Skip to main content
Erschienen in: Malaria Journal 1/2020

Open Access 01.12.2020 | Research

Efficacy of artemether-lumefantrine for treating uncomplicated Plasmodium falciparum cases and molecular surveillance of drug resistance genes in Western Myanmar

verfasst von: Yanrui Wu, Myat Thut Soe, Pyae Linn Aung, Luyi Zhao, Weilin Zeng, Lynette Menezes, Zhaoqing Yang, Myat Phone Kyaw, Liwang Cui

Erschienen in: Malaria Journal | Ausgabe 1/2020

Abstract

Background

Currently, artemisinin-based combination therapy (ACT) is the first-line anti-malarial treatment in malaria-endemic areas. However, resistance in Plasmodium falciparum to artemisinin-based combinations emerging in the Greater Mekong Sub-region is a major problem hindering malaria elimination. To continuously monitor the potential spread of ACT-resistant parasites, this study assessed the efficacy of artemether-lumefantrine (AL) for falciparum malaria in western Myanmar.

Methods

Ninety-five patients with malaria symptoms from Paletwa Township, Chin State, Myanmar were screened for P. falciparum infections in 2015. After excluding six patients with a parasite density below 100 or over 150,000/µL, 41 P. falciparum patients were treated with AL and followed for 28 days. Molecular markers associated with resistance to 4-amino-quinoline drugs (pfcrt and pfmdr1), antifolate drugs (pfdhps and pfdhfr) and artemisinin (pfk13) were genotyped to determine the prevalence of mutations associated with anti-malarial drug resistance.

Results

For the 41 P. falciparum patients (27 children and 14 adults), the 28-day AL therapeutic efficacy was 100%, but five cases (12.2%) were parasite positive on day 3 by microscopy. For the pfk13 gene, the frequency of NN insert after the position 136 was 100% in the day-3 parasite-positive group as compared to 50.0% in the day-3 parasite-negative group, albeit the difference was not statistically significant (P = 0.113). The pfk13 K189T mutation (10.0%) was found in Myanmar for the first time. The pfcrt K76T and A220S mutations were all fixed in the parasite population. In pfmdr1, the Y184F mutation was present in 23.3% of the parasite population, and found in both day-3 parasite-positive and -negative parasites. The G968A mutation of pfmdr1 gene was first reported in Myanmar. Prevalence of all the mutations in pfdhfr and pfdhps genes assessed was over 70%, with the exception of the pfdhps A581G mutation, which was 3.3%.

Conclusions

AL remained highly efficacious in western Myanmar. Pfk13 mutations associated with artemisinin resistance were not found. The high prevalence of mutations in pfcrt, pfdhfr and pfdhps suggests high-degree resistance to chloroquine and antifolate drugs. The pfmdr1 N86/184F/D1246 haplotype associated with selection by AL in Africa reached > 20% in this study. The detection of > 10% patients who were day-3 parasite-positive after AL treatment emphasizes the necessity of continuously monitoring ACT efficacy in western Myanmar.
Hinweise
Yanrui Wu, Myat Thut Soe contributed equally to the work

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12936-020-03376-5.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACT
Artemisinin-based combination therapy
GMS
The Greater Mekong Subregion
AL
Artemether-lumefantrine
DP
Dihydroartemisinin/piperaquine
PC1/2
Parasite clearance half-life
RSA
Ring-stage survival assay
pfcrt
P. falciparum chloroquine resistance transporter
pfmdr1
P. falciparum multidrug resistance 1
pfdhfr
P. falciparum dihydrofolate reductase
pfdhps
P. falciparum dihydropteroate synthase
pfk13
P. falciparum Kelch13
RDT
Rapid diagnostic test
DBS
Dried blood spots
DNA
Deoxyribonucleic acid
nested-PCR
Nested polymerase chain reaction

Background

Malaria remains a major public health problem in tropical and sub-tropical regions of the world. According to the World Malaria Report 2019, it is estimated that there were 228 million malaria cases and 405,000 malaria-related deaths worldwide in 2018 [1]. Currently, malaria control relies primarily on measures targeting vectors (insecticide-treated bed nets and indoor residual spraying) and effective anti-malarial treatment of clinical cases [2]. Since 2001, artemisinin-based combination therapy (ACT) has been recommended as the first-line treatment for Plasmodium falciparum [3], and its widespread adoption in malaria treatment policies of endemic nations has played an important role in reducing malaria-related mortality and morbidity. The development of resistance in P. falciparum to artemisinins and partner drugs is a major threat to malaria control and elimination [4].
Artemisinin resistance first emerged in western Cambodia in 2007 [5, 6], and has since been detected in all countries of the Greater Mekong Sub-region (GMS), due to spread and/or independent emergence [7, 8]. ACT includes artemisinin or one of its derivatives and a partner drug such as lumefantrine, piperaquine, mefloquine, amodiaquine, and pyronaridine. Evolution of resistance in parasites to the artemisinins and the partner drugs would lead to clinical failures of ACT. In Cambodia, clinical resistance to two ACT, artesunate/mefloquine [9] and dihydroartemisinin/piperaquine (DP) [1013], has already been identified. To halt the spread of artemisinin resistance in the GMS, ACT efficacy has been monitored in multiple sentinel sites [1420]. Furthermore, to effectively contain artemisinin resistance in the GMS, countries within the GMS aim to eliminate P. falciparum malaria from this region by 2025 [21].
Clinically, artemisinin resistance manifests as delayed parasite clearance with parasite clearance half-life (PC1/2) exceeding 5 h, resulting in lingering parasitaemia 3 days after initiation of the treatment [17]. Accurate determination of parasite PC1/2 requires sampling of peripheral parasitaemia every 6 h after administration of the artemisinin drug [22]. In resource-limited settings, the day-3 parasite-positive rate can be used as a proxy measure of delayed parasite clearance [23]. Artemisinin resistance affects the ring stage, and dormant ring-stage parasites are able to endure the onslaught of artemisinins and later cause recrudescence of the disease [24]. To capture the ring stage-associated resistance phenotype, an in vitro or ex vivo ring-stage survival assay (RSA) measuring the proportion of the 0–3 h ring-stage parasites surviving 6 h of 700 nM dihydroartemisinin treatment was developed [25, 26]. In 2014, mutations in the propeller domain of the P. falciparum kelch13 (pfk13) gene were identified to be associated with artemisinin resistance [27], providing a molecular marker for surveillance of artemisinin resistance. A large-scale survey of P. falciparum populations identified as many as 108 non-synonymous pfk13 mutations, with wide variation in geographical distribution worldwide; mutations associated with delayed parasite clearance were identified only in Southeast Asia [28]. Likewise, within the GMS, P. falciparum populations showed striking disparity in the prevalence and distribution of pfk13 mutations, with the C580Y and F446I being the predominant pfk13 mutations in east and west GMS, respectively [27, 2931]. The NN insertion between amino acids 136 and 137 was associated with artemisinin resistance and its prevalence has increased dramatically over the years along the China-Myanmar border [20, 32].
Molecular markers associated with anti-malarial resistance are useful for resistance surveillance and elucidation of evolution of resistance in parasite populations [33]. Point mutations in the P. falciparum chloroquine resistance transporter (pfcrt) and the P. falciparum multidrug resistance 1 (pfmdr1) genes are associated with resistance to chloroquine (CQ) and certain 4-amino-quinoline drugs [34]. In Africa, the extensive deployment of artemether-lumefantrine (AL) has selected parasites with the wild-type N86 and pfmdr1 haplotype N86/184F/D1246 [3539]. In the folate biosynthesis pathway, mutations in P. falciparum dihydrofolate reductase (pfdhfr) and P. falciparum dihydropteroate synthase (pfdhps) genes as well as amplification of the GTP-cyclohydrolase gene are associated with resistance to the antifolate drugs sulfadoxine-pyrimethamine (SP) [40, 41].
From 2002, ACT has been deployed for the treatment of falciparum malaria in Myanmar and three ACT, AL, DP and artesunate-mefloquine are recommended [42]. In the GMS, Myanmar has the heaviest malaria burden and its geographical position bridging Southeast Asia and South Asia highlights the need to monitor potential westward spread of resistance. To date, clinical studies to monitor the efficacies of artemisinins or ACT detected artemisinin-resistant P. falciparum only in southern and eastern Myanmar [43, 44]. In comparison, ACT remained highly efficacious in northern, northeastern (at the China-Myanmar border) and western Myanmar [19, 4549]. Molecular surveillance also detected disparate distributions and prevalence of pfk13 mutations in different regions of Myanmar [29, 30, 4648, 50], providing a quick assessment of the artemisinin resistance situation. This study evaluated the clinical efficacy of AL for treating falciparum malaria in a western township of Myanmar bordering Bangladesh and India and studied the genetic polymorphisms in genes associated with resistance to AL (pfk13, pfcrt and pfmdr1). Given the extensive use of artesunate-SP in India, this study also genotyped the mutations in the pfdhfr and pfdhps genes.

Methods

Study site and population

Patients presenting with fever (axillary temperature ≥ 37.5 °C) or a history of fever within the previous 24 h and attending clinics at the Paletwa Township, Chin State, Myanmar (Fig. 1) in 2015, were screened for P. falciparum infection using the SD Bioline Malaria Ag P.f/Pan (Alere) rapid diagnostic test (RDT). RDT-positive P. falciparum patients were recruited into this study to evaluate the efficacy of AL. Exclusion criteria included severe malaria symptoms, anti-malarial drug use in the previous month, pregnant or lactating women and those with an intention to move out of the study area in the subsequent 2 months. Written informed consent was obtained from the participants or their guardians prior to enrolment. Assent was also obtained from children aged 7 to 17 years. Finger-prick blood samples were collected to make blood smears for microscopic confirmation and determination of parasite density. Patients with parasite density outside the range of 100–150,000 parasites/µL of blood were also excluded. Dried blood spots (DBS) were also prepared on Whatman 3 filter paper, air-dried and stored in individual plastic bags with desiccant. Ethical approval for this study was obtained from the ethical review committee of The Department of Medical Research, Ministry of Health and Sports, Myanmar.

Treatment and follow-up

RDT-positive P. falciparum patients were treated with AL (Coartem®) twice daily for a 3-day course. The target dose was calculated according to patient’s body weight (1.3 mg/kg artemether and 8 mg/kg lumefantrine). Patients were instructed to take the tablets and were checked for compliance daily during the follow-up visits on days 1–3. Patients were followed up to day 28 with blood smears collected on days 0, 1, 2, 3, 7, 14, 21, 28, and on any other day if the patient displayed malaria-related symptoms. All collected blood films were assessed for the presence of parasites by microscopy, with genotyping conducted to determine if the parasites were due to a recrudescence or new infection. According to the Myanmar National Malaria Treatment Guidelines, DP is the alternative ACT in case of AL treatment failures.

Plasmodium species identification

Thick smears were stained with 10% Giemsa for 30 min and examined at the field laboratory by microscopy under oil immersion. A smear was considered parasite negative if no parasites were seen after examination of 1000 white blood cells (WBCs). Parasite density, expressed as the number of asexual stage parasites per µL of blood, was calculated by counting the number of asexual stage parasites divided by 400 WBCs, assuming 5000 WBCs/µL blood for patients ≥ 5 years and 7000 WBCs/µL blood for children younger than 5 years [51]. To further confirm P. falciparum infections, parasite DNA was extracted from DBS using the QIAamp DNA micro kit (Qiagen, Hilde, Germany). Confirmation of Plasmodium infection and differentiation of other Plasmodium species including Plasmodium vivax, Plasmodium malariae, Plasmodium ovale, and Plasmodium knowlesi, were performed using PCR primers and conditions described previously [52, 53].

Amplification and sequencing of pfk13, pfcrt, pfmdr1, pfdhps and pfdhfr genes

The entire pfk13 gene was amplified using primers and protocol described earlier [30]. Primers for nested PCR of pfdhps and pfmdr1 fragment spanning codons 967–1290 are given in Additional file 1: Table S1. Mutations in exon 2 and 4 of pfcrt gene as well as the pfmdr1 fragment covering codons 77–190 were determined as described previously [54]. The target fragments of pfdhfr spanning codons 51–164 were amplified as described earlier [55]. The primary PCR volume was 25 μL, including 1 μM of each primer, 12.5 μL Premix Taq (TaKaRa Biotechnology Co., Ltd. Japan) and 1.5 μL genomic DNA. The nested PCR volume was 50 μL with 2 μM each primer, 25 μL Premix Taq and 2 μL amplified products of the primary PCR. PCR conditions were initial denaturation at 94 °C for 5 min; 35 cycles of 94 °C for 30 s, respective annealing temperatures for 30 s, and 68 °C for 30 s; final extension at 68 °C for 5 min. The amplified PCR products were separated by electrophoresis on 2% agarose gels and visualized after ethidium bromide staining. Then PCR products of different genes were purified and sequenced commercially (Sangon Biotech Co., Ltd. China).

Sequence analysis

The reference 3D7 sequences for pfk13 (PF3D7_1343700), pfcrt (PF3D7_0709000), pfmdr1 (PF3D7_0523000), pfdhps (PF3D7_0810800) and pfdhfr (PF3D7_0417200) were obtained from the online database (https://​plasmodb.​org/​plasmo/​). All sequences were aligned to respective reference genes by using the DNASTAR (version 7.1) software.

Statistical analysis

Statistical analysis was carried out using GraphPad Prism 6.0. The general characteristics of samples were described with mean and range. Frequencies of mutations and haplotypes between day-3 parasite-positive and day-3 -negative groups were compared using Fisher’s exact test. P value < 0.05 was considered statistically significant.

Results

Efficacy of AL in the study population

A total of 95 patients with fever or fever history were screened for P. falciparum infection. Of these 47 were RDT-positive for P. falciparum infection and were treated with AL. Plasmodium falciparum infections were confirmed by PCR. Microscopic examination of day-0 smears identified 6 samples with parasite density outside the 100–150,000 parasites/µL range, which were excluded from follow-up. The 41 patients included in the efficacy analysis had a median age of 12 years (range 9–60 years), and the majority presented with fever at enrolment (Table 1). Overall, no recurrent cases were detected within the 28 days of the follow-up, giving a 100% adequate clinical and parasitological response. However, there were 5 (12.2%) patients who remained parasite positive on day 3.
Table 1
Demographic and clinical characteristics of 41 enrolled patients with P. falciparum infection
Number of patients (% male)
41 (51%)
Age in years [median/(range)]
12 (9–60)
Body weight (kg) [mean (range)]
46.3 (29–64)
Patients with fever ≥ 37.5 °C on day 0
33 (80.5%)
Day 0 temperature (℃) [mean (range)]
37.9 (37–39)
Day 0 parasite density/µL [mean (range)]
13631 (131–55,309)

Mutations in molecular markers of drug resistance

PCR amplification and sequencing were successful from 36 of the 41 patients who were followed for 28 days. There were six samples with double peaks at eight polymorphic sites of the three resistance-associated genes, pfmdr1, pfdhps and pfdhfr, suggesting these samples contained mixed-strain infections (Additional file 2: Figure S1). Sequence data from these samples were excluded from allele and haplotype frequency analysis.

Pfk13 gene

For the artemisinin resistance marker pfk13, full-length sequences from the 30 samples did not identify any mutations in the propeller domain. The K189T substitution was detected in three samples (10.0%), which were from the day-3 parasite-negative samples. The NN insertion after amino acid 136 was detected in 17 (56.7%) samples (Table 2). The NN insert was present in all day-3 parasite-positive samples compared to 50.0% in day-3 parasite-negative samples, but the difference was not statistically significant (P = 0.113).
Table 2
Prevalence of mutations in molecular markers of day 0 samples from day 3 positive and negative patients after treatment with artemether-lumefantrine
Gene
Mutation
n (%) of isolates
Pa
Day 3 positive, n = 4
Day 3 negative, n = 26
Total, n = 30
pfk13
K189T
0 (0.0)
3 (11.5)
3 (10.0)
1.000
NN insertion
4 (100.0)
13 (50.0)
17 (56.7)
0.113
pfcrt
K76T
4 (100.0)
26 (100.0)
30 (100.0)
1.000
A220S
4 (100.0)
26 (100.0)
30 (100.0)
1.000
Pfmdr1
Y184F
1 (25.0)
6 (23.1)
7 (23.3)
1.000
G968A
0 (0.0)
1 (3.8)
1 (3.3)
1.000
pfdhps
S436A
4 (100.0)
20 (76.9)
24 (80.0)
0.557
A437G
4 (100.0)
26 (100.0)
30 (100.0)
1.000
K540E
4 (100.0)
23 (88.5)
27 (90.0)
1.000
A581G
0 (0.0)
1 (3.8)
1 (3.3)
1.000
pfdhfr
N51I
4 (100.0)
20 (76.9)
24 (80.0)
0.557
C59R
4 (100.0)
26 (100.0)
30 (100.0)
1.000
S108N
4 (100.0)
26 (100.0)
30 (100.0)
1.000
I164L
3 (75.0)
19 (73.1)
22 (73.3)
1.000
aComparison between the two groups by Fisher’s exact test. Sequences with mixed types were excluded for single mutation analysis

Pfcrt and pfmdr1 genes

Sequencing data for exon 2 and 4 of the pfcrt gene revealed that the mutant K76T and A220S alleles were present in all 30 samples analysed (Table 2). Of the previously reported pfmdr1 mutations, namely N86Y, Y184F, S1034D, N1042D, and D1246Y, only the Y184F mutation was identified with a prevalence of 23.3% (Table 2). The prevalence of non-synonymous substitution G968A was 3.3% and two synonymous (G182G and T1069T) changes were 16.7 and 3.3%, respectively. There were three pfmdr1 haplotypes constructed based on the amino acid substitutions (Table 3). Among them, the wild type Y184G968 had the highest frequency (73.3%), followed by F184G968 (23.3%) and Y184A968 (3.3%).
Table 3
Prevalence of pfcrt, pfmdr1, pfdhfr and pfdhps haplotypes in P. falciparum isolates from day 3 positive and negative patients after artemether-lumefantrine treatment
Gene
Mutants
Haplotype
n (%)
Pa
Day 3 positive, n = 4
Day 3 negative, n = 26
Total, n = 30
pfk13
Wild type
K189
0 (0.0)
10 (38.5)
10 (33.3)
0.272
Single
T189
0 (0.0)
3 (11.5)
3 (10.0)
1.000
NN K189
4 (100.0)
13 (50.0)
17 (56.7)
0.113
pfcrt
Double
T76S220
4 (100.0)
26 (100.0)
30 (100)
1.000
pfmdr1
Wild type
Y184G968
3 (75.0)
19 (73.1)
22 (73.3)
1.000
Single
F184G968
1 (25.0)
6 (23.1)
7 (23.3)
1.000
Y184A968
0 (0.0)
1 (3.8)
1 (3.3)
1.000
pfdhps
Single
S436G437K540A581
0 (0.0)
2 (7.7)
2 (6.7)
1.000
Double
S436G437E540A581
0 (0.0)
3 (11.5)
3 (10.0)
1.000
A436G437K540A581
0 (0.0)
1 (3.8)
1 (3.3)
1.000
Triple
S436G437E540G581
0 (0.0)
1 (3.8)
1 (3.3)
1.000
A436G437E540A581
4 (100.0)
19 (73.1)
23 (76.7)
0.548
pfdhfr
Double
N51R59N108I164
0 (0.0)
2 (7.7)
2 (6.7)
1.000
Triple
N51R59N108L164
0 (0.0)
4 (15.4)
4 (13.3)
1.000
I51R59N108I164
1 (25.0)
5 (19.2)
6 (20.0)
1.000
Quadruple
I51R59N108L164
3 (75.0)
15 (57.7)
18 (60.0)
0.632
pfdhpspfdhfr
Quadruple
S436G437K540A581–N51R59N108L164
0 (0.0)
2 (7.7)
2 (6.7)
1.000
S436G437E540A581–N51R59N108I164
0 (0.0)
1 (3.8)
1 (3.3)
1.000
Quintuple
S436G437E540A581I51R59N108I164
0 (0.0)
2 (7.7)
2 (6.7)
1.000
> Quintuple
S436G437E540G581I51R59N108I164
0 (0.0)
1 (3.8)
1 (3.3)
1.000
A436G437K540A581I51R59N108L164
0 (0.0)
1 (3.8)
1 (3.3)
1.000
A436G437E540A581–N51R59N108I164
0 (0.0)
1 (3.8)
1 (3.3)
1.000
A436G437E540A581–N51R59N108L164
0 (0.0)
2 (7.7)
2 (6.7)
1.000
A436G437E540A581I51R59N108I164
1 (25.0)
2 (7.7)
3 (10.0)
0.360
A436G437E540A581I51R59N108L164
3 (75.0)
14 (53.8)
17 (56.7)
0.613
Mutant amino acids are highlighted in bold
aComparison between the two groups by Fisher’s exact test. Sequences with mixed types were excluded for haplotype analysis

Pfdhps and pfdhfr genes

There were no wild-type parasites at the pfdhps and pfdhfr genes (Table 2). Most of the mutations (S436A, A437G and K540E in the pfdhps gene; N51I, C59R, S108 N and I164L in the pfdhfr gene) exceeded 70%, whereas A581G in pfdhps was low at 3.3%. For the pfdhps gene, five haplotypes were found in the samples and the triple mutant haplotype A436G437E540A581 was the most common (76.7%) compared with the triple mutant haplotype S436G437E540G581 (3.3%). The double mutant haplotypes were A436G437K540A581 (3.3%) and S436G437E540A581 (10.0%) and the single mutant haplotype was S436G437K540A581 (6.7%). For pfdhfr, there were four haplotypes and the quadruple mutant haplotype I51R59N108L164 was found most frequent (60.0%) followed by the two triple mutant haplotypes I51R59N108I164 (20.0%) and N51R59N108L164 (13.3%) and the double mutant haplotype N51R59N108I164 (6.7%).
Since quintuple mutations in pfdhps (437G and 540E) and pfdhfr (51I, 59R and 108N) were linked to clinical treatment failure of SP [56], the combination of the pfdhps and pfdhfr mutations was further evaluated. Of the total of eight pfdhps-pfdhfr haplotypes, the haplotype with triple pfdhps and quadruple pfdhfr mutations (A436G437E540A581I51R59N108L164) was the most common at 56.7%. Three additional haplotypes (A436G437E540A581I51R59N108I164, S436G437E540A581I51R59N108I164 and S436G437E540G581I51R59N108I164), which all contained the aforementioned quintuple mutations were equally represented at 20.0%.

Discussion

The emergence and spread of P. falciparum resistance to artemisinin in GMS is of great concern and demands the monitoring of clinical efficacy of ACT in malaria-endemic areas of the region. Myanmar occupies an important position in artemisinin resistance containment, because it was among the highest malaria burden countries in the GMS and is geographically linked to the Indian sub-continent [57]. Since the detection of artemisinin resistance in Cambodia [6], delayed parasite clearance in patients after ACT or artesunate treatment was first detected in southern Myanmar in 2010 [42, 44]. One study conducted in northern Myanmar reported 30% of day-3 parasite positivity after treatment with DP in 2013 [48]. The artemisinin resistance phenotype was also documented in eastern (37.1%) [43] and northeastern (23.1%) [20] Myanmar after treatment with artesunate. In southeastern Myanmar, 20% of the cases were still parasitaemic on day 3 after treatment with AL [49]. Despite the presence of artemisinin resistance, ACT still demonstrated high therapeutic efficacies (95.9–100%) in the above areas. In western Myanmar, artemisinin resistance has not been detected. This study confirmed the absence of clinical artemisinin resistance in western Myanmar, with AL demonstrating 100% therapeutic efficacy with no recrudescence within 28 days of follow-up. Although the number of patients tested here was relatively small, the day-28 therapeutic efficacy of AL was consistent with previous studies conducted in the same area [46, 47]. However, the day-3 parasite-positive cases (12.2%) just exceeded the 10% threshold recommended by WHO for suspected emergence of artemisinin resistance.
Artemisinin resistance has been associated with mutations in the propeller domain of pfk13 [27]. Several mutations including N458Y, Y493H, R539T, I543T, and C580Y have been genetically validated to confer artemisinin resistance [58]. The NN insert outside of the propeller domain has also been reported to be correlated with artemisinin resistance, initially in China-Myanmar border [20]. This insert has increased in prevalence over the years and reached 100% in samples collected in 2014–2016 [32]. No mutations in the propeller domain of the pfk13 gene were identified in the present study, whereas the NN insert was present in 56.7% patients. This is consistent with a recent study of asymptomatic P. falciparum infections in this region showing NN insert as the most popular mutation [59]. Although all of the day-3 parasite-positive samples in the present study harboured NN insert compared to 50% among the day-3 parasite-negative cases, the sample size was too small to perform a robust assessment of the potential association of the NN insert with day-3 parasitaemia. Further investigations are needed to explore the functions of this mutation. The K189T mutation was identified in Myanmar for the first time. This mutation was previously observed in northeast India near Myanmar [60, 61], but it was not associated with increased clearance half-life [14]. The study findings suggest that continuous monitoring of pfk13 gene mutations and RSA in western Myanmar is warranted.
Several studies investigated the relationship between AL treatment and selection of molecular markers associated with treatment failures. Whereas there was no indication of artemisinin resistance-associated pfk13 mutations, markedly increased prevalence of pfmdr1 N86 and pfcrt K76 wild-type alleles was associated with extensive use of AL [62]. An in vitro study linked the wild-type pfmdr1 N86 with reduced lumefantrine activity [36], consistent with the selection of wild-type K76 by lumefantrine [63]. In pfmdr1, AL results in the selection of the N86/184F/D1246 haplotype [3739]. The present study showed that all samples were fixed at K76T and A220S mutations in pfcrt, but remained wild type at the pfmdr1 N86 and D1246. The high prevalence of mutations in pfcrt gene may be the result of continued drug pressure of CQ for treating P. vivax infections in Myanmar [64]. In pfmdr1 gene, Y184F had a frequency of 23.3% and there was no statistically significant association between Y184F and the day-3 parasite-positive and -negative phenotypes (P = 1.000, Fisher’s exact test). These results were similar to a recent study, which showed the extremely low frequency of N86Y and a moderate prevalence of Y184F in asymptomatic malaria carriers in western Myanmar [59]. The moderate prevalence of the N86/184F/D1246 haplotype associated with AL selection desires further monitoring.
In recognition of the extensive deployment of the artesunate-SP in India, this study also evaluated pfdhfr and pfdhps mutations and detected high prevalence of pfdhfr (N51I, C59R, S108 N and I164L) and pfdhps (S436A, A437G and K540E) mutations. Interestingly, these mutations were even more prevalent than previously reported from central Myanmar [59]. The quintuple mutant of pfdhps gene (437G and 540E) and pfdhfr gene (51I, 59R and 108N) was the significant predictor of clinical treatment failure [56]. Four combined haplotypes containing these quintuple mutations exceeded 70%. In addition, the pfhdfr I164L mutation associated with SP failures in Asia [65] also had > 70% prevalence, indicating high-degree SP resistance in this region.
While this study constitutes continued efforts of monitoring the efficacy of anti-malarial drugs in the GMS, it has several limitations. The study reflects the situation that was 5 years ago, and an update is highly desired. The number of patients recruited to this study was small, and an expanded sample size is needed to obtain more accurate estimates of the resistance phenotype. In addition, future studies should extend the follow-up period to 42 days. Furthermore, future studies should also include larger areas along the western Myanmar border to better capture the broad picture of ACT efficacy.

Conclusions

This study showed that AL was still efficacious for treating uncomplicated falciparum malaria in western Myanmar. Yet, the appearance of day-3 parasitaemia after AL treatment is a warning sign of potential development of artemisinin resistance. Whereas no mutations were identified in pfk13, resistance-conferring mutations in pfcrt, pfdhps and pfdhfr genes were highly prevalent, suggesting parasites from this region were resistant to chloroquine and antifolate drugs, and potentially other 4-aminoquinoline drugs. Given the strategic location of Myanmar and the high proportion of P. falciparum malaria in western Myanmar, continuous surveillance of therapeutic efficacy of ACT and molecular markers of resistance to both artemisinin and partner drugs, is strongly recommended, which echoes with the WHO’s advice that anti-malarial drug efficacy should be monitored at least once every 24 months in order to provide critical evidence for timely modification of malaria treatment policy [66].

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12936-020-03376-5.

Acknowledgements

The authors thank the staff at the clinics and patients for participation in this study.
Ethical approval for the study was given by the ethical review committee of The Department of Medical Research, Myanmar. Written informed consent was gathered from the participants or their guardians prior to enrolment.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

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 2019. Geneva: World Health Organization; 2019. WHO. World malaria report 2019. Geneva: World Health Organization; 2019.
2.
Zurück zum Zitat Bhatt S, Weiss DJ, Cameron E, Bisanzio D, Mappin B, Dalrymple U, et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015;526:207–11.PubMedPubMedCentral Bhatt S, Weiss DJ, Cameron E, Bisanzio D, Mappin B, Dalrymple U, et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015;526:207–11.PubMedPubMedCentral
3.
Zurück zum Zitat WHO. Antimalarial drug combination therapy. Report of a WHO Technical Consultation. Geneva: World Health Organization; 2001. WHO. Antimalarial drug combination therapy. Report of a WHO Technical Consultation. Geneva: World Health Organization; 2001.
4.
Zurück zum Zitat Menard D, Dondorp A. Antimalarial drug resistance: a threat to malaria elimination. Cold Spring Harb Perspect Med. 2017;7:a025619.PubMedPubMedCentral Menard D, Dondorp A. Antimalarial drug resistance: a threat to malaria elimination. Cold Spring Harb Perspect Med. 2017;7:a025619.PubMedPubMedCentral
5.
Zurück zum Zitat Dondorp AM, Nosten F, Yi P, Das D, Phyo AP, Tarning J, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361:455–67.PubMedPubMedCentral Dondorp AM, Nosten F, Yi P, Das D, Phyo AP, Tarning J, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361:455–67.PubMedPubMedCentral
6.
Zurück zum Zitat Noedl H, Se Y, Schaecher K, Smith BL, Socheat D, Fukuda MM, et al. Evidence of artemisinin-resistant malaria in western Cambodia. N Engl J Med. 2008;359:2619–20.PubMed Noedl H, Se Y, Schaecher K, Smith BL, Socheat D, Fukuda MM, et al. Evidence of artemisinin-resistant malaria in western Cambodia. N Engl J Med. 2008;359:2619–20.PubMed
7.
Zurück zum Zitat Imwong M, Suwannasin K, Kunasol C, Sutawong K, Mayxay M, Rekol H, et al. The spread of artemisinin-resistant Plasmodium falciparum in the Greater Mekong subregion: a molecular epidemiology observational study. Lancet Infect Dis. 2017;17:491–7.PubMedPubMedCentral Imwong M, Suwannasin K, Kunasol C, Sutawong K, Mayxay M, Rekol H, et al. The spread of artemisinin-resistant Plasmodium falciparum in the Greater Mekong subregion: a molecular epidemiology observational study. Lancet Infect Dis. 2017;17:491–7.PubMedPubMedCentral
8.
Zurück zum Zitat Takala-Harrison S, Jacob CG, Arze C, Cummings MP, Silva JC, Dondorp AM, et al. Independent emergence of artemisinin resistance mutations among Plasmodium falciparum in Southeast Asia. J Infect Dis. 2015;211:670–9.PubMed Takala-Harrison S, Jacob CG, Arze C, Cummings MP, Silva JC, Dondorp AM, et al. Independent emergence of artemisinin resistance mutations among Plasmodium falciparum in Southeast Asia. J Infect Dis. 2015;211:670–9.PubMed
9.
Zurück zum Zitat Wongsrichanalai C, Meshnick SR. Declining artesunate-mefloquine efficacy against falciparum malaria on the Cambodia-Thailand border. Emerg Infect Dis. 2008;14:716–9.PubMedPubMedCentral Wongsrichanalai C, Meshnick SR. Declining artesunate-mefloquine efficacy against falciparum malaria on the Cambodia-Thailand border. Emerg Infect Dis. 2008;14:716–9.PubMedPubMedCentral
10.
Zurück zum Zitat Amaratunga C, Lim P, Suon S, Sreng S, Mao S, Sopha C, et al. Dihydroartemisinin-piperaquine resistance in Plasmodium falciparum malaria in Cambodia: a multisite prospective cohort study. Lancet Infect Dis. 2016;16:357–65.PubMedPubMedCentral Amaratunga C, Lim P, Suon S, Sreng S, Mao S, Sopha C, et al. Dihydroartemisinin-piperaquine resistance in Plasmodium falciparum malaria in Cambodia: a multisite prospective cohort study. Lancet Infect Dis. 2016;16:357–65.PubMedPubMedCentral
11.
Zurück zum Zitat Leang R, Taylor WR, Bouth DM, Song L, Tarning J, Char MC, et al. Evidence of Plasmodium falciparum malaria multidrug resistance to artemisinin and piperaquine in Western Cambodia: dihydroartemisinin-piperaquine open-label multicenter clinical assessment. Antimicrob Agents Chemother. 2015;59:4719–26.PubMedPubMedCentral Leang R, Taylor WR, Bouth DM, Song L, Tarning J, Char MC, et al. Evidence of Plasmodium falciparum malaria multidrug resistance to artemisinin and piperaquine in Western Cambodia: dihydroartemisinin-piperaquine open-label multicenter clinical assessment. Antimicrob Agents Chemother. 2015;59:4719–26.PubMedPubMedCentral
12.
Zurück zum Zitat Saunders DL, Vanachayangkul P, Lon C. Dihydroartemisinin-piperaquine failure in Cambodia. N Engl J Med. 2014;371:484–5.PubMed Saunders DL, Vanachayangkul P, Lon C. Dihydroartemisinin-piperaquine failure in Cambodia. N Engl J Med. 2014;371:484–5.PubMed
13.
Zurück zum Zitat Spring MD, Lin JT, Manning JE, Vanachayangkul P, Somethy S, Bun R, et al. Dihydroartemisinin-piperaquine failure associated with a triple mutant including kelch13 C580Y in Cambodia: an observational cohort study. Lancet Infect Dis. 2015;15:683–91.PubMed Spring MD, Lin JT, Manning JE, Vanachayangkul P, Somethy S, Bun R, et al. Dihydroartemisinin-piperaquine failure associated with a triple mutant including kelch13 C580Y in Cambodia: an observational cohort study. Lancet Infect Dis. 2015;15:683–91.PubMed
14.
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.PubMedPubMedCentral 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.PubMedPubMedCentral
15.
Zurück zum Zitat Hien TT, Thuy-Nhien NT, Phu NH, Boni MF, Thanh NV, Nha-Ca NT, et al. In vivo susceptibility of Plasmodium falciparum to artesunate in Binh Phuoc Province. Vietnam. Malar J. 2012;11:355.PubMed Hien TT, Thuy-Nhien NT, Phu NH, Boni MF, Thanh NV, Nha-Ca NT, et al. In vivo susceptibility of Plasmodium falciparum to artesunate in Binh Phuoc Province. Vietnam. Malar J. 2012;11:355.PubMed
16.
Zurück zum Zitat Huang F, Tang L, Yang H, Zhou S, Sun X, Liu H. Therapeutic efficacy of artesunate in the treatment of uncomplicated Plasmodium falciparum malaria and anti-malarial, drug-resistance marker polymorphisms in populations near the China-Myanmar border. Malar J. 2012;11:278.PubMedPubMedCentral Huang F, Tang L, Yang H, Zhou S, Sun X, Liu H. Therapeutic efficacy of artesunate in the treatment of uncomplicated Plasmodium falciparum malaria and anti-malarial, drug-resistance marker polymorphisms in populations near the China-Myanmar border. Malar J. 2012;11:278.PubMedPubMedCentral
17.
Zurück zum Zitat Phyo AP, Nkhoma S, Stepniewska K, Ashley EA, Nair S, McGready R, et al. Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet. 2012;379:1960–6.PubMedPubMedCentral Phyo AP, Nkhoma S, Stepniewska K, Ashley EA, Nair S, McGready R, et al. Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet. 2012;379:1960–6.PubMedPubMedCentral
18.
Zurück zum Zitat Thanh NV, Thuy-Nhien N, Tuyen NT, Tong NT, Nha-Ca NT, Dong LT, et al. Rapid decline in the susceptibility of Plasmodium falciparum to dihydroartemisinin-piperaquine in the south of Vietnam. Malar J. 2017;16:27.PubMedPubMedCentral Thanh NV, Thuy-Nhien N, Tuyen NT, Tong NT, Nha-Ca NT, Dong LT, et al. Rapid decline in the susceptibility of Plasmodium falciparum to dihydroartemisinin-piperaquine in the south of Vietnam. Malar J. 2017;16:27.PubMedPubMedCentral
19.
Zurück zum Zitat Wang Y, Yang Z, Yuan L, Zhou G, Parker D, Lee MC, et al. Clinical efficacy of dihydroartemisinin-piperaquine for the treatment of uncomplicated Plasmodium falciparum malaria at the China-Myanmar border. Am J Trop Med Hyg. 2015;93:577–83.PubMedPubMedCentral Wang Y, Yang Z, Yuan L, Zhou G, Parker D, Lee MC, et al. Clinical efficacy of dihydroartemisinin-piperaquine for the treatment of uncomplicated Plasmodium falciparum malaria at the China-Myanmar border. Am J Trop Med Hyg. 2015;93:577–83.PubMedPubMedCentral
20.
Zurück zum Zitat Wang Z, Wang Y, Cabrera M, Zhang Y, Gupta B, Wu Y, et al. Artemisinin resistance at the China-Myanmar border and association with mutations in the K13 propeller gene. Antimicrob Agents Chemother. 2015;59:6952–9.PubMedPubMedCentral Wang Z, Wang Y, Cabrera M, Zhang Y, Gupta B, Wu Y, et al. Artemisinin resistance at the China-Myanmar border and association with mutations in the K13 propeller gene. Antimicrob Agents Chemother. 2015;59:6952–9.PubMedPubMedCentral
21.
Zurück zum Zitat WHO. Eliminating malaria in the Greater Mekong Subregion: United to end a deadly disease. Geneva: World Health Organization; 2016. WHO. Eliminating malaria in the Greater Mekong Subregion: United to end a deadly disease. Geneva: World Health Organization; 2016.
22.
Zurück zum Zitat Flegg JA, Guerin PJ, Nosten F, Ashley EA, Phyo AP, Dondorp AM, et al. Optimal sampling designs for estimation of Plasmodium falciparum clearance rates in patients treated with artemisinin derivatives. Malar J. 2013;12:411.PubMedPubMedCentral Flegg JA, Guerin PJ, Nosten F, Ashley EA, Phyo AP, Dondorp AM, et al. Optimal sampling designs for estimation of Plasmodium falciparum clearance rates in patients treated with artemisinin derivatives. Malar J. 2013;12:411.PubMedPubMedCentral
23.
Zurück zum Zitat WHO. Global plan for artemisinin resistance containment (GPARC). Geneva: World Health Organization; 2011. WHO. Global plan for artemisinin resistance containment (GPARC). Geneva: World Health Organization; 2011.
24.
Zurück zum Zitat Teuscher F, Gatton ML, Chen N, Peters J, Kyle DE, Cheng Q. Artemisinin-induced dormancy in Plasmodium falciparum: duration, recovery rates, and implications in treatment failure. J Infect Dis. 2010;202:1362–8.PubMedPubMedCentral Teuscher F, Gatton ML, Chen N, Peters J, Kyle DE, Cheng Q. Artemisinin-induced dormancy in Plasmodium falciparum: duration, recovery rates, and implications in treatment failure. J Infect Dis. 2010;202:1362–8.PubMedPubMedCentral
25.
Zurück zum Zitat Witkowski B, Amaratunga C, Khim N, Sreng S, Chim P, Kim S, et al. Novel phenotypic assays for the detection of artemisinin-resistant Plasmodium falciparum malaria in Cambodia: in vitro and ex vivo drug-response studies. Lancet Infect Dis. 2013;13:1043–9.PubMedPubMedCentral Witkowski B, Amaratunga C, Khim N, Sreng S, Chim P, Kim S, et al. Novel phenotypic assays for the detection of artemisinin-resistant Plasmodium falciparum malaria in Cambodia: in vitro and ex vivo drug-response studies. Lancet Infect Dis. 2013;13:1043–9.PubMedPubMedCentral
26.
Zurück zum Zitat Zhang J, Feng GH, Zou CY, Su PC, Liu HE, Yang ZQ. Overview of the improvement of the ring-stage survival assay-a novel phenotypic assay for the detection of artemisinin-resistant Plasmodium falciparum. Zool Res. 2017;38:317–20.PubMedPubMedCentral Zhang J, Feng GH, Zou CY, Su PC, Liu HE, Yang ZQ. Overview of the improvement of the ring-stage survival assay-a novel phenotypic assay for the detection of artemisinin-resistant Plasmodium falciparum. Zool Res. 2017;38:317–20.PubMedPubMedCentral
27.
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.PubMed 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.PubMed
28.
Zurück zum Zitat Menard D, Khim N, Beghain J, Adegnika AA, Shafiul-Alam M, Amodu O, et al. A worldwide map of Plasmodium falciparum K13-propeller polymorphisms. N Engl J Med. 2016;374:2453–64.PubMedPubMedCentral Menard D, Khim N, Beghain J, Adegnika AA, Shafiul-Alam M, Amodu O, et al. A worldwide map of Plasmodium falciparum K13-propeller polymorphisms. N Engl J Med. 2016;374:2453–64.PubMedPubMedCentral
29.
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.PubMedPubMedCentral 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.PubMedPubMedCentral
30.
Zurück zum Zitat Wang Z, Shrestha S, Li X, Miao J, Yuan L, Cabrera M, Grube C, Yang Z, Cui L. Prevalence of K13-propeller polymorphisms in Plasmodium falciparum from China-Myanmar border in 2007–2012. Malar J. 2015;14:168.PubMedPubMedCentral Wang Z, Shrestha S, Li X, Miao J, Yuan L, Cabrera M, Grube C, Yang Z, Cui L. Prevalence of K13-propeller polymorphisms in Plasmodium falciparum from China-Myanmar border in 2007–2012. Malar J. 2015;14:168.PubMedPubMedCentral
31.
Zurück zum Zitat Ye R, Hu D, Zhang Y, Huang Y, Sun X, Wang J, et al. Distinctive origin of artemisinin-resistant Plasmodium falciparum on the China-Myanmar border. Sci Rep. 2016;6:20100.PubMedPubMedCentral Ye R, Hu D, Zhang Y, Huang Y, Sun X, Wang J, et al. Distinctive origin of artemisinin-resistant Plasmodium falciparum on the China-Myanmar border. Sci Rep. 2016;6:20100.PubMedPubMedCentral
32.
Zurück zum Zitat Zhang J, Li N, Siddiqui FA, Xu S, Geng J, He X, et al. In vitro susceptibility of Plasmodium falciparum isolates from the China-Myanmar border area to artemisinins and correlation with K13 mutations. Int J Parasitol Drugs Drug Resist. 2019;10:20–7.PubMedPubMedCentral Zhang J, Li N, Siddiqui FA, Xu S, Geng J, He X, et al. In vitro susceptibility of Plasmodium falciparum isolates from the China-Myanmar border area to artemisinins and correlation with K13 mutations. Int J Parasitol Drugs Drug Resist. 2019;10:20–7.PubMedPubMedCentral
33.
34.
Zurück zum Zitat Sanchez CP, Dave A, Stein WD, Lanzer M. Transporters as mediators of drug resistance in Plasmodium falciparum. Int J Parasitol. 2010;40:1109–18.PubMed Sanchez CP, Dave A, Stein WD, Lanzer M. Transporters as mediators of drug resistance in Plasmodium falciparum. Int J Parasitol. 2010;40:1109–18.PubMed
35.
Zurück zum Zitat Sisowath C, Stromberg J, Martensson A, Msellem M, Obondo C, Bjorkman A, Gil JP. In vivo selection of Plasmodium falciparum pfmdr1 86 N coding alleles by artemether-lumefantrine (Coartem). J Infect Dis. 2005;191:1014–7.PubMed Sisowath C, Stromberg J, Martensson A, Msellem M, Obondo C, Bjorkman A, Gil JP. In vivo selection of Plasmodium falciparum pfmdr1 86 N coding alleles by artemether-lumefantrine (Coartem). J Infect Dis. 2005;191:1014–7.PubMed
36.
Zurück zum Zitat Mwai L, Kiara SM, Abdirahman A, Pole L, Rippert A, Diriye A, et al. In vitro activities of piperaquine, lumefantrine, and dihydroartemisinin in Kenyan Plasmodium falciparum isolates and polymorphisms in pfcrt and pfmdr1. Antimicrob Agents Chemother. 2009;53:5069–73.PubMedPubMedCentral Mwai L, Kiara SM, Abdirahman A, Pole L, Rippert A, Diriye A, et al. In vitro activities of piperaquine, lumefantrine, and dihydroartemisinin in Kenyan Plasmodium falciparum isolates and polymorphisms in pfcrt and pfmdr1. Antimicrob Agents Chemother. 2009;53:5069–73.PubMedPubMedCentral
37.
Zurück zum Zitat Mbaye A, Dieye B, Ndiaye YD, Bei AK, Muna A, Deme AB, et al. Selection of N86F184D1246 haplotype of Pfmrd1 gene by artemether-lumefantrine drug pressure on Plasmodium falciparum populations in Senegal. Malar J. 2016;15:433.PubMedPubMedCentral Mbaye A, Dieye B, Ndiaye YD, Bei AK, Muna A, Deme AB, et al. Selection of N86F184D1246 haplotype of Pfmrd1 gene by artemether-lumefantrine drug pressure on Plasmodium falciparum populations in Senegal. Malar J. 2016;15:433.PubMedPubMedCentral
38.
Zurück zum Zitat Malmberg M, Ngasala B, Ferreira PE, Larsson E, Jovel I, Hjalmarsson A, et al. Temporal trends of molecular markers associated with artemether-lumefantrine tolerance/resistance in Bagamoyo district, Tanzania. Malar J. 2013;12:103.PubMedPubMedCentral Malmberg M, Ngasala B, Ferreira PE, Larsson E, Jovel I, Hjalmarsson A, et al. Temporal trends of molecular markers associated with artemether-lumefantrine tolerance/resistance in Bagamoyo district, Tanzania. Malar J. 2013;12:103.PubMedPubMedCentral
39.
Zurück zum Zitat Thomsen TT, Madsen LB, Hansson HH, Tomas EV, Charlwood D, Bygbjerg IC, Alifrangis M. 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.PubMedPubMedCentral Thomsen TT, Madsen LB, Hansson HH, Tomas EV, Charlwood D, Bygbjerg IC, Alifrangis M. 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.PubMedPubMedCentral
40.
Zurück zum Zitat Gregson A, Plowe CV. Mechanisms of resistance of malaria parasites to antifolates. Pharmacol Rev. 2005;57:117–45.PubMed Gregson A, Plowe CV. Mechanisms of resistance of malaria parasites to antifolates. Pharmacol Rev. 2005;57:117–45.PubMed
41.
Zurück zum Zitat Heinberg A, Kirkman L. The molecular basis of antifolate resistance in Plasmodium falciparum: looking beyond point mutations. Ann N Y Acad Sci. 2015;1342:10–8.PubMedPubMedCentral Heinberg A, Kirkman L. The molecular basis of antifolate resistance in Plasmodium falciparum: looking beyond point mutations. Ann N Y Acad Sci. 2015;1342:10–8.PubMedPubMedCentral
42.
Zurück zum Zitat WHO. Global report on antimalarial efficacy and drug resistance: 2000–2010. Geneva: World Health Organization; 2010. WHO. Global report on antimalarial efficacy and drug resistance: 2000–2010. Geneva: World Health Organization; 2010.
43.
Zurück zum Zitat Bonnington CA, Phyo AP, Ashley EA, Imwong M, Sriprawat K, Parker DM, Proux S, White NJ, Nosten F. Plasmodium falciparum Kelch 13 mutations and treatment response in patients in Hpa-Pun District, Northern Kayin State, Myanmar. Malar J. 2017;16:480.PubMedPubMedCentral Bonnington CA, Phyo AP, Ashley EA, Imwong M, Sriprawat K, Parker DM, Proux S, White NJ, Nosten F. Plasmodium falciparum Kelch 13 mutations and treatment response in patients in Hpa-Pun District, Northern Kayin State, Myanmar. Malar J. 2017;16:480.PubMedPubMedCentral
44.
Zurück zum Zitat Kyaw MP, Nyunt MH, Chit K, Aye MM, Aye KH, Aye MM, et al. Reduced susceptibility of Plasmodium falciparum to artesunate in southern Myanmar. PLoS ONE. 2013;8:e57689.PubMedPubMedCentral Kyaw MP, Nyunt MH, Chit K, Aye MM, Aye KH, Aye MM, et al. Reduced susceptibility of Plasmodium falciparum to artesunate in southern Myanmar. PLoS ONE. 2013;8:e57689.PubMedPubMedCentral
45.
Zurück zum Zitat Myint MK, Rasmussen C, Thi A, Bustos D, Ringwald P, Lin K. Therapeutic efficacy and artemisinin resistance in northern Myanmar: evidence from in vivo and molecular marker studies. Malar J. 2017;16:143.PubMedPubMedCentral Myint MK, Rasmussen C, Thi A, Bustos D, Ringwald P, Lin K. Therapeutic efficacy and artemisinin resistance in northern Myanmar: evidence from in vivo and molecular marker studies. Malar J. 2017;16:143.PubMedPubMedCentral
46.
Zurück zum Zitat Nyunt MH, Hlaing T, Oo HW, Tin-Oo LL, Phway HP, Wang B, et al. Molecular assessment of artemisinin resistance markers, polymorphisms in the k13 propeller, and a multidrug-resistance gene in the eastern and western border areas of Myanmar. Clin Infect Dis. 2015;60:1208–15.PubMed Nyunt MH, Hlaing T, Oo HW, Tin-Oo LL, Phway HP, Wang B, et al. Molecular assessment of artemisinin resistance markers, polymorphisms in the k13 propeller, and a multidrug-resistance gene in the eastern and western border areas of Myanmar. Clin Infect Dis. 2015;60:1208–15.PubMed
47.
Zurück zum Zitat Nyunt MH, Soe MT, Myint HW, Oo HW, Aye MM, Han SS, et al. Clinical and molecular surveillance of artemisinin resistant falciparum malaria in Myanmar (2009–2013). Malar J. 2017;16:333.PubMedPubMedCentral Nyunt MH, Soe MT, Myint HW, Oo HW, Aye MM, Han SS, et al. Clinical and molecular surveillance of artemisinin resistant falciparum malaria in Myanmar (2009–2013). Malar J. 2017;16:333.PubMedPubMedCentral
48.
Zurück zum Zitat Tun KM, Jeeyapant A, Imwong M, Thein M, Aung SS, Hlaing TM, et al. Parasite clearance rates in Upper Myanmar indicate a distinctive artemisinin resistance phenotype: a therapeutic efficacy study. Malar J. 2016;15:185.PubMedPubMedCentral Tun KM, Jeeyapant A, Imwong M, Thein M, Aung SS, Hlaing TM, et al. Parasite clearance rates in Upper Myanmar indicate a distinctive artemisinin resistance phenotype: a therapeutic efficacy study. Malar J. 2016;15:185.PubMedPubMedCentral
49.
Zurück zum Zitat Tun KM, Jeeyapant A, Myint AH, Kyaw ZT, Dhorda M, Mukaka M, et al. Effectiveness and safety of 3 and 5 day courses of artemether-lumefantrine for the treatment of uncomplicated falciparum malaria in an area of emerging artemisinin resistance in Myanmar. Malar J. 2018;17:258.PubMedPubMedCentral Tun KM, Jeeyapant A, Myint AH, Kyaw ZT, Dhorda M, Mukaka M, et al. Effectiveness and safety of 3 and 5 day courses of artemether-lumefantrine for the treatment of uncomplicated falciparum malaria in an area of emerging artemisinin resistance in Myanmar. Malar J. 2018;17:258.PubMedPubMedCentral
50.
Zurück zum Zitat Win AA, Imwong M, Kyaw MP, Woodrow CJ, Chotivanich K, Hanboonkunupakarn B, et al. K13 mutations and pfmdr1 copy number variation in Plasmodium falciparum malaria in Myanmar. Malar J. 2016;15:110.PubMedPubMedCentral Win AA, Imwong M, Kyaw MP, Woodrow CJ, Chotivanich K, Hanboonkunupakarn B, et al. K13 mutations and pfmdr1 copy number variation in Plasmodium falciparum malaria in Myanmar. Malar J. 2016;15:110.PubMedPubMedCentral
51.
Zurück zum Zitat Liu H, Feng G, Zeng W, Li X, Bai Y, Deng S, et al. A more appropriate white blood cell count for estimating malaria parasite density in Plasmodium vivax patients in northeastern Myanmar. Acta Trop. 2016;156:152–6.PubMedPubMedCentral Liu H, Feng G, Zeng W, Li X, Bai Y, Deng S, et al. A more appropriate white blood cell count for estimating malaria parasite density in Plasmodium vivax patients in northeastern Myanmar. Acta Trop. 2016;156:152–6.PubMedPubMedCentral
52.
Zurück zum Zitat Buppan P, Putaporntip C, Pattanawong U, Seethamchai S, Jongwutiwes S. Comparative detection of Plasmodium vivax and Plasmodium falciparum DNA in saliva and urine samples from symptomatic malaria patients in a low endemic area. Malar J. 2010;9:72.PubMedPubMedCentral Buppan P, Putaporntip C, Pattanawong U, Seethamchai S, Jongwutiwes S. Comparative detection of Plasmodium vivax and Plasmodium falciparum DNA in saliva and urine samples from symptomatic malaria patients in a low endemic area. Malar J. 2010;9:72.PubMedPubMedCentral
53.
Zurück zum Zitat Johnston SP, Pieniazek NJ, Xayavong MV, Slemenda SB, Wilkins PP, da Silva AJ. PCR as a confirmatory technique for laboratory diagnosis of malaria. J Clin Microbiol. 2006;44:1087–9.PubMedPubMedCentral Johnston SP, Pieniazek NJ, Xayavong MV, Slemenda SB, Wilkins PP, da Silva AJ. PCR as a confirmatory technique for laboratory diagnosis of malaria. J Clin Microbiol. 2006;44:1087–9.PubMedPubMedCentral
54.
Zurück zum Zitat Dorsey G, Kamya MR, Singh A, Rosenthal PJ. Polymorphisms in the Plasmodium falciparum pfcrt and pfmdr-1 genes and clinical response to chloroquine in Kampala, Uganda. J Infect Dis. 2001;183:1417–20.PubMed Dorsey G, Kamya MR, Singh A, Rosenthal PJ. Polymorphisms in the Plasmodium falciparum pfcrt and pfmdr-1 genes and clinical response to chloroquine in Kampala, Uganda. J Infect Dis. 2001;183:1417–20.PubMed
55.
Zurück zum Zitat Garg S, Saxena V, Kanchan S, Sharma P, Mahajan S, Kochar D, Das A. Novel point mutations in sulfadoxine resistance genes of Plasmodium falciparum from India. Acta Trop. 2009;110:75–9.PubMed Garg S, Saxena V, Kanchan S, Sharma P, Mahajan S, Kochar D, Das A. Novel point mutations in sulfadoxine resistance genes of Plasmodium falciparum from India. Acta Trop. 2009;110:75–9.PubMed
56.
Zurück zum Zitat Gosling RD, Gesase S, Mosha JF, Carneiro I, Hashim R, Lemnge M, et al. Protective efficacy and safety of three antimalarial regimens for intermittent preventive treatment for malaria in infants: a randomised, double-blind, placebo-controlled trial. Lancet. 2009;374:1521–32.PubMed Gosling RD, Gesase S, Mosha JF, Carneiro I, Hashim R, Lemnge M, et al. Protective efficacy and safety of three antimalarial regimens for intermittent preventive treatment for malaria in infants: a randomised, double-blind, placebo-controlled trial. Lancet. 2009;374:1521–32.PubMed
57.
Zurück zum Zitat Nwe TW, Oo T, Wai KT, Zhou S, van Griensven J, Chinnakali P, et al. Malaria profiles and challenges in artemisinin resistance containment in Myanmar. Infect Dis Poverty. 2017;6:76.PubMedPubMedCentral Nwe TW, Oo T, Wai KT, Zhou S, van Griensven J, Chinnakali P, et al. Malaria profiles and challenges in artemisinin resistance containment in Myanmar. Infect Dis Poverty. 2017;6:76.PubMedPubMedCentral
58.
Zurück zum Zitat Straimer J, Gnadig NF, Witkowski B, Amaratunga C, Duru V, Ramadani AP, et al. Drug resistance. K13-propeller mutations confer artemisinin resistance in Plasmodium falciparum clinical isolates. Science. 2015;347:428–31.PubMed Straimer J, Gnadig NF, Witkowski B, Amaratunga C, Duru V, Ramadani AP, et al. Drug resistance. K13-propeller mutations confer artemisinin resistance in Plasmodium falciparum clinical isolates. Science. 2015;347:428–31.PubMed
59.
Zurück zum Zitat Zhao Y, Liu Z, Soe MT, Wang L, Soe TN, Wei H, et al. Genetic variations associated with drug resistance markers in asymptomatic Plasmodium falciparum infections in Myanmar. Genes (Basel). 2019;10. Zhao Y, Liu Z, Soe MT, Wang L, Soe TN, Wei H, et al. Genetic variations associated with drug resistance markers in asymptomatic Plasmodium falciparum infections in Myanmar. Genes (Basel). 2019;10.
60.
Zurück zum Zitat Chhibber-Goel J, Sharma A. Profiles of Kelch mutations in Plasmodium falciparum across South Asia and their implications for tracking drug resistance. Int J Parasitol Drugs Drug Resist. 2019;11:49–58.PubMedPubMedCentral Chhibber-Goel J, Sharma A. Profiles of Kelch mutations in Plasmodium falciparum across South Asia and their implications for tracking drug resistance. Int J Parasitol Drugs Drug Resist. 2019;11:49–58.PubMedPubMedCentral
61.
Zurück zum Zitat Das S, Manna S, Saha B, Hati AK, Roy S. Novel pfkelch13 gene polymorphism associates with artemisinin resistance in Eastern India. Clin Infect Dis. 2019;69:1144–52.PubMed Das S, Manna S, Saha B, Hati AK, Roy S. Novel pfkelch13 gene polymorphism associates with artemisinin resistance in Eastern India. Clin Infect Dis. 2019;69:1144–52.PubMed
62.
Zurück zum Zitat Raman J, Kagoro FM, Mabuza A, Malatje G, Reid A, Frean J, Barnes KI. Absence of kelch13 artemisinin resistance markers but strong selection for lumefantrine-tolerance molecular markers following 18 years of artemisinin-based combination therapy use in Mpumalanga Province, South Africa (2001–2018). Malar J. 2019;18:280.PubMedPubMedCentral Raman J, Kagoro FM, Mabuza A, Malatje G, Reid A, Frean J, Barnes KI. Absence of kelch13 artemisinin resistance markers but strong selection for lumefantrine-tolerance molecular markers following 18 years of artemisinin-based combination therapy use in Mpumalanga Province, South Africa (2001–2018). Malar J. 2019;18:280.PubMedPubMedCentral
63.
Zurück zum Zitat Sisowath C, Petersen I, Veiga MI, Martensson A, Premji Z, Bjorkman A, et al. In vivo selection of Plasmodium falciparum parasites carrying the chloroquine-susceptible pfcrt K76 allele after treatment with artemether-lumefantrine in Africa. J Infect Dis. 2009;199:750–7.PubMedPubMedCentral Sisowath C, Petersen I, Veiga MI, Martensson A, Premji Z, Bjorkman A, et al. In vivo selection of Plasmodium falciparum parasites carrying the chloroquine-susceptible pfcrt K76 allele after treatment with artemether-lumefantrine in Africa. J Infect Dis. 2009;199:750–7.PubMedPubMedCentral
64.
Zurück zum Zitat Htun MW, Mon NCN, Aye KM, Hlaing CM, Kyaw MP, Handayuni I, et al. Chloroquine efficacy for Plasmodium vivax in Myanmar in populations with high genetic diversity and moderate parasite gene flow. Malar J. 2017;16:281.PubMedPubMedCentral Htun MW, Mon NCN, Aye KM, Hlaing CM, Kyaw MP, Handayuni I, et al. Chloroquine efficacy for Plasmodium vivax in Myanmar in populations with high genetic diversity and moderate parasite gene flow. Malar J. 2017;16:281.PubMedPubMedCentral
65.
Zurück zum Zitat Ochong E, Bell DJ, Johnson DJ, D’Alessandro U, Mulenga M, Muangnoicharoen S, et al. Plasmodium falciparum strains harboring dihydrofolate reductase with the I164L mutation are absent in Malawi and Zambia even under antifolate drug pressure. Antimicrob Agents Chemother. 2008;52:3883–8.PubMedPubMedCentral Ochong E, Bell DJ, Johnson DJ, D’Alessandro U, Mulenga M, Muangnoicharoen S, et al. Plasmodium falciparum strains harboring dihydrofolate reductase with the I164L mutation are absent in Malawi and Zambia even under antifolate drug pressure. Antimicrob Agents Chemother. 2008;52:3883–8.PubMedPubMedCentral
66.
Zurück zum Zitat WHO. Artemisinin and artemisinin-based combination therapy resistance. Geneva: World Health Organization; 2017. WHO. Artemisinin and artemisinin-based combination therapy resistance. Geneva: World Health Organization; 2017.
Metadaten
Titel
Efficacy of artemether-lumefantrine for treating uncomplicated Plasmodium falciparum cases and molecular surveillance of drug resistance genes in Western Myanmar
verfasst von
Yanrui Wu
Myat Thut Soe
Pyae Linn Aung
Luyi Zhao
Weilin Zeng
Lynette Menezes
Zhaoqing Yang
Myat Phone Kyaw
Liwang Cui
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2020
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-020-03376-5

Weitere Artikel der Ausgabe 1/2020

Malaria Journal 1/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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