sequencing validation of novel SNPs [39]. All genotypes were called from background adjusted peak intensities, which were normalized and called by batch. In brief, batches underwent calling using a heuristic algorithm which identifies intensity ranges for each SNP in single infection samples, and called mixed base loci (“heterozygous”) based on those range thresholds, adjusting for background intensity. Batches were plate based and contained between 96 and 384 samples. These are necessary to generate per assay ranges of intensities to calculate background intensity levels.

Parasite diversity and multiple infection assessment

The malaria parasite diversity and multiplicity of infection were assessed using the Sanger’s P. falciparum barcodes formed by concatenated genotypes at 101 SNPs across P. falciparum genome. These SNPs are all bi-allelic, at low to medium frequency in global parasite populations, and were chosen for their usefulness in analyses of relationship between P. falciparum parasites. They have been used previously to estimate levels of relationship and importation [40] as well as complexity of infection. Multiplicity of infection estimates were produced by both the programmes COIL [41] using default parameters and The Real McCOIL [42] using the bi-allelic SNP barcodes.

Drug resistance markers assessment

Specific mutations in P. falciparum genome were screened using the Agena MassARRAY® iPLEX platform as above. The following codons in the P. falciparum genome were screened for substitution: K76, N326, I356 in the Pfcrt gene, the N86, Y184, D1246 in the Pfmdr1 gene, the N51, C59, S108, I164 in the Pfdhfr gene, the S436, A337, K540, A581, A613 in Pfdhps V127, D128 in the Pfarps10 gene, the D193 in Pf-ferredoxin gene, the E415 in Pf-exonuclease gene, and T484 in Pfmdr2. Single isolate mutations or combination of mutations in these codons are associated with resistance to anti-malarial drugs as specified in the background section.

Statistical analysis

This was an inclusive study. All the confirmed malaria case detected during the study period in the two sites were enrolled. A Chi square test was used to compare the prevalence of molecular markers in the two study sites. When the effective ≥ 30, the Z-test was considered. The significant P value was set as equal or inferior to 0.05.
For the Real McCOIL determination, the categorical method was used with the following parameters, maxCOI = 25, threshold_ind = 20, threshold_site = 20, totalrun = 10,000, burnin = 1000, M0 = 15, e1 = 0.05, e2 = 0.05, err_method = 1. Principal component analysis was performed to assess and compare the parasite diversity between the two study sites.

Results

In total, 270 P. falciparum samples (214 from Dangassa and 56 from Nioro-du-Sahel) were analysed. The analysis of anti-malarial drug genes showed very high rates of mutations. The chloroquine-resistance Pfcrt-76T genotype was significantly more prevalent in Dangassa [64.4% (45.9% 76T + 18.4% 76K/76T)] compared to Nioro-du-Sahel [45.2% (37.7% 76T + 7.5% 76K/76T)] (p = 0.025) (Fig. 1). This study revealed a very high but similar rate of codon-substitution in the pyrimethamine-resistance Pfdhfr gene at positions 51, 59, 108 and 164 in both Dangassa and Nioro-du-Sahel (Fig. 2) (p = 0.2). Respectively, 88.3% and 80% of the isolates from Dangassa and Nioro showed at least one substitution at these codons. However the prevalence of the particular pyrimethamine-resistance Pfdhfr_51I-19R-108N genotype reached 14.1% and 19.6% in Dangassa and Nioro-du-Sahel, respectively. Higher rate of codon substitution was observed in the sulfadoxine-resistance Pfdhps gene at positions 436, 437, 540, 581 and 613. These substitutions were more prevalent in Dangassa as compared to Nioro-du-Sahel (p value = 0.035). Up to 17.8% of the isolates from Dangassa against 7% for the isolates from Nioro-du-Sahel accumulated at least two (2) codon-substitutions were detected at these codons (Fig. 3).
The particular sulfadoxine-resistance haplotype Pfdhps_437G-540E was found in four isolates from Dangassa out of which only one exhibits the pyrimethamine-resistance Pfdhfr_51I-19R-108N genotype (the quintuple Pfdhps_437G-540E/Pfdhfr_51I-19R-108N substitution).
The amodiaquine-resistance Pfmdr1-N86Y mutation was identified in 14.9% (6% Pfmdr1-Y86 + 8.8% Pfmdr1-N86/Y86) and 14.2% (8.9% Pfmdr1-Y86 + 8.3% Pfmdr1-N86/Y86) of the samples from Dangassa and Nioro-du-Sahel, respectively.
More importantly, the lumefantrine-reduced susceptibility Pfmdr1_184F mutation was found in 39.9% and 48.2% of samples from Dangassa and Nioro-du-Sahel, respectively. Only two (2) isolates from Dangassa exhibited the piperaquine-resistance Exo-E415G mutation. Such mutation was not found in Nioro. No artemisinins-resistance PGB was detected in the genotyped isolates. However, isolated codon substitutions constitutive of the artemisinin resistance PGB were found. Further information regarding the codon variation in studied genes is available in Additional file 1: Table S1.
A principal component analysis of genotyping data from the Sanger 101 SNPs barcode showed very high P. falciparum genomic diversity in both Dangassa and Nioro-du-Sahel. However, no aggregation of specific P. falciparum genotype was found either in Dangassa or in Nioro (Fig. 4). Higher proportion of multiplicity of infection was observed in Dangassa using either COIL (p = 0.04) or Real McCOIL (p = 0.02) method based on the Sanger 101 SNPs barcode (Table 1).
Table 1
Multiplicity of infection in Dangassa and Nioro-du-Sahel patients, either with the MOI-COIL or the MOI-McCOIL
Index of multiplicity of infection
Dangassa (n = 214)
Nioro-du-sahel (n = 56)
Difference p
MOI-COIL
 1
139
65.3
44
83.0
 
 2
73
34.3
9
17.0
 
 3
1
0.5
0
0.0
0.043
MOI-McCOIL
 1
151
70.9
47
88.7
 
 2
61
28.6
6
11.3
 
 3
1
0.5
0
0.0
0.029

Discussion

To protect the artemisinin molecules from resistance emergence in Africa and delay the spread of this resistance in the world, the WHO recommended ACT for uncomplicated malaria treatment in 2001 [4]. Such a strategy requires monitoring P. falciparum resistance to anti-malarial drugs in different geographical areas and adapting the appropriate drug combination. Moreover, the monitoring of the malaria parasite genetic variation will help to predict the dynamic of spread of the drug resistance through varying geographical areas. To assess molecular markers of anti-malarial drugs resistance and the genetic diversity in P. falciparum circulating isolates through the Malian territory, a passive case detection study was carried out in Dangassa and Nioro, two distinct epidemiological and eco-climatic zones in Mali. The revealed a high prevalence of the chloroquine resistance Pfcrt_76T in both localities. The Pfcrt_76T mutation was found in 64.3% in Dangassa and 42.5% in Nioro-du-Sahel. This is the first data report of anti-malarial drug resistance marker in Nioro, but a previous study has reported the prevalence of the chloroquine-resistance Pfcrt_76T mutation was of 85% in 2002 and 64.5% in 2003 in Kollé, a village sharing the same geographical area with Dangassa [31]. These observations suggest no decrease in the prevalence of chloroquine resistance marker in this region from 2002 to 2016 despite the official removal of chloroquine by the Malian NMCP since the years 2000s. Several studies in countries where chloroquine was removed reported substantial decrease in the prevalence of the chloroquine resistance marker suggesting a possible future reintroduction of this low-cost drug [4346].
The data from this study revealed a very low prevalence of amodiaquine resistance-associated mutation Pfmdr1_86Y mutant parasite (two (2/214) in Dangassa and one (1/56) in Nioro-du-Sahel). The Pfmdr1_86Y mutation has been associated with amodiaquine resistance in Burkina Faso [47]. The Pfmdr_86Y mutant isolates was reported at the prevalence of 49.3% in Kolle, in 2002; however, the authors could not find association between this mutation and amodiaquine resistance in an in vivo assay [31]. Amodiaquine is one of most used anti-malarial drugs in Mali; it is associated with artemisinin in ACT and is also used in association with the SP in seasonal malaria control strategy (SMC) in children under five (5) years old in Mali.
Surprisingly, a high prevalence of Pfmdr1_Y184F, the lumefantrine-reduced susceptibility mutation was found in both Dangassa (39.9%) and Nioro-du-Sahel (48.2%). A higher prevalence of this mutation (53.8%) was reported in Senegal in 2013–2014 [48]. This finding was intriguing since lumefantrine has never been made available as a monotherapy in Mali. Lumefantrine has always been administrated in combination with artemether. Two possible scenarios may explain this finding. First, P. falciparum is less susceptible to the lumefantrine. This could impede on the efficacy of artemisinin-based strategy since artemether–lumefantrine is the most used ACT in Mali. Second, the Pfmdr1_Y184F mutation is not a good lumefantrine-resistance marker in Mali. Although several studies have reported an association between Pfmdr1_Y184F mutation and ex vivo reduced susceptibility to lumefantrine [49, 50], another in vivo studies could not establish this association [51]. Further in vitro investigations are needed to tease this observation apart.
The codon substitutions were assessed in the Pfdhfr and Pfdhp genes to identify SP resistant parasites, characterized by the inheritance of the Pfdhfr_51R-59N-108I/Pfdhps_437G-540E quintuple mutation. A high level of codon substitution was observed in these genes. The pyrimethamine-resistance Pfdhfr_51R-59N-108I genotype was found in 14.1% in Dangassa and 19.6% in Nioro-du-Sahel suggesting a decrease in the prevalence of this haplotype in the south of Mali as compared to the prevalence’s of 31.5% and 42.9% reported respectively 2002 and 2003 in Kolle [31]. The sulfadoxine resistance haplotype Pfdhps_437G-540E was found in only four isolates in Dangassa. Only one isolate presented the Pfdhfr_51R-59N-108I/Pfdhps_437G-540E quintuple substitution in Dangassa. SP is widely used in Mali for pregnant woman malaria chemoprophylaxis and seasonal malaria chemoprophylaxis in children under 5 years old. The data from this study suggest that the use of SP is still relevant in Mali, but previous reports suggest that such a malaria control strategy appears to increase the prevalence of SP-resistant parasite in Mali [32]. A very high rate of codon substitution was observed in these two genes (Pfdhfr and Pfdhps) in both Dangassa and Nioro-du-Sahel (Additional file 2).
No Pfarps10_127M-128Y/H-Pffd_193Y-Pfcrt_326S-356T-Pfmdr2_484I artemisinin resistance genetic background genotype was found during this study. Several mutations in P. falciparum kelch 13 protein propeller domain have been associated to resistance to artemisinins in Asia [2830]. A genome wide association study revealed that these mutations always arise on the above specific genetic background [27]. Even though, no artemisinin resistance has been reported yet in Mali, [5, 52], a close monitoring for artemisinin resistance is essential. Further analysis of individual mutation constitutive of artemisinin resistance genetic background genotype may provide appropriate tool to predict artemisinin resistance emergence in Africa, where mutation in the Pfkelch13 propeller domain were not associated with artemisinin resistance [53].
P. falciparum genetic diversity is correlated with the level of transmission in different continental regions [5456], and is influenced within regions by factors such as altitude, vector availability, urbanization, and malaria control strategies [57, 58]. In this study, it was hypothesized that the eco-climatic difference determines the malaria transmission variability between the two sites, and would impact the fitness of malaria parasites. Therefore, specific genetic variant of parasites could be selected in each site. Using the Sanger’s P. falciparum barcodes formed by concatenated genotypes at 101 SNPs across P. falciparum genome, the genetic diversity in P. falciparum circulating isolates was assessed in the study localities. These genotyping data showed a very high genetic diversity in P. falciparum isolates in both Dangassa and Nioro-du-Sahel supporting previous finding in Africa, even though those studies explored the msp1, msp2 or glurp genes size polymorphisms [59, 60]. No aggregation of specific P. falciparum genotype was observed at either study site. This finding may be due to the fact that the two sites are not geographically isolated. A higher complexity of P. falciparum infection was found in Dangassa, compared to Nioro-du-Sahel. These findings reflect the difference of transmission intensity between the two study sites. The complexity of infection is known to be associated with the intensity of malaria transmission [61, 62].

Conclusions

This study revealed high diversity in P. falciparum circulating isolates in Mali. However no genetic population structure was found in either low or high transmission areas. No artemisinin resistance genetic background was found, but high prevalence of the lumefantrine resistance marker (Pfmdr1_Y184F genotype), the most used associated molecule to artemisinin in Mali, was detected. Further investigations are required to explore the lumefantrine resistance in Mali.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12936-019-2986-5.

Acknowledgements

We thank the parents, guardians and children who participated into this study, and the technical, clinical and nursing staff for assistance. We are grateful to many colleagues at MRTC for providing critical reviews of the manuscripts which helped improve it. This publication uses data from the MalariaGEN SpotMalaria Project as described online https://​www.​malariagen.​net/​projects/​spotmalaria pending citeable publication; the project is coordinated by the MalariaGEN Resource Centre with funding from Wellcome (206194, 090770). The authors would like to thank the staff of Wellcome Sanger Institute Sample Management, Genotyping, Sequencing and Informatics teams for their contribution”.
Before starting this study, we obtained a community consent from traditional and customary chiefs prior to the study. The study was approved by the ethics committee of the faculty of medicine and Pharmacy of the University of Sciences, Technics and Technologies of Bamako (USTTB), Mali. Written informed consent was obtained from a parent or guardian of each enrolled child.
All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat WHO. World malaria report 2018. Geneva: World Health Organization; 2018. WHO. World malaria report 2018. Geneva: World Health Organization; 2018.
2.
Zurück zum Zitat Bloland PB. Drug resistance in malaria. Geneva: World Health Organization, WHO/CDS/CSR/DRS; 2001. Bloland PB. Drug resistance in malaria. Geneva: World Health Organization, WHO/CDS/CSR/DRS; 2001.
4.
Zurück zum Zitat WHO. Antimalarial drug combination therapy. Geneva: World Health Organization; 2001. WHO. Antimalarial drug combination therapy. Geneva: World Health Organization; 2001.
5.
Zurück zum Zitat Lopera-Mesa TM, Doumbia S, Chiang S, Zeituni AE, Konate DS, Doumbouya M, et al. Plasmodium falciparum clearance rates in response to artesunate in Malian children with malaria: effect of acquired immunity. J Infect Dis. 2013;207:1655–63.PubMedPubMedCentralCrossRef Lopera-Mesa TM, Doumbia S, Chiang S, Zeituni AE, Konate DS, Doumbouya M, et al. Plasmodium falciparum clearance rates in response to artesunate in Malian children with malaria: effect of acquired immunity. J Infect Dis. 2013;207:1655–63.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Abdulla S, Ashley EA, Bassat Q, Bethell D, Bjorkman A, Borrmann S, et al. Baseline data of parasite clearance in patients with falciparum malaria treated with an artemisinin derivative: an individual patient data meta-analysis. Malar J. 2015;14:359.PubMedCrossRef Abdulla S, Ashley EA, Bassat Q, Bethell D, Bjorkman A, Borrmann S, et al. Baseline data of parasite clearance in patients with falciparum malaria treated with an artemisinin derivative: an individual patient data meta-analysis. Malar J. 2015;14:359.PubMedCrossRef
7.
Zurück zum Zitat Dondorp AM, Fairhurst RM, Slutsker L, Macarthur JR, Breman JG, Guerin PJ, et al. The threat of artemisinin-resistant malaria. N Engl J Med. 2011;365:1073–5.PubMedPubMedCentralCrossRef Dondorp AM, Fairhurst RM, Slutsker L, Macarthur JR, Breman JG, Guerin PJ, et al. The threat of artemisinin-resistant malaria. N Engl J Med. 2011;365:1073–5.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat WHO. Global plan for artemisinin resistance containment (GPARC). Geneva: World Health Organization; 2012. WHO. Global plan for artemisinin resistance containment (GPARC). Geneva: World Health Organization; 2012.
10.
Zurück zum Zitat Hastings IM. Molecular markers as indicators of antimalarial drug failure rates. Trop Med Int Health. 2007;12:1298–301.PubMedCrossRef Hastings IM. Molecular markers as indicators of antimalarial drug failure rates. Trop Med Int Health. 2007;12:1298–301.PubMedCrossRef
11.
Zurück zum Zitat Foote SJ, Kyle DE, Martin RK, Oduola AM, Forsyth K, Kemp DJ, et al. Several alleles of the multidrug-resistance gene are closely linked to chloroquine resistance in Plasmodium falciparum. Nature. 1990;345:255–8.PubMedCrossRef Foote SJ, Kyle DE, Martin RK, Oduola AM, Forsyth K, Kemp DJ, et al. Several alleles of the multidrug-resistance gene are closely linked to chloroquine resistance in Plasmodium falciparum. Nature. 1990;345:255–8.PubMedCrossRef
12.
Zurück zum Zitat Fidock DA, Nomura T, Talley AK, Cooper RA, Dzekunov SM, Ferdig MT, et al. Mutations in the P falciparum digestive vacuole transmembrane protein PfCRT and evidence for their role in chloroquine resistance. Mol Cell. 2000;6:861–71.PubMedPubMedCentralCrossRef Fidock DA, Nomura T, Talley AK, Cooper RA, Dzekunov SM, Ferdig MT, et al. Mutations in the P falciparum digestive vacuole transmembrane protein PfCRT and evidence for their role in chloroquine resistance. Mol Cell. 2000;6:861–71.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Baro NK, Callaghan PS, Roepe PD. Function of resistance conferring Plasmodium falciparum chloroquine resistance transporter isoforms. Biochemistry. 2013;52:4242–9.PubMedCrossRef Baro NK, Callaghan PS, Roepe PD. Function of resistance conferring Plasmodium falciparum chloroquine resistance transporter isoforms. Biochemistry. 2013;52:4242–9.PubMedCrossRef
15.
Zurück zum Zitat Durand R, Jafari S, Vauzelle J, Delabre JF, Jesic Z, Le Bras J. Analysis of pfcrt point mutations and chloroquine susceptibility in isolates of Plasmodium falciparum. Mol Biochem Parasitol. 2001;114:95–102.PubMedCrossRef Durand R, Jafari S, Vauzelle J, Delabre JF, Jesic Z, Le Bras J. Analysis of pfcrt point mutations and chloroquine susceptibility in isolates of Plasmodium falciparum. Mol Biochem Parasitol. 2001;114:95–102.PubMedCrossRef
16.
Zurück zum Zitat Chusacultanachai S, Thiensathit P, Tarnchompoo B, Sirawaraporn W, Yuthavong Y. Novel antifolate resistant mutations of Plasmodium falciparum dihydrofolate reductase selected in Escherichia coli. Mol Biochem Parasitol. 2002;120:61–72.PubMedCrossRef Chusacultanachai S, Thiensathit P, Tarnchompoo B, Sirawaraporn W, Yuthavong Y. Novel antifolate resistant mutations of Plasmodium falciparum dihydrofolate reductase selected in Escherichia coli. Mol Biochem Parasitol. 2002;120:61–72.PubMedCrossRef
17.
Zurück zum Zitat Gregson A, Plowe CV. Mechanisms of resistance of malaria parasites to antifolates. Pharmacol Rev. 2005;57:117–45.PubMedCrossRef Gregson A, Plowe CV. Mechanisms of resistance of malaria parasites to antifolates. Pharmacol Rev. 2005;57:117–45.PubMedCrossRef
18.
Zurück zum Zitat Peterson DS, Walliker D, Wellems TE. Evidence that a point mutation in dihydrofolate reductase-thymidylate synthase confers resistance to pyrimethamine in falciparum malaria. Proc Natl Acad Sci USA. 1988;85:9114–8.PubMedCrossRefPubMedCentral Peterson DS, Walliker D, Wellems TE. Evidence that a point mutation in dihydrofolate reductase-thymidylate synthase confers resistance to pyrimethamine in falciparum malaria. Proc Natl Acad Sci USA. 1988;85:9114–8.PubMedCrossRefPubMedCentral
19.
Zurück zum Zitat Wang P, Lee CS, Bayoumi R, Djimde A, Doumbo O, Swedberg G, et al. Resistance to antifolates in Plasmodium falciparum monitored by sequence analysis of dihydropteroate synthetase and dihydrofolate reductase alleles in a large number of field samples of diverse origins. Mol Biochem Parasitol. 1997;89:161–77.PubMedCrossRef Wang P, Lee CS, Bayoumi R, Djimde A, Doumbo O, Swedberg G, et al. Resistance to antifolates in Plasmodium falciparum monitored by sequence analysis of dihydropteroate synthetase and dihydrofolate reductase alleles in a large number of field samples of diverse origins. Mol Biochem Parasitol. 1997;89:161–77.PubMedCrossRef
20.
Zurück zum Zitat Nsobya SL, Kiggundu M, Nanyunja S, Joloba M, Greenhouse B, Rosenthal PJ. In vitro sensitivities of Plasmodium falciparum to different antimalarial drugs in Uganda. Antimicrob Agents Chemother. 2010;54:1200–6.PubMedPubMedCentralCrossRef Nsobya SL, Kiggundu M, Nanyunja S, Joloba M, Greenhouse B, Rosenthal PJ. In vitro sensitivities of Plasmodium falciparum to different antimalarial drugs in Uganda. Antimicrob Agents Chemother. 2010;54:1200–6.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Echeverry DF, Holmgren G, Murillo C, Higuita JC, Bjorkman A, Gil JP, et al. Polymorphisms in the pfcrt and pfmdr1 genes of Plasmodium falciparum and in vitro susceptibility to amodiaquine and desethylamodiaquine. Am J Trop Med Hyg. 2007;77:1034–8.PubMedCrossRef Echeverry DF, Holmgren G, Murillo C, Higuita JC, Bjorkman A, Gil JP, et al. Polymorphisms in the pfcrt and pfmdr1 genes of Plasmodium falciparum and in vitro susceptibility to amodiaquine and desethylamodiaquine. Am J Trop Med Hyg. 2007;77:1034–8.PubMedCrossRef
22.
Zurück zum Zitat Sidhu AB, Valderramos SG, Fidock DA. Pfmdr1 mutations contribute to quinine resistance and enhance mefloquine and artemisinin sensitivity in Plasmodium falciparum. Mol Microbiol. 2005;57:913–26.PubMedCrossRef Sidhu AB, Valderramos SG, Fidock DA. Pfmdr1 mutations contribute to quinine resistance and enhance mefloquine and artemisinin sensitivity in Plasmodium falciparum. Mol Microbiol. 2005;57:913–26.PubMedCrossRef
23.
Zurück zum Zitat Malmberg M, Ferreira PE, Tarning J, Ursing J, Ngasala B, Bjorkman A, et al. Plasmodium falciparum drug resistance phenotype as assessed by patient antimalarial drug levels and its association with pfmdr1 polymorphisms. J Infect Dis. 2013;207:842–7.PubMedCrossRef Malmberg M, Ferreira PE, Tarning J, Ursing J, Ngasala B, Bjorkman A, et al. Plasmodium falciparum drug resistance phenotype as assessed by patient antimalarial drug levels and its association with pfmdr1 polymorphisms. J Infect Dis. 2013;207:842–7.PubMedCrossRef
24.
Zurück zum Zitat Taylor AR, Flegg JA, Holmes CC, Guerin PJ, Sibley CH, Conrad MD, et al. Artemether–lumefantrine and dihydroartemisinin–piperaquine exert inverse selective pressure on Plasmodium falciparum drug sensitivity-associated haplotypes in Uganda. Open Forum Infect Dis. 2017;4:229.CrossRef Taylor AR, Flegg JA, Holmes CC, Guerin PJ, Sibley CH, Conrad MD, et al. Artemether–lumefantrine and dihydroartemisinin–piperaquine exert inverse selective pressure on Plasmodium falciparum drug sensitivity-associated haplotypes in Uganda. Open Forum Infect Dis. 2017;4:229.CrossRef
25.
Zurück zum Zitat Sisowath C, Ferreira PE, Bustamante LY, Dahlstrom S, Martensson A, Bjorkman A, et al. The role of pfmdr1 in Plasmodium falciparum tolerance to artemether–lumefantrine in Africa. Trop Med Int Health. 2007;12:736–42.PubMedCrossRef Sisowath C, Ferreira PE, Bustamante LY, Dahlstrom S, Martensson A, Bjorkman A, et al. The role of pfmdr1 in Plasmodium falciparum tolerance to artemether–lumefantrine in Africa. Trop Med Int Health. 2007;12:736–42.PubMedCrossRef
26.
Zurück zum Zitat Amato R, Lim P, Miotto O, Amaratunga C, Dek D, Pearson RD, et al. Genetic markers associated with dihydroartemisinin–piperaquine failure in Plasmodium falciparum malaria in Cambodia: a genotype-phenotype association study. Lancet Infect Dis. 2017;17:164–73.PubMedCrossRef Amato R, Lim P, Miotto O, Amaratunga C, Dek D, Pearson RD, et al. Genetic markers associated with dihydroartemisinin–piperaquine failure in Plasmodium falciparum malaria in Cambodia: a genotype-phenotype association study. Lancet Infect Dis. 2017;17:164–73.PubMedCrossRef
27.
Zurück zum Zitat Miotto O, Amato R, Ashley EA, MacInnis B, Almagro-Garcia J, Amaratunga C, et al. Genetic architecture of artemisinin-resistant Plasmodium falciparum. Nat Genet. 2015;47:226–34.PubMedPubMedCentralCrossRef Miotto O, Amato R, Ashley EA, MacInnis B, Almagro-Garcia J, Amaratunga C, et al. Genetic architecture of artemisinin-resistant Plasmodium falciparum. Nat Genet. 2015;47:226–34.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, et al. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2014;371:411–23.PubMedPubMedCentralCrossRef Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, et al. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2014;371:411–23.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois AC, Khim N, et al. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature. 2014;505:50–5.PubMedCrossRef Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois AC, Khim N, et al. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature. 2014;505:50–5.PubMedCrossRef
30.
Zurück zum Zitat Tun KM, Imwong M, Lwin KM, Win AA, Hlaing TM, Hlaing T, et al. Spread of artemisinin-resistant Plasmodium falciparum in Myanmar: a cross-sectional survey of the K13 molecular marker. Lancet Infect Dis. 2015;15:415–21.PubMedPubMedCentralCrossRef Tun KM, Imwong M, Lwin KM, Win AA, Hlaing TM, Hlaing T, et al. Spread of artemisinin-resistant Plasmodium falciparum in Myanmar: a cross-sectional survey of the K13 molecular marker. Lancet Infect Dis. 2015;15:415–21.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Tekete M, Djimde AA, Beavogui AH, Maiga H, Sagara I, Fofana B, et al. Efficacy of chloroquine, amodiaquine and sulphadoxine-pyrimethamine for the treatment of uncomplicated falciparum malaria: revisiting molecular markers in an area of emerging AQ and SP resistance in Mali. Malar J. 2009;8:34.PubMedPubMedCentralCrossRef Tekete M, Djimde AA, Beavogui AH, Maiga H, Sagara I, Fofana B, et al. Efficacy of chloroquine, amodiaquine and sulphadoxine-pyrimethamine for the treatment of uncomplicated falciparum malaria: revisiting molecular markers in an area of emerging AQ and SP resistance in Mali. Malar J. 2009;8:34.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Maiga H, Lasry E, Diarra M, Sagara I, Bamadio A, Traore A, et al. Seasonal malaria chemoprevention with sulphadoxine-pyrimethamine and amodiaquine selects Pfdhfr-dhps quintuple mutant genotype in Mali. PLoS One. 2016;11:e0162718.PubMedPubMedCentralCrossRef Maiga H, Lasry E, Diarra M, Sagara I, Bamadio A, Traore A, et al. Seasonal malaria chemoprevention with sulphadoxine-pyrimethamine and amodiaquine selects Pfdhfr-dhps quintuple mutant genotype in Mali. PLoS One. 2016;11:e0162718.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Snow RW, Craig MH, Deichmann U, le Sueur D. A preliminary continental risk map for malaria mortality among African children. Parasitol Today. 1999;15:99–104.PubMedCrossRef Snow RW, Craig MH, Deichmann U, le Sueur D. A preliminary continental risk map for malaria mortality among African children. Parasitol Today. 1999;15:99–104.PubMedCrossRef
34.
Zurück zum Zitat Tanser FC, Sharp B, le Sueur D. Potential effect of climate change on malaria transmission in Africa. Lancet. 2003;362:1792–8.PubMedCrossRef Tanser FC, Sharp B, le Sueur D. Potential effect of climate change on malaria transmission in Africa. Lancet. 2003;362:1792–8.PubMedCrossRef
35.
Zurück zum Zitat Sogoba N, Keita M, Diakite M, Diallo M, Baber I, Manoukis N, et al. Malaria transmission along the Niger river in a Sudan Savanna area of Mali. Am J Trop Med Hyg. 2009;81:S170. Sogoba N, Keita M, Diakite M, Diallo M, Baber I, Manoukis N, et al. Malaria transmission along the Niger river in a Sudan Savanna area of Mali. Am J Trop Med Hyg. 2009;81:S170.
36.
Zurück zum Zitat WHO. Severe falciparum malaria World Health Organization, Communicable Diseases Cluster. Trans R Soc Trop Med Hyg. 2000;94(Suppl 1):S1–90. WHO. Severe falciparum malaria World Health Organization, Communicable Diseases Cluster. Trans R Soc Trop Med Hyg. 2000;94(Suppl 1):S1–90.
37.
Zurück zum Zitat Zhang L, Cui X, Schmitt K, Hubert R, Navidi W, Arnheim N. Whole genome amplification from a single cell: implications for genetic analysis. Proc Natl Acad Sci USA. 1992;89:5847–51.PubMedCrossRefPubMedCentral Zhang L, Cui X, Schmitt K, Hubert R, Navidi W, Arnheim N. Whole genome amplification from a single cell: implications for genetic analysis. Proc Natl Acad Sci USA. 1992;89:5847–51.PubMedCrossRefPubMedCentral
38.
Zurück zum Zitat Oyola SO, Ariani CV, Hamilton WL, Kekre M, Amenga-Etego LN, Ghansah A, et al. Whole genome sequencing of Plasmodium falciparum from dried blood spots using selective whole genome amplification. Malar J. 2016;15:597.PubMedPubMedCentralCrossRef Oyola SO, Ariani CV, Hamilton WL, Kekre M, Amenga-Etego LN, Ghansah A, et al. Whole genome sequencing of Plasmodium falciparum from dried blood spots using selective whole genome amplification. Malar J. 2016;15:597.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Manske M, Miotto O, Campino S, Auburn S, Almagro-Garcia J, Maslen G, et al. Analysis of Plasmodium falciparum diversity in natural infections by deep sequencing. Nature. 2012;487:375–9.PubMedPubMedCentralCrossRef Manske M, Miotto O, Campino S, Auburn S, Almagro-Garcia J, Maslen G, et al. Analysis of Plasmodium falciparum diversity in natural infections by deep sequencing. Nature. 2012;487:375–9.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Chang HH, Wesolowski A, Sinha I, Jacob CG, Mahmud A, Uddin D, et al. Mapping imported malaria in Bangladesh using parasite genetic and human mobility data. Elife. 2019;8:e43481.PubMedPubMedCentralCrossRef Chang HH, Wesolowski A, Sinha I, Jacob CG, Mahmud A, Uddin D, et al. Mapping imported malaria in Bangladesh using parasite genetic and human mobility data. Elife. 2019;8:e43481.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Galinsky K, Valim C, Salmier A, de Thoisy B, Musset L, Legrand E, et al. COIL: a methodology for evaluating malarial complexity of infection using likelihood from single nucleotide polymorphism data. Malar J. 2015;14:4.PubMedPubMedCentralCrossRef Galinsky K, Valim C, Salmier A, de Thoisy B, Musset L, Legrand E, et al. COIL: a methodology for evaluating malarial complexity of infection using likelihood from single nucleotide polymorphism data. Malar J. 2015;14:4.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Chang HH, Worby CJ, Yeka A, Nankabirwa J, Kamya MR, Staedke SG, et al. THE REAL McCOIL: a method for the concurrent estimation of the complexity of infection and SNP allele frequency for malaria parasites. PLoS Comput Biol. 2017;13:e1005348.PubMedPubMedCentralCrossRef Chang HH, Worby CJ, Yeka A, Nankabirwa J, Kamya MR, Staedke SG, et al. THE REAL McCOIL: a method for the concurrent estimation of the complexity of infection and SNP allele frequency for malaria parasites. PLoS Comput Biol. 2017;13:e1005348.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Ndam NT, Basco LK, Ngane VF, Ayouba A, Ngolle EM, Deloron P, et al. Reemergence of chloroquine-sensitive pfcrt K76 Plasmodium falciparum genotype in southeastern Cameroon. Malar J. 2017;16:130.PubMedPubMedCentralCrossRef Ndam NT, Basco LK, Ngane VF, Ayouba A, Ngolle EM, Deloron P, et al. Reemergence of chloroquine-sensitive pfcrt K76 Plasmodium falciparum genotype in southeastern Cameroon. Malar J. 2017;16:130.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Thomsen TT, Madsen LB, Hansson HH, Tomas EV, Charlwood D, Bygbjerg IC, et al. Rapid selection of Plasmodium falciparum chloroquine resistance transporter gene and multidrug resistance gene-1 haplotypes associated with past chloroquine and present artemether–lumefantrine use in Inhambane District, southern Mozambique. Am J Trop Med Hyg. 2013;88:536–41.PubMedPubMedCentralCrossRef Thomsen TT, Madsen LB, Hansson HH, Tomas EV, Charlwood D, Bygbjerg IC, et al. Rapid selection of Plasmodium falciparum chloroquine resistance transporter gene and multidrug resistance gene-1 haplotypes associated with past chloroquine and present artemether–lumefantrine use in Inhambane District, southern Mozambique. Am J Trop Med Hyg. 2013;88:536–41.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Laufer MK, Thesing PC, Dzinjalamala FK, Nyirenda OM, Masonga R, Laurens MB, et al. A longitudinal trial comparing chloroquine as monotherapy or in combination with artesunate, azithromycin or atovaquone-proguanil to treat malaria. PLoS One. 2012;7:e42284.PubMedPubMedCentralCrossRef Laufer MK, Thesing PC, Dzinjalamala FK, Nyirenda OM, Masonga R, Laurens MB, et al. A longitudinal trial comparing chloroquine as monotherapy or in combination with artesunate, azithromycin or atovaquone-proguanil to treat malaria. PLoS One. 2012;7:e42284.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Kublin JG, Cortese JF, Njunju EM, Mukadam RA, Wirima JJ, Kazembe PN, et al. Reemergence of chloroquine-sensitive Plasmodium falciparum malaria after cessation of chloroquine use in Malawi. J Infect Dis. 2003;187:1870–5.PubMedCrossRef Kublin JG, Cortese JF, Njunju EM, Mukadam RA, Wirima JJ, Kazembe PN, et al. Reemergence of chloroquine-sensitive Plasmodium falciparum malaria after cessation of chloroquine use in Malawi. J Infect Dis. 2003;187:1870–5.PubMedCrossRef
47.
Zurück zum Zitat Dokomajilar C, Lankoande ZM, Dorsey G, Zongo I, Ouedraogo JB, Rosenthal PJ. Roles of specific Plasmodium falciparum mutations in resistance to amodiaquine and sulfadoxine-pyrimethamine in Burkina Faso. Am J Trop Med Hyg. 2006;75:162–5.PubMedCrossRef Dokomajilar C, Lankoande ZM, Dorsey G, Zongo I, Ouedraogo JB, Rosenthal PJ. Roles of specific Plasmodium falciparum mutations in resistance to amodiaquine and sulfadoxine-pyrimethamine in Burkina Faso. Am J Trop Med Hyg. 2006;75:162–5.PubMedCrossRef
48.
Zurück zum Zitat Boussaroque A, Fall B, Madamet M, Wade KA, Fall M, Nakoulima A, et al. Prevalence of anti-malarial resistance genes in Dakar, Senegal from, to 2014. Malar J. 2013;15:347.CrossRef Boussaroque A, Fall B, Madamet M, Wade KA, Fall M, Nakoulima A, et al. Prevalence of anti-malarial resistance genes in Dakar, Senegal from, to 2014. Malar J. 2013;15:347.CrossRef
49.
Zurück zum Zitat Wurtz N, Fall B, Pascual A, Fall M, Baret E, Camara C, et al. Role of Pfmdr1 in in vitro Plasmodium falciparum susceptibility to chloroquine, quinine, monodesethylamodiaquine, mefloquine, lumefantrine, and dihydroartemisinin. Antimicrob Agents Chemother. 2014;58:7032–40.PubMedPubMedCentralCrossRef Wurtz N, Fall B, Pascual A, Fall M, Baret E, Camara C, et al. Role of Pfmdr1 in in vitro Plasmodium falciparum susceptibility to chloroquine, quinine, monodesethylamodiaquine, mefloquine, lumefantrine, and dihydroartemisinin. Antimicrob Agents Chemother. 2014;58:7032–40.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Dama S, Niangaly H, Ouattara A, Sagara I, Sissoko S, Traore OB, et al. Reduced ex vivo susceptibility of Plasmodium falciparum after oral artemether–lumefantrine treatment in Mali. Malar J. 2017;16:59.PubMedPubMedCentralCrossRef Dama S, Niangaly H, Ouattara A, Sagara I, Sissoko S, Traore OB, et al. Reduced ex vivo susceptibility of Plasmodium falciparum after oral artemether–lumefantrine treatment in Mali. Malar J. 2017;16:59.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Baraka V, Mavoko HM, Nabasumba C, Francis F, Lutumba P, Alifrangis M, et al. Impact of treatment and re-treatment with artemether–lumefantrine and artesunate-amodiaquine on selection of Plasmodium falciparum multidrug resistance gene-1 polymorphisms in the Democratic Republic of Congo and Uganda. PLoS One. 2018;13:e0191922.PubMedPubMedCentralCrossRef Baraka V, Mavoko HM, Nabasumba C, Francis F, Lutumba P, Alifrangis M, et al. Impact of treatment and re-treatment with artemether–lumefantrine and artesunate-amodiaquine on selection of Plasmodium falciparum multidrug resistance gene-1 polymorphisms in the Democratic Republic of Congo and Uganda. PLoS One. 2018;13:e0191922.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Maiga AW, Fofana B, Sagara I, Dembele D, Dara A, Traore OB, et al. No evidence of delayed parasite clearance after oral artesunate treatment of uncomplicated falciparum malaria in Mali. Am J Trop Med Hyg. 2012;87:23–8.PubMedPubMedCentralCrossRef Maiga AW, Fofana B, Sagara I, Dembele D, Dara A, Traore OB, et al. No evidence of delayed parasite clearance after oral artesunate treatment of uncomplicated falciparum malaria in Mali. Am J Trop Med Hyg. 2012;87:23–8.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat WWARN K13 Genotype-Phenotype Study Group. Association of mutations in the Plasmodium falciparum kelch13 gene (Pf3D7_1343700) with parasite clearance rates after artemisinin-based treatments-a WWARN individual patient data meta-analysis. BMC Med. 2019;17:1.CrossRef WWARN K13 Genotype-Phenotype Study Group. Association of mutations in the Plasmodium falciparum kelch13 gene (Pf3D7_1343700) with parasite clearance rates after artemisinin-based treatments-a WWARN individual patient data meta-analysis. BMC Med. 2019;17:1.CrossRef
54.
Zurück zum Zitat Anderson TJ, Haubold B, Williams JT, Estrada-Franco JG, Richardson L, Mollinedo R, et al. Microsatellite markers reveal a spectrum of population structures in the malaria parasite Plasmodium falciparum. Mol Biol Evol. 2000;17:1467–82.PubMedCrossRef Anderson TJ, Haubold B, Williams JT, Estrada-Franco JG, Richardson L, Mollinedo R, et al. Microsatellite markers reveal a spectrum of population structures in the malaria parasite Plasmodium falciparum. Mol Biol Evol. 2000;17:1467–82.PubMedCrossRef
55.
Zurück zum Zitat Mu J, Awadalla P, Duan J, McGee KM, Joy DA, McVean GA, et al. Recombination hotspots and population structure in Plasmodium falciparum. PLoS Biol. 2005;3:e335.PubMedPubMedCentralCrossRef Mu J, Awadalla P, Duan J, McGee KM, Joy DA, McVean GA, et al. Recombination hotspots and population structure in Plasmodium falciparum. PLoS Biol. 2005;3:e335.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Joy DA, Feng X, Mu J, Furuya T, Chotivanich K, Krettli AU, et al. Early origin and recent expansion of Plasmodium falciparum. Science. 2003;300:318–21.PubMedCrossRef Joy DA, Feng X, Mu J, Furuya T, Chotivanich K, Krettli AU, et al. Early origin and recent expansion of Plasmodium falciparum. Science. 2003;300:318–21.PubMedCrossRef
57.
Zurück zum Zitat Anthony TG, Conway DJ, Cox-Singh J, Matusop A, Ratnam S, Shamsul S, et al. Fragmented population structure of Plasmodium falciparum in a region of declining endemicity. J Infect Dis. 2005;191:1558–64.PubMedCrossRef Anthony TG, Conway DJ, Cox-Singh J, Matusop A, Ratnam S, Shamsul S, et al. Fragmented population structure of Plasmodium falciparum in a region of declining endemicity. J Infect Dis. 2005;191:1558–64.PubMedCrossRef
58.
Zurück zum Zitat Tsumori Y, Ndounga M, Sunahara T, Hayashida N, Inoue M, Nakazawa S, et al. Plasmodium falciparum: differential selection of drug resistance alleles in contiguous urban and peri-urban areas of Brazzaville, Republic of Congo. PLoS One. 2011;6:e23430.PubMedPubMedCentralCrossRef Tsumori Y, Ndounga M, Sunahara T, Hayashida N, Inoue M, Nakazawa S, et al. Plasmodium falciparum: differential selection of drug resistance alleles in contiguous urban and peri-urban areas of Brazzaville, Republic of Congo. PLoS One. 2011;6:e23430.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Babiker HA, Lines J, Hill WG, Walliker D. Population structure of Plasmodium falciparum in villages with different malaria endemicity in east Africa. Am J Trop Med Hyg. 1997;56:141–7.PubMedCrossRef Babiker HA, Lines J, Hill WG, Walliker D. Population structure of Plasmodium falciparum in villages with different malaria endemicity in east Africa. Am J Trop Med Hyg. 1997;56:141–7.PubMedCrossRef
60.
Zurück zum Zitat Ghanchi NK, Martensson A, Ursing J, Jafri S, Bereczky S, Hussain R, et al. Genetic diversity among Plasmodium falciparum field isolates in Pakistan measured with PCR genotyping of the merozoite surface protein 1 and 2. Malar J. 2010;9:1.PubMedPubMedCentralCrossRef Ghanchi NK, Martensson A, Ursing J, Jafri S, Bereczky S, Hussain R, et al. Genetic diversity among Plasmodium falciparum field isolates in Pakistan measured with PCR genotyping of the merozoite surface protein 1 and 2. Malar J. 2010;9:1.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Basco LK, Ringwald P. Molecular epidemiology of malaria in Yaounde, Cameroon. VII. Analysis of recrudescence and reinfection in patients with uncomplicated falciparum malaria. Am J Trop Med Hyg. 2000;63:215–21.PubMedCrossRef Basco LK, Ringwald P. Molecular epidemiology of malaria in Yaounde, Cameroon. VII. Analysis of recrudescence and reinfection in patients with uncomplicated falciparum malaria. Am J Trop Med Hyg. 2000;63:215–21.PubMedCrossRef
62.
Zurück zum Zitat Happi CT, Gbotosho GO, Sowunmi A, Falade CO, Akinboye DO, Gerena L, et al. Molecular analysis of Plasmodium falciparum recrudescent malaria infections in children treated with chloroquine in Nigeria. Am J Trop Med Hyg. 2004;70:20–6.PubMedCrossRef Happi CT, Gbotosho GO, Sowunmi A, Falade CO, Akinboye DO, Gerena L, et al. Molecular analysis of Plasmodium falciparum recrudescent malaria infections in children treated with chloroquine in Nigeria. Am J Trop Med Hyg. 2004;70:20–6.PubMedCrossRef
Metadaten
Titel
A comprehensive analysis of drug resistance molecular markers and Plasmodium falciparum genetic diversity in two malaria endemic sites in Mali
verfasst von
Seidina A. S. Diakité
Karim Traoré
Ibrahim Sanogo
Taane G. Clark
Susana Campino
Modibo Sangaré
Djeneba Dabitao
Antoine Dara
Drissa S. Konaté
Fousseyni Doucouré
Amadou Cissé
Bourama Keita
Mory Doumbouya
Merepen A. Guindo
Mahamoudou B. Toure
Nafomon Sogoba
Seydou Doumbia
Gordon A. Awandare
Mahamadou Diakité
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2019
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-019-2986-5

Weitere Artikel der Ausgabe 1/2019

Malaria Journal 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Neu im Fachgebiet Innere Medizin

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

Krebspatienten impfen: Was? Wen? Und wann nicht?

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

Nierenultraschall: Tipps vom Profi

22.04.2024 | DGIM 2024 | Kongressbericht | Nachrichten

„KI sieht, was wir nicht sehen“

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.