INTRODUCTION
Malaria is a mosquito-transmitted parasitic disease that occurs primarily in impoverished tropical and subtropical areas of the world. In the Greater Mekong Subregion (GMS), which consists of Cambodia, China’s Yunnan and Guangxi provinces, the Lao People’s Democratic Republic (Laos), Myanmar, Thailand, and Vietnam, malaria has been one of the most severe public health issues, hampering socioeconomic development.1–3 Recent decades have welcomed bourgeoning economic growth and significant improvement in public health in GMS countries. Driven by increasing political commitment and motivated by recent achievements in malaria control,3,4 the six GMS nations have endorsed a regional malaria elimination plan with an ultimate goal of eliminating Plasmodium falciparum malaria by 2025 and all malaria by 2030 in all countries of the GMS.5 Recently, after 3 years with no indigenous malaria cases, China was certified as malaria-free by WHO, marking a major success in the decades-long fight against this disease. However, various setbacks have been encountered in other GMS countries due to existing and emerging challenges (detailed in the following).
Malaria control and elimination rely on accurate and timely knowledge of the distribution of malaria incidence and prevalence, delivery of effective chemotherapy, and implementation of operative vector-management strategies. The complex and fast-evolving malaria epidemiology in the GMS is reflected in its immense spatial heterogeneity and the emerging dominance of Plasmodium vivax, a parasite species with remarkable resilience to conventional malaria control methods.6 In addition, artemisinin (ART) resistance in P. falciparum, detected initially in Western Cambodia a decade ago, has received augmented local and international concerns.7–10 Failure to contain ART-resistant parasites and the emergence of resistance elsewhere in the GMS escalated the urgency for a regional plan of malaria elimination.11,12 Further, the effectiveness of two core vector control interventions (insecticide-treated nets and indoor residue spraying) has been declining due to the development of insecticide resistance and increased outdoor biting of vectors.13,14 To address these problems, the Southeast Asia International Center of Excellence for Malaria Research (ICEMR) has developed a multidisciplinary program, aiming to understand how human migration, antimalarial drug resistance, and vector adaptations contribute to continuous malaria transmission at international borders so that integrative control strategies can be developed. To realize this scientific goal, we have strategically selected representative sentinel sites along the international borders of China, Myanmar, and Thailand, where malaria epidemiology is drastically different from each other. Using systems approaches and innovative technologies, we want to dissect the tripartite interactions among migrant human populations, diverse mosquito vectors, and multidrug-resistant (MDR) parasites to develop novel control strategies to propel the course of regional malaria elimination.
EPIDEMIOLOGY OF BORDER MALARIA
Spatial epidemiology.
The distribution of malaria in the GMS exhibits extreme heterogeneity at both macro and microgeographical scales.15,16 The six GMS countries have advanced to different stages of malaria elimination, with Myanmar having the highest malaria incidence (almost 70% of the regional burden). Although border malaria (concentrated malaria transmission along international borders) is a shared phenomenon of each country,17 intensified control efforts have led to isolated pockets of malaria transmission.18 In Thailand, malaria has declined over the last several decades, but pockets of malaria transmission persist along the Thai–Myanmar border (Figure 1). Of the 927 border districts, 637 (69%) reported malaria incidence in the past 3 years and 307 (33%) in 2021. Similarly, during the final phase of malaria elimination in China, malaria in the border counties of Yunnan province displayed large spatiotemporal changes with incidence clustered in several hotspot townships.16 While the P. falciparum clusters shifted locations and cluster size each year, high-incidence vivax malaria clusters persisted.16 Within villages, malaria also exhibited evident transmission hotspots, probably depending on the local ecology of vectors.19,20
Spatial distribution of malaria incidence in the border areas of Thailand, 2017–2021. The figure illustrates persistent border malaria despite the gradual reduction of annual malaria incidence. Neighboring countries and scale bar are marked in one panel. This figure appears in color at www.ajtmh.org.
Citation: The American Journal of Tropical Medicine and Hygiene 107, 4_Suppl; 10.4269/ajtmh.21-1267
Another conspicuous change in malaria epidemiology is the increasing dominance of P. vivax malaria.21,22 Surveillance of clinical malaria cases at the China–Myanmar border detected an increase in the proportion of vivax malaria from ∼60% in 2011 to > 97% in 2016, with occasional vivax malaria outbreaks.21 Such a trend has persisted in more recent years (Figure 2). The proportional increase of vivax malaria is partially attributed to its ability to relapse, which requires 14-day primaquine (PQ) radical cure, a regimen with ubiquitously poor compliance. In a cohort of 7,000 village residents on the Western Thai border, we detected 410 malaria cases by microscopy in 6.5 years. Among them, 67 people had multiple malaria episodes within 1 year of the initial infection, and 60% of these recurring infections were due to P.vivax.23 The resilience of vivax malaria to conventional malaria control measures necessitates new tools for its elimination.
Dynamics of confirmed P. vivax and P. falciparum cases from passive case detection at the Laiza township hospital in Myanmar, 2017–2021. The Inset panel shows the dominance of P. vivax. The vivax malaria outbreak in 2016–2017 was effectively suppressed by vector-based control efforts (IRS and street fumigation). Case rebounds were noticed in 2020–2021, which may be due to reduced control efforts during COVID-19. This figure appears in color at www.ajtmh.org.
Citation: The American Journal of Tropical Medicine and Hygiene 107, 4_Suppl; 10.4269/ajtmh.21-1267
Risk factors for malaria transmission.
Risk factors for malaria infection vary by parasite species, geography, and demographic attributes. Plasmodium falciparum is more geographically restricted and clusters in rural, remote areas with poor healthcare access—especially along borders. In much of the GMS, P. falciparum infection clusters in adult males who are exposed to the parasite through travel to hotspots of the disease (e.g., forested areas).21,24,25 Certain occupations (e.g., farming, military) bear a significantly higher risk of malaria while students have an increased risk of vivax malaria.21,24 Individuals with poor access to health services, with linguistic barriers, of ethnic minorities, and without citizenship may also have a higher likelihood of infection.20,21,26 For both vivax and falciparum infections, individuals who have a history of malaria infection are more likely to have subsequent infections.21,24 Housing characteristics are also related to the risk of infection, presumably associated with Anopheles permeability (open structures, building materials, distance to mosquito breeding habitats, etc.).20,27,28 In addition, housing can be a proxy for other factors like socioeconomic status, which influences occupation and access to healthcare. Identifying high-risk populations facilitates the implementation of targeted malaria control measures. Delivering health education messages to hotspot villages29 and malaria prevention packages to forest-goers and farmers staying in farm huts will help change risky behaviors and reduce malaria infection.30,31
Malaria parasite detection and surveillance.
In low-endemic malaria settings in border communities, most Plasmodium infections appear to be asymptomatic and submicroscopic,32 requiring sensitive molecular tools for detection. We have demonstrated that submicroscopic infections can infect mosquitoes,33 constituting a critical reservoir for persistent transmission. In clinical settings, malaria diagnosis is routinely performed using light microscopy and rapid diagnostic tests (RDTs). RDTs have recently gained considerable traction in the GMS and play an indispensable role in evidence-based treatment, especially in hard-to-reach remote communities along international borders, where quality microscopy is often inaccessible. As most of the RDTs deployed in the GMS for P. falciparum are based on the detection of histidine-rich protein (HRP) 2 protein, our recent findings on the emergence of parasites with pfhrp2 deletion in the Western GMS suggest potential challenges for the continued use of such RDTs.34,35 Consistent with the suboptimal performance of RDTs against nonfalciparum and nonvivax human parasite species found in Southeast Asia,36 we also demonstrated the failure of a conventional RDT to diagnose high-density (> 500 parasites/mL) acute febrile infections of Plasmodium malariae and Plasmodium ovale in the China–Myanmar border area.37
Microscopy and RDTs have limited utility for active surveillance because parasite densities in asymptomatic infections are often below their detection thresholds. Recognizing these limitations, we have conducted studies to compare potential new solutions, aiming to identify pragmatic tools for disease surveillance in the GMS. The recent advent of an ultrasensitive RDT (uRDT) for P. falciparum, having a detection limit 10 times lower than conventional RDTs, prompted the team to investigate its utility for active surveillance.38 Our study conducted in endemic areas of Myanmar demonstrated that uRDTs have approximately 20% increased sensitivity in detecting subclinical P. falciparum infections when compared with standard RDTs.39 Ultrasensitive RDTs still have lower sensitivity than molecular assays and are unlikely to identify all subclinical infections, but they are a promising improvement in our ability to monitor P. falciparum. The increasing predominance of P. vivax demands the development of uRDTs for this species.
The program evaluated several molecular diagnostics, including qPCR, nested PCR to detect parasite rRNA genes, nested reverse-transcriptase PCR (nRT-PCR) to detect parasite rRNAs, and capture and ligation-probe PCR (CLIP-PCR) to detect parasite rRNAs in cross-sectional surveys.40,41 The rRNA-based method has the highest sensitivity and rivals that of high-volume PCR,42 but the RNA detection requires a much smaller blood volume and is more suitable for active surveillance in many places. Applying nRT-PCR to finger-prick blood samples from community surveys in Northeastern Myanmar uncovered an infection prevalence of nearly 20% compared with 1% by light microscopy,27,40 further demonstrating the feasibility and the gain of using a sensitive molecular tool. As costs are one major impediment to molecular testing, a simple and flexible method of sample pooling was devised,39 which can be tailored to different endemicities. As most infections in areas approaching elimination are asymptomatic and submicroscopic,33 molecular surveillance in sentinel sites is essential for guiding targeted control practices, determining the effects of control measures, and monitoring the progress toward elimination. Further fine-tuning these molecular tools to differentiate the drug resistant and sensitive parasites in a clinical setting would also be crucial for timely adjusting drug policies.
Migration and malaria introduction.
Border malaria poses a vital threat to malaria elimination and requires multinational cooperation.43 Heavy population flow along the extremely porous borders makes neighboring countries vulnerable to malaria introduction and reintroduction.44,45 Human migration may be partially responsible for the cross-national spread of ART-resistant strains with specific multidrug resistance genotypes.46 The association of a higher risk of malaria with the migrant population and those with travel to Myanmar highlights the significance of malaria introduction by migratory populations in the border region.20,47,48 Although passive case detection activity in the Southwestern border of China only showed strong evidence of imported P. falciparum malaria,47 subsequent genetic studies at the China–Myanmar border using microsatellite markers revealed genetically homogenous populations for both parasite species on both sides, indicating extensive parasite gene flow not constrained by the political border.49,50 Analysis of parasite migration patterns within and between the two sides of the international border detected unidirectional migration of parasites from Myanmar to China, providing genetic evidence of parasite migration in the border region. Especially for P. vivax, a parasite that can travel long distances by infected migrants as silent liver hypnozoites, there is an urgency to identify the sources and sinks of the parasites to enable timely targeted control. The use of polymorphic antigen markers such as Pvmsp3α and 3β has revealed highly diverse P. vivax populations in Western Thailand border despite low endemicity, and detected clonal expansion events in Southern Thailand, likely resulting from relaxed control efforts.51–53 Using microsatellite markers, we found drastically divergent P. vivax populations in the Eastern and Western Thailand borders, with the central malaria-free zone as a gene flow barrier.54 The possibility to distinguish these parasite populations using as few as four microsatellite markers will simplify the tracking of parasite migration, at least among the Thailand borders. We also found that microsatellites could be used to assess the temporal population changes as a means to monitor the progress of malaria control. Although the genetic diversity of P. vivax populations over time may remain high, the decreased multiplicity of infection and increased multilocus linkage disequilibrium may reflect a reduction in the parasite population size.55 In Eastern GMS, where P. vivax populations are less geographically isolated and genetically distinct, whole-genome sequencing (WGS) and the derived SNP barcode may be necessary to distinguish closely related parasite strains and identify the origins of the parasite.56,57 The genomic information from spatially representative parasite populations would identify potential migration patterns using shared identity-by-descent segments,56,58 providing the scientific basis for enhanced monitoring of parasite introduction by migrant populations.
Zoonotic Plasmodium knowlesi malaria.
Since the first cluster of Plasmodium knowlesi malaria cases in humans was reported in 2004 in Malaysian Borneo,59 reports of P. knowlesi incidence have increased strikingly, including in all countries of the GMS—Thailand,60–64 Laos,65,66 Cambodia,67 Myanmar,68,69 and Vietnam.65,70,71 This wide range of P. knowlesi in Southeast Asia largely reflects the distribution of the zoonotic hosts (the long-tailed and pig-tailed macaques) and vectors of the Leucosphyrus group of anopheline mosquitoes.72,73 This parasite is probably historically present in the GMS rather than newly emergent. In recent years, we and others have identified an increasing trend of clinical P. knowlesi cases in Thailand.63,64 Increased incidences of P. knowlesi are likely due to environmental changes such as deforestation, increased forest-related human activities, and potentially peridomestic transmission.74 Plasmodium knowlesi diagnosis is challenging75—it is often misdiagnosed by microscopy due to its resemblance to P. malariae and P. falciparum, current RDTs are not sufficiently sensitive to detect P. knowlesi, and confirmation requires the use of molecular methods.61,76 Its presence as coinfections with other human malaria parasites and in asymptomatic infections also complicates diagnosis and detection, resulting in an underestimate of its real burden.61,65,68,69,71,77,78 Since the regional malaria elimination efforts are meant to target all Plasmodium species,79 it is also time to consider eliminating P. knowlesi and other monkey malaria parasites infecting humans (P. cynomolgi, P. inui, etc.).80–82 The diverse factors associated with the transmission of these zoonotic malaria parasites present a challenge for their elimination, as conventional vector-based control efforts in the domestic environment are ineffective in protecting against sylvatic transmission. Strategies such as repellent and chemoprophylaxis targeting high-risk populations like forest-goers are advocated to accelerate malaria transmission in the GMS.31
MOSQUITO ECOLOGY AND INSECTICIDE RESISTANCE
Vector ecology.
Malaria vectors in the GMS consist of many Anopheles species with varying abundance and importance in malaria transmission among different geographical regions.83,84 Many vector species are in species complexes, including several morphologically similar species and possibly cryptic species. The abundance, diversity, distribution, survivorship, biting behaviors, and vectorial status of different vectors can be influenced by environmental changes, such as deforestation and extensive use of insecticides in both public health and agricultural sectors. As “forest malaria” is a major contributor to residual malaria incidence,85,86 deforestation and landscape changes will have a significant impact on vector ecology and malaria transmission.87 Our study conducted in the China–Myanmar border area showed that adult An. sinensis and An. minimus, the main malaria vectors in this region, had much higher survivorship in deforested than forested areas.88 Deforestation also enhanced the survival of An. minimus larvae and accelerated larval development.89 Our vector surveillance studies conducted in sentinel sites of China, Myanmar, and Thailand have detected major changes in Anopheles composition and seasonal dynamics (Table 1). An. minimus was the predominant vector in all the surveys.30,90–92 Consistent with An. minimus being a highly adaptive vector, population genetic analysis revealed similar population genetic structure of past and present An. minimus populations and substantial gene flow among different geographical populations.93 These studies also revealed increased abundance of other vectors such as An. annularis and An. barbirostris s.l., some of which may support outdoor transmission.30,90–92 The vector species composition is further complicated by the presence of morphologically identical cryptic species. In Western Thailand, An. minimus A and An. harrisoni (An. minimus C) are two cryptic species often found in the same locations.94 Our recent molecular studies of An. minimus species collected from Western Thailand showed that ∼11% of the morphologically identified An. minimus belonged to a cryptic species (lineage B), which deserves further investigation to understand its bionomics, vectorial status, and species evolution.93
Anopheles species compositions in different study sites and study periods at the international borders of the GMS
Species | China–Myanmar border (2012–2014)a | Tak, Thailand (2011–2013)b | Tak, Thailand (2015)c | |||
---|---|---|---|---|---|---|
N | % | N | % | N | % | |
An. minimus | 13,038 | 84.6 | 1,204 | 40.3 | 3,725 | 49.5 |
An. maculatus | 530 | 3.4 | 640 | 21.4 | 999 | 13.3 |
An. culicifacies | 437 | 2.8 | 51 | 1.7 | 1054 | 14.0 |
An. vagus | 220 | 1.4 | 13 | 0.4 | 38 | 0.5 |
An. sinensis | 161 | 1.0 | 1 | – | – | – |
An. barbirostris | 133 | 0.9 | 105 | 3.5 | 185 | 2.5 |
An. paeditaeniatus | 127 | 0.8 | 63 | 2.1 | 102 | 1.4 |
An. kochi | 39 | 0.3 | 161 | 5.4 | 41 | 0.6 |
An. tessellatus | 39 | 0.3 | 157 | 5.3 | 97 | 1.3 |
An. annularis | 7 | 0.0 | 431 | 14.4 | 851 | 11.3 |
An. jeypariensis | 277 | 1.8 | – | – | – | – |
An. splendidus | 237 | 1.5 | – | – | – | – |
An. varuna | – | – | 41 | 1.4 | 3 | 0.0 |
An. sawadwongporni | – | – | 1 | 0.0 | 293 | 3.9 |
Other Anopheles | 175 | 1.1 | 118 | 4.0 | 133 | 1.8 |
Total | 15,410 | 100 | 2,986 | 100 | 7,519 | 100 |
Mosquitoes were collected by CDC light traps. This table illustrates major changes of primary vector species in different sentinel sites. Anopheles species with ≥ 1% abundance were listed by species names, while the rest was summarized as “Other Anopheles.”
In addition to changes in vector species, the malaria vectors in the GMS showed different levels of adaptations to the microecology with dramatic variations among villages. Their different seasonal dynamics underlie their roles in malaria transmission in different seasons.90–92 Residual malaria transmission was traced to farm huts and outdoor agriculture sites, where human biting rates were the highest with An. minimus, An. dirus, and An. maculatus as the primary vectors.30 In Western Thailand, An. minimus and An. maculatus are the main vectors during the two annual malaria transmission peaks while An. minimus group is the key primary vector in the dry season,94 the Maculatus group is most abundant in the wet season with within-group species-specific variations.91 Collectively, this knowledge of the species composition, distribution, bionomics, and dynamics in the international border regions is needed to guide vector control efforts.
Extent, distribution, and mechanisms of insecticide resistance.
Fast emerging and increasing insecticide resistance of malaria vectors has been implicated as a significant threat to malaria prevention by vector control. Understanding the status, distribution, and mechanisms of insecticide resistance in local malaria vector populations is critical for resistance management and effective malaria control and elimination. We have been monitoring the resistance of malaria vectors to multiple insecticides using the WHO tube test in multiple study sites in China, Thailand, and Myanmar since 2011. The two best-known resistance mechanisms (target site resistance and metabolic detoxification) were investigated in field populations of Anopheles mosquitoes. High-level resistance to the four major classes of insecticides (pyrethroids, organochlorines, organophosphates, and carbamates) was observed in An. sinenesis populations from Southern and Central China,95–97 and the Eastern Coastal region of China.98 Three nonsynonymous knockdown resistance (kdr) mutations (L1014F, L1014C, and L1014S) were detected at codon L1014 of the para-type sodium channel gene in An. sinensis from China, and these kdr mutation alleles exhibited a patchy distribution in frequency from Southern to Central China. Near fixation of kdr mutation was detected in populations from Central China but no kdr mutations were found in Southwestern China, suggesting that kdr alone is insufficient to predict pyrethroid resistance.99 The G119S mutation of the ace-1 gene in An. sinensis was moderately frequent in Southern and Central China but fixed in the Eastern Coastal region of China.96–98 Recently, high-level resistance to deltamethrin (mortality rate, 40–80%) was observed in multiple Anopheles species, including An. minimus s.l. from Thailand in 2018, and the two major vector species complexes (An. hyrcanus s.l. and An. barbirostris s.l.) from Myanmar in 2019 (unpublished data). However, the kdr L1014 mutations or the ace-1 G119S mutation were not detected in any of the Anopheles species analyzed from Thailand and Myanmar, suggesting other mechanisms responsible for pyrethroid and organophosphate resistance (unpublished data). The classification and statistical regression analysis found that metabolic detoxification was the most important resistance mechanism, whereas target site insensitivity of L1014 kdr mutation played a less critical role.96 We have used transcriptome and WGS to identify transcripts and SNPs associated with insecticide resistance.100,101 These studies highlight the complex network of mechanisms conferring resistance to multiple chemical insecticides in mosquito vectors, and it has important implications for designing and implementing improved vector resistance management strategies.
ANTIMALARIAL DRUG RESISTANCE
ART-based combination therapies (ACTs) are the frontline treatment of P. falciparum and are also recommended as a unified treatment of P. vivax. The emergence of P. falciparum parasites resistant to ART and partner drugs significantly compromised the efficacies of two ACTs—artesunate-mefloquine (AS-MQ) and dihydroartemisinin-piperaquine (DHA-PPQ).102–107 Clinical ART resistance is manifested as delayed parasite clearance with a parasite clearance half-life of > 5.5 hours, compared with ∼2 hours typically associated with ART-sensitive parasites.8,108–110 Day-3 blood smear parasite-positivity is also a crude measure of ART resistance, with a 10% cutoff for suspected ART resistance.111,112 In vitro, ART resistance is measured by the ring-stage survival assay (RSA), which measures the survival rate of early ring-stage parasites exposed to a 6-hour pulse of 700 nM of DHA, with an RSA value of ≥ 1% considered as an indication of ART resistance.113 Genetically, mutations in the propeller domain of the Kelch-domain protein K13 were identified as the key determinants of ART resistance.114 Of the > 200 PfK13 mutations identified in the global parasite populations,115,116 many have been confirmed in clinical efficacy studies111,117,118 while some have been validated genetically for in vitro ART resistance.119–122
Monitoring clinical efficacy of ACTs in Western GMS.
We have focused our efforts on monitoring the emergence and spread of ART resistance in Myanmar, given its disproportionate malaria burden in the GMS and its bridging position with South Asia. In Northeastern Myanmar bordering China, the evaluation of DHA-PPQ in 71 patients with uncomplicated falciparum malaria in 2012–2013 demonstrated a 42-day cure rate of 100% and a day-3 parasite-positive rate of 7%.123 Similarly, we also found a 28-day cure rate of 100% for artemether-lumefantrine in 41 falciparum patients at the Western border of Myanmar in 2015, although the day-3 positivity rate exceeded 10% in the latter study.124 Assessment of 44 culture-adapted clinical isolates for RSA demonstrated increased ring survival rates in parasites with PfK13 mutations.125 In addition, day-3 parasite-positive isolates had ∼10 times higher RSA values than day-3 parasite-negative isolates. These studies set the stage for using in vivo efficacy study, in vitro RSA, and molecular surveillance as complementary approaches to monitoring ART resistance.
Longitudinal in vitro drug susceptibility and molecular markers of resistance.
Our efforts over the past decade to procure clinical isolates from the China–Myanmar border area and establish continuous culture have allowed us to follow the dynamics of in vitro drug susceptibility longitudinally.126–129 From these studies, in vitro sensitivities to 4-aminoquinolines, antifolates, and ARTs deserve some attention. Although chloroquine (CQ) has been withdrawn from treating P. falciparum malaria for some time, CQ resistance is consistently high, corresponding with the prevailing occurrence of the Dd2-like pfcrt genotype, the primary determinant of CQ resistance.126,128 The use of CQ as the frontline treatment of P. vivax malaria may have continually exerted collateral selection pressure on the sympatric P. falciparum. Similarly, although the antifolate drugs were withdrawn quite some time ago, parasites exhibited continuous increases in resistance to pyrimethamine, and major mutations in the pfdhfr and pfdhps genes mediating antifolate resistance remain highly prevalent.126,128 The drug that replaced CQ in this region is PPQ monotherapy,130 also the partner drug for the commonly used ACT, DHA-PPQ. Despite previous reports of clinical resistance to PPQ and identification of pfcrt mutations, which may be associated with PPQ resistance,131,132 recent studies showed that the efficacy of DHA-PPQ for uncomplicated P. falciparum malaria remained high.123,133 Parasites collected over the years were relatively susceptible to PPQ with temporal fluctuations in IC50 or IC90.129 We did not observe parasites with either plasmepsin 2/3 amplification or new pfcrt mutations (H97Y, F145I, M343L, and G353V),129 which were described in the DHA-PPQ-resistant populations in Cambodia.134–138
PfK13-mediated and non-PfK13 ART resistance mechanisms.
PfK13 mutations have also experienced drastic spatiotemporal changes in the GMS. In the Eastern GMS, the C580Y mutation was predominant and has swept rapidly across Cambodia and the Eastern GMS.114,115,122 In the Western GMS, the F446I mutation is the most prevalent.139–141 Table 2 summarizes the results from our molecular surveillance of PfK13 mutations in the Western GMS. An updated distribution map of major PfK13 mutations in endemic sites of the GMS is shown in Figure 3. On the Eastern border of Myanmar, F446I has gained a steady increase in prevalence between 2007 and 2013.127,140 In the 2014–2016 samples, the G533S mutation emerged and became the second most prevalent at 44%. This new mutation was associated with increased RSA values.127 Analysis of asymptomatic P. falciparum infections from cross-sectional surveys conducted in the Eastern, Northern, and Western border areas of Myanmar during 2015–2018 detected the F446I mutation only on the Eastern border, suggesting that ART resistance has not spread to or emerged in the Western and Northern borders (Table 2).142 To determine whether the PfK13 mutations found in the Western GMS indeed confer ART resistance in vitro, we engineered the F446I, N458Y, C469Y, F495L, and C580Y mutations in the 3D7 background and confirmed that the N458Y and C580Y mutations conferred significant increases in ring survival rate.121 Conversely, reverting the F446I, N458Y, C469Y, and C580Y mutations to the wild type (WT) in field isolates resulted in significant decreases in RSA values in all except for the C469Y mutation. Although all tested PfK13 mutations incurred different levels of fitness cost in the transgenic parasites, the F446I and C580Y mutations were almost as fit as the WT,121 which may explain their high prevalences in the field parasite populations. In addition, transgenic parasites with these two mutations also exhibited a prolonged ring stage, presumably enabling the parasites to better survive ART treatment, which has a short half-life.
Amino acid substitutions detected in the PfK13 gene of P. falciparum populations at different border areas of Myanmar
Mutation | China–Myanmar border (and East Myanmar) | Banmauk, North Myanmar | Paletwa, West Myanmar | ||
---|---|---|---|---|---|
2007–2012 (N = 191)a | (2013–2016) (N = 74)b | 2017–2018 (N = 53)c | 2015 (N = 30)d | 2017 (N = 22)c | |
N11Y | 1 (0.5) | – | – | – | – |
K189T | 3 (1.6) | 2 (2.7) | 4 (17.4) | 3 (10.0) | – |
E252Q | 1 (0.5) | – | – | – | – |
R255K | 1 (0.5) | – | – | – | – |
I352T | 1 (0.5) | – | – | – | – |
I376V | 2 (1.0) | – | – | – | – |
P441L | 1 (0.5) | – | – | – | – |
P443S | 1 (0.5) | – | – | – | – |
F446I | 52 (27.2) | 44 (59.5) | – | – | – |
N458Y | 1 (0.5) | 1 (1.4) | – | – | – |
C469Y | 2 (1.0) | – | – | – | – |
L492S | 1 (0.5) | – | – | – | – |
F495L | 2 (1.0) | – | – | – | – |
G533S | – | 15 (20.3) | – | – | – |
P574L | 12 (6.3) | – | – | – | – |
C580Y | 3 (1.6) | – | – | – | – |
A676D | 2 (1.0) | – | – | – | – |
H719N | 2 (1.0) | – | – | – | – |
Total | 88 (46.1) | 62 (83.8) | 4 (17.4) | 3 (10.0) | 0 (0) |
Map of the GMS showing the prevalence and distribution of major PfK13 mutations as pie graphs above the malaria incidence heatmap of the region in 2020. The PfK13 mutation status was updated using malaria parasites collected primarily in 2016 from Cambodia,122 Laos,192 Vietnam,193 Thailand,194,195 and Myanmar.124,127,142 This figure appears in color at www.ajtmh.org.
Citation: The American Journal of Tropical Medicine and Hygiene 107, 4_Suppl; 10.4269/ajtmh.21-1267
Investigations into the PfK13-mediated ART resistance mechanisms suggest the involvement of heme-facilitated ART activation and oxidative stress responses.143–146 Since heme is an abundant product of hemoglobin digestion, reduced hemoglobin uptake and digestion would lower ART activation and increase ART resistance. Mutations in the hemoglobinase falcipain 2a resulting from in vitro ART selection suggest its involvement in ART resistance.114,147 Falcipain 2a harbors geographically divergent mutations, a likely result of drug selection. Analysis of falcipain 2a mutation haplotypes in field isolates from the China–Myanmar border area showed that some mutations might reduce the enzyme activity, resulting in increased ART resistance.148 Future studies using isogenic parasite lines will better define the role of falcipain 2a in mediating ART resistance.
ART-resistant parasites carrying the WT PfK13 allele have inspired research on additional players in ART resistance.149,150 The archived clinical isolates have provided the opportunity to perform detailed in vitro studies and uncover the genetic determinants of resistance. Genome-wide association studies (GWAS) of P. falciparum isolates collected from the China–Myanmar border area allowed us to identify mutations in genes from multiple pathways such as autophagy (ATG18) and DNA repair (Rad5) to be associated with increased ART resistance in field isolates.151 Further probing into these pathways will determine whether they are directly responsible for ART resistance or constitute background mutations ameliorating fitness costs resulting from causal mutations.118
VIVAX MALARIA TRANSMISSION
As P. falciparum incidence declines, eliminating vivax malaria is a major challenge for the “last mile” of malaria elimination in the GMS. The unique biology of P. vivax—hypnozoite formation responsible for relapses, early gametocytogenesis enabling transmission before clinical symptoms, and invasion of reticulocytes resulting in low parasitemia—underlies the resilience of this parasite to conventional malaria control measures. In addition, host genetics, drug resistance of the parasite, and changing vector species and populations may also contribute to the persistence and increasing predominance of this parasite.152
Implementation of radical cure.
Currently, PQ is the only drug approved for radical cure of vivax malaria in this region. However, PQ is under-prescribed because it can cause acute hemolytic anemia in patients with glucose-6-dehydrogenase (G6PD) deficiency. In the Kachin ethnicity of Northeast Myanmar, G6PD deficiency reached > 20% prevalence153 but patients are not screened routinely for G6PD status before initiating treatment of P. vivax malaria. In Thailand and Myanmar, the Mahidol variant (487G>A) is the most predominant and often accounts for ∼90% of all mutations.153–156 The Mahidol variant is associated with different levels of protection against vivax malaria.157,158 Although it is classified as a mild-deficient variant with 30–60% enzyme activity,159 some patients with this variant could have < 1% of the normal G6PD activity.155,160–162 Thus, PQ administration in these patients carries a significant risk of severe hemolysis. We have documented a clinical case of severe hemolysis in a vivax patient after receiving a 3-day low dose PQ (0.25 mg/kg/day) that required blood transfusion.163 In Northeast Myanmar, where the G6PD Mahidol variant is prevalent, we conducted an observational study in a cohort of 152 vivax patients to follow the risk of acute hemolysis after treatment with the standard CQ and 14-day PQ regimen. We found that almost 1/3 of the patients experienced clinically concerning declines in hemoglobin, with five requiring blood transfusion (unpublished). Risk in this area is likely exacerbated by preexisting anemia due to host genetics, such as thalassemia and hemoglobin E and other factors, such as helminth infections and poor nutrition.162 Thus, the standard 14-day PQ regimen carries a significant risk of acute hemolytic anemia in vivax patients without G6PD testing in Northeast Myanmar.
Another host factor that affects the effectiveness of PQ for radical cure of vivax malaria is the hepatic enzyme cytochrome P450 (CYP) 2D6,164 which mediates activation of PQ to its active metabolite(s).165,166 In clinical trials to assess the effectiveness of PQ for preventing relapses, treatment failures were associated with impaired CYP2D6 function.167–169 In Southern China, where malaria has recently been eliminated, malaria importation is a concern, and relapsing malaria has steadily increased in the proportion of the imported cases.170,171 We identified a clinical P. vivax case with multiple relapses, potentially due to poor metabolism of the CYP2D6 enzyme.172 Thus, the knowledge of the prevalence of low metabolizer CYP2D6 variants in a population is a prerequisite for planning large-scale PQ administration for vivax malaria elimination.
Chloroquine efficacy and drug resistance.
Chloroquine remains the mainstay treatment of blood-stage P. vivax infections in the GMS, though a unified ACT for both P. falciparum and P. vivax is advocated.173 Clinical failures of CQ treatment of vivax malaria are reported sporadically in the GMS,174–177 making efficacy monitoring imperative. Our studies in Northeastern Myanmar also detected declining efficacy of CQ for vivax malaria, with the detection of cases where CQ/PQ treatment failed to clear parasitemia within 7 days, suggesting high-grade resistance.178,179 Given the schizonticidal activity of PQ, these studies may underestimate the CQ resistance status. We also monitored the susceptibilities of the P. vivax clinical isolates to a panel of commonly used antimalarials using an ex vivo assay from 2012 to 2016.180 For CQ, parasites displayed a wide range of susceptibility, including > 10% parasites with IC50 values exceeding 220 nM, a cutoff value used to define CQ resistance.181,182 Only the median IC50 values for pyronaridine had an increasing trend from 2.9 in 2012–2013 to 15.5 nM in 2016.181,182 The latter value was much greater than that reported for P. vivax parasites from Papua, Indonesia.183
To date, the molecular mechanism of P. vivax CQ resistance remains unknown. Studies have focused on pvmdr1 gene, which has geographically divergent nonsynonymous SNPs.184 Within the GMS, pvmdr1 also showed a significant spatial difference in the prevalence of mutations.185–188 The pvmdr1 Y976F mutation was highly prevalent in Cambodian parasites but was either absent or less frequent in samples from Thailand, the China–Myanmar border region, and Myanmar.187–191 Longitudinal molecular surveillance at the China–Myanmar border showed that the Y976F and F1076L prevalences showed an opposite trend.186 The Y976F mutation was present at a moderate frequency of 18.5% in 2008 but sharply decreased to 1.5% in 2012 and completely disappeared in 2015. In contrast, the F1076L mutation continually increased from 33.3% in 2008 to 41.7% in 2012–2013 and 77.8% in 2015. However, we did not detect an association between these two mutations with CQ resistance.180,182 With evidence of the emerging CQ resistance in this region, it is imperative to continuously monitor in vivo and ex vivo CQ sensitivities, coupling these with genetic studies such as GWAS to elucidate the resistance mechanisms.
CONCLUSION AND FUTURE WORK
With the scale-up of malaria control efforts in the GMS regional malaria elimination campaign, malaria epidemiology has experienced drastic changes with varying degrees of reduction in malaria incidence in the regional countries. Regardless, border regions continue to have persistent malaria transmission, with cross-border introduction constituting a critical threat to malaria elimination. Rigorous surveillance of malaria in border townships needs to be maintained so that real-time information can guide the implementation of existing and new elimination strategies. Vector control measures effectively suppress malaria outbreaks and need to be implemented or strengthened in high-incidence areas. These measures must be regularly adjusted in response to changing prevalence and behaviors of primary vector species and resistance to popular pesticides. There must also be close phenotypic and molecular monitoring of insecticide resistance, especially in areas where insecticide resistance is emerging, such as Western Thailand. Although P. falciparum malaria incidence continues to decline, and there is no indication of the emergence or spread of ART-resistant parasites in the Western GMS, continuous studies are still required to identify novel resistance mechanisms, determine ACT efficacy, and monitor the spread of ART resistance. The dominant status of P. vivax requires the development and implementation of effective control measures, such as mass drug administration of PQ in combination with mass screening for G6PD deficiency to eliminate the liver stages. New strategies also need to be implemented to determine the burden of zoonotic malaria, understand the ecology of transmission, and identify the high-risk population for targeted prevention. In sum, continued research will help develop integrated tools for countries to move from low or very low endemicity to complete elimination.
ACKNOWLEDGMENTS
We would like to thank the staff at our malaria research stations along the international borders of Thailand, China, and Myanmar for technical support. We also thank the patients and their guardians for their participation in clinical studies.
REFERENCES
- 1.↑
Delacollette C et al.2009. Malaria trends and challenges in the Greater Mekong Subregion. Southeast Asian J Trop Med Public Health 40: 674–691.
- 2.↑
Cui L et al.2012. Malaria in the Greater Mekong Subregion: heterogeneity and complexity. Acta Trop 121: 227–239.
- 3.↑
Hewitt S , Delacollette C , Chavez I , 2013. Malaria situation in the Greater Mekong subregion. Southeast Asian J Trop Med Public Health 44 (Suppl 1):46–72, discussion 306–307.
- 4.↑
WHO , 2011. Roll Back Malaria: Eliminating Malaria: Learning from the Past, Looking Ahead, Number 8. World Health Organization, Geneva, Switzerland: Progress & Impact Series.
- 5.↑
WHO , 2015. Strategy for Malaria Elimination in the Greater Mekong Subregion (2015–2030), World Health Organization, Geneva, Switzerland, 64.
- 6.↑
WHO , 2015. Control and Elimination of Plasmodium Vivax Malaria—A Technical Brief. Available at: https://www.who.int/malaria/publications/atoz/9789241509244/en/.
- 7.↑
Dondorp AM et al.2009. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 361: 455–467.
- 8.↑
Amaratunga C et al.2012. Artemisinin-resistant Plasmodium falciparum in Pursat Province, Western Cambodia: a parasite clearance rate study. Lancet Infect Dis 12: 851–858.
- 9.↑
Noedl H , Se Y , Schaecher K , Smith BL , Socheat D , Fukuda MM , 2008. Evidence of artemisinin-resistant malaria in Western Cambodia. N Engl J Med 359: 2619–2620.
- 10.↑
Noedl H et al.2010. Artemisinin resistance in Cambodia: a clinical trial designed to address an emerging problem in Southeast Asia. Clin Infect Dis 51: e82–e89.
- 11.↑
Smith Gueye C , Newby G , Hwang J , Phillips AA , Whittaker M , MacArthur JR , Gosling RD , Feachem RG , 2014. The challenge of artemisinin resistance can only be met by eliminating Plasmodium falciparum malaria across the Greater Mekong Subregion. Malar J 13: 286.
- 13.↑
Tananchai C , Pattanakul M , Nararak J , Sinou V , Manguin S , Chareonviriyaphap T , 2019. Diversity and biting patterns of Anopheles species in a malaria endemic area, Umphang Valley, Tak Province, Western Thailand. Acta Trop 190: 183–192.
- 14.↑
Chaumeau V , Cerqueira D , Zadrozny J , Kittiphanakun P , Andolina C , Chareonviriyaphap T , Nosten F , Corbel V , 2017. Insecticide resistance in malaria vectors along the Thailand–Myanmar border. Parasit Vectors 10: 165.
- 15.↑
Cui L , Cao Y , Kaewkungwal J , Khamsiriwatchara A , Lawpoolsri S , Soe TN , Kyaw MK , Sattabongkot J , 2018. Malaria elimination in the Greater Mekong Subregion: challenges and prospects. Manguin S, Dev V, eds. Towards Malaria Elimination: A Leap Forward. IntechOpen, London, UK. 179–200.
- 16.↑
Xu X , Zhou G , Wang Y , Hu Y , Ruan Y , Fan Q , Yang Z , Yan G , Cui L , 2016. Microgeographic heterogeneity of border malaria during elimination phase, Yunnan Province, China, 2011–2013. Emerg Infect Dis 22: 1363–1370.
- 17.↑
Parker DM , Carrara VI , Pukrittayakamee S , McGready R , Nosten FH , 2015. Malaria ecology along the Thailand–Myanmar border. Malar J 14: 388.
- 18.↑
Zeng W , Bai Y , Wang M , Wang Z , Deng S , Ruan Y , Feng S , Yang Z , Cui L , 2017. Significant divergence in sensitivity to antimalarial drugs between neighboring Plasmodium falciparum populations along the eastern border of Myanmar. Antimicrob Agents Chemother 61: e01689–e16.
- 19.↑
Hu Y et al.2016. Seasonal dynamics and microgeographical spatial heterogeneity of malaria along the China–Myanmar border. Acta Trop 157: 12–19.
- 20.↑
Parker DM et al.2015. Microgeography and molecular epidemiology of malaria at the Thailand–Myanmar border in the malaria pre-elimination phase. Malar J 14: 198.
- 21.↑
Geng J et al.2019. Increasing trends of malaria in a border area of the Greater Mekong Subregion. Malar J 18: 309.
- 22.↑
Mercado CEG et al.2019. Spatiotemporal epidemiology, environmental correlates, and demography of malaria in Tak Province, Thailand (2012–2015). Malar J 18: 240.
- 23.↑
Lawpoolsri S , Sattabongkot J , Sirichaisinthop J , Cui L , Kiattibutr K , Rachaphaew N , Suk-Uam K , Khamsiriwatchara A , Kaewkungwal J , 2019. Epidemiological profiles of recurrent malaria episodes in an endemic area along the Thailand–Myanmar border: a prospective cohort study. Malar J 18: 124.
- 24.↑
Li N et al.2013. Risk factors associated with slide positivity among febrile patients in a conflict zone of North-Eastern Myanmar along the China–Myanmar border. Malar J 12: 361.
- 25.↑
Saita S , Silawan T , Parker DM , Sriwichai P , Phuanukoonnon S , Sudathip P , Maude RJ , White LJ , Pan-Ngum W , 2019. Spatial heterogeneity and temporal trends in malaria on the Thai–Myanmar Border (2012–2017): a retrospective observational study. Trop Med Infect Dis 4: 62.
- 26.↑
Parker DM , Wood JW , Tomita S , DeWitte S , Jennings J , Cui L , 2014. Household ecology and out-migration among ethnic Karen along the Thai–Myanmar border. Demogr Res 30: 1129–1156.
- 27.↑
Zhao Y , Zeng J , Liu Q , He Y , Zhang J , Yang Z , Fan Q , Wang Q , Cui L , Cao Y , 2018. Risk factors for asymptomatic malaria infections from seasonal cross-sectional surveys along the China–Myanmar border. Malar J 17: 247.
- 28.↑
Aung PL , Soe MT , Oo TL , Khin A , Thi A , Zhao Y , Cao Y , Cui L , Kyaw MP , Parker DM , 2021. Predictors of malaria rapid diagnostic test positivity in a high burden area of Paletwa Township, Chin State in Western Myanmar. Infect Dis Poverty 10: 6.
- 29.↑
Aung PL , Pumpaibool T , Soe TN , Burgess J , Menezes LJ , Kyaw MP , Cui L , 2019. Health education through mass media announcements by loudspeakers about malaria care: prevention and practice among people living in a malaria endemic area of Northern Myanmar. Malar J 18: 362.
- 30.↑
Edwards HM , Sriwichai P , Kirabittir K , Prachumsri J , Chavez IF , Hii J , 2019. Transmission risk beyond the village: entomological and human factors contributing to residual malaria transmission in an area approaching malaria elimination on the Thailand–Myanmar border. Malar J 18: 221.
- 31.↑
Nofal SD , Peto TJ , Adhikari B , Tripura R , Callery J , Bui TM , von Seidlein L , Pell C , 2019. How can interventions that target forest-goers be tailored to accelerate malaria elimination in the Greater Mekong Subregion? A systematic review of the qualitative literature. Malar J 18: 32.
- 32.↑
Baum E et al.2016. Common asymptomatic and submicroscopic malaria infections in Western Thailand revealed in longitudinal molecular and serological studies: a challenge to malaria elimination. Malar J 15: 333.
- 33.↑
Kiattibutr K et al.2017. Infectivity of symptomatic and asymptomatic Plasmodium vivax infections to a Southeast Asian vector, Anopheles dirus. Int J Parasitol 47: 163–170.
- 34.↑
Li P , Xing H , Zhao Z , Yang Z , Cao Y , Li W , Yan G , Sattabongkot J , Cui L , Fan Q , 2015. Genetic diversity of Plasmodium falciparum histidine-rich protein 2 in the China–Myanmar border area. Acta Trop 152: 26–31.
- 35.↑
Gibbons J et al.2020. Lineage-specific expansion of Plasmodium falciparum parasites with pfhrp2 deletion in the Greater Mekong subregion. J Infect Dis 222: 1561–1569.
- 36.↑
Yerlikaya S , Campillo A , Gonzalez IJ , 2018. A systematic review: performance of rapid diagnostic tests for the detection of Plasmodium knowlesi, Plasmodium malariae, and Plasmodium ovale monoinfections in human blood. J Infect Dis 218: 265–276.
- 37.↑
Shikur B , Deressa W , Lindtjorn B , 2016. Association between malaria and malnutrition among children aged under-five years in Adami Tulu District, South-Central Ethiopia: a case-control study. BMC Public Health 16: 174.
- 38.↑
Das S , Peck RB , Barney R , Jang IK , Kahn M , Zhu M , Domingo GJ , 2018. Performance of an ultra-sensitive Plasmodium falciparum HRP2-based rapid diagnostic test with recombinant HRP2, culture parasites, and archived whole blood samples. Malar J 17: 118.
- 39.↑
Liu Z et al.2019. Geographical heterogeneity in prevalence of subclinical malaria infections at sentinel endemic sites of Myanmar. Parasit Vectors 12: 83.
- 40.↑
Zhao Y et al.2017. Comparison of methods for detecting asymptomatic malaria infections in the China–Myanmar border area. Malar J 16: 159.
- 41.↑
Baum E et al.2015. Submicroscopic and asymptomatic Plasmodium falciparum and Plasmodium vivax infections are common in Western Thailand—molecular and serological evidence. Malar J 14: 95.
- 42.↑
Imwong M , Hanchana S , Malleret B , Renia L , Day NP , Dondorp A , Nosten F , Snounou G , White NJ , 2014. High-throughput ultrasensitive molecular techniques for quantifying low-density malaria parasitemias. J Clin Microbiol 52: 3303–3309.
- 43.↑
Sturrock HJ , Roberts KW , Wegbreit J , Ohrt C , Gosling RD , 2015. Tackling imported malaria: an elimination endgame. Am J Trop Med Hyg 93: 139–144.
- 44.↑
Jitthai N , 2013. Migration and malaria. Southeast Asian J Trop Med Public Health 44 (Suppl 1):166–200, discussion 306–307.
- 45.↑
Guyant P , Canavati SE , Chea N , Ly P , Whittaker MA , Roca-Feltrer A , Yeung S , 2015. Malaria and the mobile and migrant population in Cambodia: a population movement framework to inform strategies for malaria control and elimination. Malar J 14: 252.
- 46.↑
Hamilton WL et al.2019. Evolution and expansion of multidrug-resistant malaria in Southeast Asia: a genomic epidemiology study. Lancet Infect Dis 19: 943–951.
- 47.↑
Zhou G et al.2014. Clinical malaria along the China–Myanmar border, Yunnan Province, China, January 2011–August 2012. Emerg Infect Dis 20: 675–678.
- 48.↑
Zhao X , Thanapongtharm W , Lawawirojwong S , Wei C , Tang Y , Zhou Y , Sun X , Cui L , Sattabongkot J , Kaewkungwal J , 2020. Malaria risk map using spatial multi-criteria decision analysis along Yunnan border during the pre-elimination period. Am J Trop Med Hyg 103: 793–809.
- 58.↑
Lo E , Zhou G , Oo W , Lee MC , Baum E , Felgner PL , Yang Z , Cui L , Yan G , 2015. Molecular inference of sources and spreading patterns of Plasmodium falciparum malaria parasites in internally displaced persons settlements in Myanmar–China border area. Infect Genet Evol 33: 189–196.
- 50.↑
Lo E , Lam N , Hemming-Schroeder E , Nguyen J , Zhou G , Lee MC , Yang Z , Cui L , Yan G , 2017. Frequent spread of Plasmodium vivax malaria maintains high genetic diversity at the Myanmar–China border, without distance and landscape barriers. J Infect Dis 216: 1254–1263.
- 51.↑
Putaporntip C , Miao J , Kuamsab N , Sattabongkot J , Sirichaisinthop J , Jongwutiwes S , Cui L , 2014. The Plasmodium vivax merozoite surface protein 3beta sequence reveals contrasting parasite populations in southern and northwestern Thailand. PLOS Negl Trop Dis 8: e3336.
- 52.↑
Gupta B , Parker DM , Fan Q , Reddy BP , Yan G , Sattabongkot J , Cui L , 2016. Microgeographically diverse Plasmodium vivax populations at the Thai–Myanmar border. Infect Genet Evol 45: 341–346.
- 53.↑
Gupta B , Reddy BP , Fan Q , Yan G , Sirichaisinthop J , Sattabongkot J , Escalante AA , Cui L , 2015. Molecular evolution of PvMSP3alpha Block II in Plasmodium vivax from diverse geographic origins. PLOS ONE 10: e0135396.
- 54.↑
Kittichai V , Koepfli C , Nguitragool W , Sattabongkot J , Cui L , 2017. Substantial population structure of Plasmodium vivax in Thailand facilitates identification of the sources of residual transmission. PLOS Negl Trop Dis 11: e0005930.
- 55.↑
Li Y et al.2020. Dynamics of Plasmodium vivax populations in border areas of the Greater Mekong sub-region during malaria elimination. Malar J 19: 145.
- 56.↑
Brashear AM , Fan Q , Hu Y , Li Y , Zhao Y , Wang Z , Cao Y , Miao J , Barry A , Cui L , 2020. Population genomics identifies a distinct Plasmodium vivax population on the China–Myanmar border of Southeast Asia. PLOS Negl Trop Dis 14: e0008506.
- 57.↑
Brashear AM et al.2020. New Plasmodium vivax Genomes from the China–Myanmar Border. Front Microbiol 11: 1930.
- 58.↑
Shetty AC et al.2019. Genomic structure and diversity of Plasmodium falciparum in Southeast Asia reveal recent parasite migration patterns. Nat Commun 10: 2665.
- 59.↑
Singh B , Kim Sung L , Matusop A , Radhakrishnan A , Shamsul SS , Cox-Singh J , Thomas A , Conway DJ , 2004. A large focus of naturally acquired Plasmodium knowlesi infections in human beings. Lancet 363: 1017–1024.
- 60.↑
Putaporntip C , Hongsrimuang T , Seethamchai S , Kobasa T , Limkittikul K , Cui L , Jongwutiwes S , 2009. Differential prevalence of Plasmodium infections and cryptic Plasmodium knowlesi malaria in humans in Thailand. J Infect Dis 199: 1143–1150.
- 61.↑
Jongwutiwes S , Buppan P , Kosuvin R , Seethamchai S , Pattanawong U , Sirichaisinthop J , Putaporntip C , 2011. Plasmodium knowlesi malaria in humans and macaques, Thailand. Emerg Infect Dis 17: 1799–1806.
- 62.↑
Sermwittayawong N , Singh B , Nishibuchi M , Sawangjaroen N , Vuddhakul V , 2012. Human Plasmodium knowlesi infection in Ranong Province, southwestern border of Thailand. Malar J 11: 36.
- 63.↑
Ngernna S et al.2019. Case report: case series of human Plasmodium knowlesi infection on the southern border of Thailand. Am J Trop Med Hyg 101: 1397–1401.
- 64.↑
Sugaram R , Boondej P , Srisutham S , Kunasol C , Pagornrat W , Boonyuen U , Dondorp AM , Saejeng A , Sudathip P , Imwong M , 2021. Genetic population of Plasmodium knowlesi during pre-malaria elimination in Thailand. Malar J 20: 454.
- 65.↑
Pongvongsa T , Culleton R , Ha H , Thanh L , Phongmany P , Marchand RP , Kawai S , Moji K , Nakazawa S , Maeno Y , 2018. Human infection with Plasmodium knowlesi on the Laos–Vietnam border. Trop Med Health 46: 33.
- 66.↑
Iwagami M et al.2018. First case of human infection with Plasmodium knowlesi in Laos. PLOS Negl Trop Dis 12: e0006244.
- 67.↑
Khim N et al.2011. Plasmodium knowlesi infection in humans, Cambodia, 2007–2010. Emerg Infect Dis 17: 1900–1902.
- 68.↑
Jiang N , Chang Q , Sun X , Lu H , Yin J , Zhang Z , Wahlgren M , Chen Q , 2010. Co-infections with Plasmodium knowlesi and other malaria parasites, Myanmar. Emerg Infect Dis 16: 1476–1478.
- 69.↑
Ghinai I et al.2017. Malaria epidemiology in central Myanmar: identification of a multi-species asymptomatic reservoir of infection. Malar J 16: 16.
- 70.↑
Van den Eede P , Van HN , Van Overmeir C , Vythilingam I , Duc TN , Hung le X , Manh HN , Anne J , D’Alessandro U , Erhart A , 2009. Human Plasmodium knowlesi infections in young children in central Vietnam. Malar J 8: 249.
- 71.↑
Marchand RP , Culleton R , Maeno Y , Quang NT , Nakazawa S , 2011. Co-infections of Plasmodium knowlesi, P. falciparum, and P. vivax among humans and Anopheles dirus mosquitoes, southern Vietnam. Emerg Infect Dis 17: 1232–1239.
- 72.↑
Putaporntip C , Jongwutiwes S , Thongaree S , Seethamchai S , Grynberg P , Hughes AL , 2010. Ecology of malaria parasites infecting Southeast Asian macaques: evidence from cytochrome b sequences. Mol Ecol 19: 3466–3476.
- 73.↑
Zhang X , Kadir KA , Quintanilla-Zarinan LF , Villano J , Houghton P , Du H , Singh B , Smith DG , 2016. Distribution and prevalence of malaria parasites among long-tailed macaques (Macaca fascicularis) in regional populations across Southeast Asia. Malar J 15: 450.
- 74.↑
Cuenca PR , Key S , Jumail A , Surendra H , Ferguson HM , Drakeley CJ , Fornace K , 2021. Epidemiology of the zoonotic malaria Plasmodium knowlesi in changing landscapes. Adv Parasitol 113: 225–286.
- 75.↑
Grigg MJ et al.2021. Plasmodium knowlesi detection methods for human infections—diagnosis and surveillance. Adv Parasitol 113: 77–130.
- 76.↑
Jongwutiwes S , Putaporntip C , Iwasaki T , Sata T , Kanbara H , 2004. Naturally acquired Plasmodium knowlesi malaria in human, Thailand. Emerg Infect Dis 10: 2211–2213.
- 77.↑
Shimizu S et al.2020. Malaria cross-sectional surveys identified asymptomatic infections of Plasmodium falciparum, Plasmodium vivax and Plasmodium knowlesi in Surat Thani, a Southern Province of Thailand. Int J Infect Dis 96: 445–451.
- 78.↑
Imwong M et al.2019. Asymptomatic natural human infections with the Simian malaria parasites Plasmodium cynomolgi and Plasmodium knowlesi. J Infect Dis 219: 695–702.
- 79.↑
Lover AA , Baird JK , Gosling R , Price RN , 2018. Malaria elimination: time to target all species. Am J Trop Med Hyg 99: 17–23.
- 80.↑
Putaporntip C , Kuamsab N , Pattanawong U , Yanmanee S , Seethamchai S , Jongwutiwes S , 2021. Plasmodium cynomolgi co-infections among symptomatic malaria patients, Thailand. Emerg Infect Dis 27: 590–593.
- 81.↑
Putaporntip C , Kuamsab N , Seethamchai S , Pattanawong U , Rojrung R , Yanmanee S , Cheng CW , Jongwutiwes S , 2022. Cryptic Plasmodium inui and P. fieldi infections among symptomatic malaria patients in Thailand. Clin Infect Disciab1060. doi: 10.1093/cid/ciab1060.
- 82.↑
Sai-Ngam P et al.2022. Case series of three malaria patients from Thailand infected with the simian parasite, Plasmodium cynomolgi. Malar J 21: 142.
- 83.↑
WHO , 2007. Anopheline Species Complexes in South and South-East Asia. World Health Organization, Geneva, Switzerland: SEARO Technical Publication No. 57: 102.
- 84.↑
Suwonkerd W , Ritthison W , Ngo CT , Tainchum K , Bangs MJ , Chareonviriphap T , 2013. Vector Biology and Malaria Transmission in Southeast Asia. Manguin S, ed. Anopheles Mosquitoes—New Insights into Malaria Vectors. IntechOpen, London, UK. 273–325.
- 85.↑
Erhart A et al.2005. Epidemiology of forest malaria in central Vietnam: a large scale cross-sectional survey. Malar J 4: 58.
- 86.↑
Lyttleton C , 2016. Deviance and resistance: malaria elimination in the Greater Mekong Subregion. Soc Sci Med 150: 144–152.
- 87.↑
Rerolle F , Dantzer E , Lover AA , Marshall JM , Hongvanthong B , Sturrock HJ , Bennett A , 2021. Spatio-temporal associations between deforestation and malaria incidence in Lao PDR. eLife 10: e56974.
- 88.↑
Zhong D , Wang X , Xu T , Zhou G , Wang Y , Lee MC , Hartsel JA , Cui L , Zheng B , Yan G , 2016. Effects of microclimate condition changes due to land use and land cover changes on the survivorship of malaria vectors in China–Myanmar border region. PLOS ONE 11: e0155301.
- 89.↑
Wang X , Zhou G , Zhong D , Wang Y , Yang Z , Cui L , Yan G , 2016. Life-table studies revealed significant effects of deforestation on the development and survivorship of Anopheles minimus larvae. Parasit Vectors 9: 323.
- 90.↑
Sriwichai P , Samung Y , Sumruayphol S , Kiattibutr K , Kumpitak C , Payakkapol A , Kaewkungwal J , Yan G , Cui L , Sattabongkot J , 2016. Natural human Plasmodium infections in major Anopheles mosquitoes in western Thailand. Parasit Vectors 9: 17.
- 91.↑
Sumruayphol S , Chaiphongpachara T , Samung Y , Ruangsittichai J , Cui L , Zhong D , Sattabongkot J , Sriwichai P , 2020. Seasonal dynamics and molecular differentiation of three natural Anopheles species (Diptera: Culicidae) of the Maculatus group (Neocellia series) in malaria hotspot villages of Thailand. Parasit Vectors 13: 574.
- 92.↑
Wang Y , Zhong D , Cui L , Lee MC , Yang Z , Yan G , Zhou G , 2015. Population dynamics and community structure of Anopheles mosquitoes along the China–Myanmar border. Parasit Vectors 8: 445.
- 93.↑
Bunmee K , Thaenkham U , Saralamba N , Ponlawat A , Zhong D , Cui L , Sattabongkot J , Sriwichai P , 2021. Population genetic structure of the malaria vector Anopheles minimus in Thailand based on mitochondrial DNA markers. Parasit Vectors 14: 496.
- 94.↑
Chatpiyaphat K , Sumruayphol S , Dujardin JP , Samung Y , Phayakkaphon A , Cui L , Ruangsittichai J , Sungvornyothin S , Sattabongkot J , Sriwichai P , 2021. Geometric morphometrics to distinguish the cryptic species Anopheles minimus and An. harrisoni in malaria hot spot villages, Western Thailand. Med Vet Entomol 35: 293–301.
- 95.↑
Zhong D et al.2013. Relationship between knockdown resistance, metabolic detoxification and organismal resistance to pyrethroids in Anopheles sinensis. PLOS ONE 8: e55475.
- 96.↑
Chang X , Zhong D , Fang Q , Hartsel J , Zhou G , Shi L , Fang F , Zhu C , Yan G , 2014. Multiple resistances and complex mechanisms of Anopheles sinensis mosquito: a major obstacle to mosquito-borne diseases control and elimination in China. PLOS Negl Trop Dis 8: e2889.
- 97.↑
Qin Q , Li Y , Zhong D , Zhou N , Chang X , Li C , Cui L , Yan G , Chen XG , 2014. Insecticide resistance of Anopheles sinensis and An. vagus in Hainan Island, a malaria-endemic area of China. Parasit Vectors 7: 92.
- 98.↑
Chen S et al.2019. Insecticide resistance status and mechanisms of Anopheles sinensis (Diptera: Culicidae) in Wenzhou, an important coastal port city in China. J Med Entomol 56: 803–810.
- 99.↑
Chang X et al.2016. Landscape genetic structure and evolutionary genetics of insecticide resistance gene mutations in Anopheles sinensis. Parasit Vectors 9: 228.
- 100.↑
Zhu G et al.2014. Transcriptome profiling of pyrethroid resistant and susceptible mosquitoes in the malaria vector, Anopheles sinensis. BMC Genomics 15: 448.
- 101.↑
Chang X , Zhong D , Wang X , Bonizzoni M , Li Y , Zhou G , Cui L , Wei X , Yan G , 2020. Genomic variant analyses in pyrethroid resistant and susceptible malaria vector, Anopheles sinensis. G3 (Bethesda) 10: 2185–2193.
- 102.↑
Wongsrichanalai C , Meshnick SR , 2008. Declining artesunate-mefloquine efficacy against falciparum malaria on the Cambodia–Thailand border. Emerg Infect Dis 14: 716–719.
- 103.↑
Rogers WO , Sem R , Tero T , Chim P , Lim P , Muth S , Socheat D , Ariey F , Wongsrichanalai C , 2009. Failure of artesunate-mefloquine combination therapy for uncomplicated Plasmodium falciparum malaria in southern Cambodia. Malar J 8: 10.
- 104.↑
Amaratunga C et al.2016. Dihydroartemisinin-piperaquine resistance in Plasmodium falciparum malaria in Cambodia: a multisite prospective cohort study. Lancet Infect Dis 16: 357–365.
- 105.↑
Saunders DL , Vanachayangkul P , Lon C , 2014. Dihydroartemisinin-piperaquine failure in Cambodia. N Engl J Med 371: 484–485.
- 106.↑
Leang R et al.2015. Evidence of Plasmodium falciparum malaria multidrug resistance to artemisinin and piperaquine in western Cambodia: dihydroartemisinin-piperaquine open-label multicenter clinical assessment. Antimicrob Agents Chemother 59: 4719–4726.
- 107.↑
Spring MD et al.2015. Dihydroartemisinin-piperaquine failure associated with a triple mutant including kelch13 C580Y in Cambodia: an observational cohort study. Lancet Infect Dis 15: 683–691.
- 108.↑
Phyo AP et al.2012. Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet 379: 1960–1966.
- 109.↑
Ashley EA et al.2014. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 371: 411–423.
- 110.↑
WHO , 2019. Artemisinin Resistance and Artemisinin-Based Combination Therapy Efficacy. Available at: https://www.who.int/docs/default-source/documents/publications/gmp/who-cds-gmp-2019-17-eng.pdf?ua=1. Accessed August 18, 2022.
- 111.↑
White LJ et al.2015. Defining the in vivo phenotype of artemisinin-resistant falciparum malaria: a modelling approach. PLOS Med 12: e1001823.
- 112.↑
WWARN Artemisinin-based Combination Therapy Africa Baseline Study Group , 2015. Clinical determinants of early parasitological response to ACTs in African patients with uncomplicated falciparum malaria: a literature review and meta-analysis of individual patient data. BMC Med 13: 212.
- 113.↑
Witkowski B et al.2013. 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 13: 1043–1049.
- 114.↑
Ariey F et al.2014. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature 505: 50–55.
- 115.↑
Malaria GEN Plasmodium falciparum Community Project, 2016. Genomic epidemiology of artemisinin resistant malaria. eLife 5: e08714.
- 116.↑
Menard D et al.2016. A worldwide map of Plasmodium falciparum K13-propeller polymorphisms. N Engl J Med 374: 2453–2464.
- 117.↑
Huang F et al.2015. A single mutation in K13 predominates in southern China and is associated with delayed clearance of Plasmodium falciparum following artemisinin treatment. J Infect Dis 212: 1629–1635.
- 118.↑
Miotto O et al.2015. Genetic architecture of artemisinin-resistant Plasmodium falciparum. Nat Genet 47: 226–234.
- 119.↑
Straimer J et al.2015. Drug resistance. K13-propeller mutations confer artemisinin resistance in Plasmodium falciparum clinical isolates. Science 347: 428–431.
- 120.↑
Ghorbal M , Gorman M , Macpherson CR , Martins RM , Scherf A , Lopez-Rubio JJ , 2014. Genome editing in the human malaria parasite Plasmodium falciparum using the CRISPR-Cas9 system. Nat Biotechnol 32: 819–821.
- 121.↑
Siddiqui FA et al.2020. Role of Plasmodium falciparum Kelch 13 protein mutations in P. falciparum populations from northeastern Myanmar in mediating artemisinin resistance. MBio 11: e01134–e19.
- 122.↑
Stokes BH et al.2021. Plasmodium falciparum K13 mutations in Africa and Asia impact artemisinin resistance and parasite fitness. eLife 10: e66277.
- 123.↑
Wang Y et al.2015. Clinical efficacy of dihydroartemisinin-piperaquine for the treatment of uncomplicated Plasmodium falciparum malaria at the China–Myanmar border. Am J Trop Med Hyg 93: 577–583.
- 124.↑
Wu Y , Soe MT , Aung PL , Zhao L , Zeng W , Menezes L , Yang Z , Kyaw MP , Cui L , 2020. Efficacy of artemether-lumefantrine for treating uncomplicated Plasmodium falciparum cases and molecular surveillance of drug resistance genes in western Myanmar. Malar J 19: 304.
- 125.↑
Wang Z et al.2015. Artemisinin resistance at the China–Myanmar border and association with mutations in the K13 propeller gene. Antimicrob Agents Chemother 59: 6952–6959.
- 126.↑
Bai Y et al.2018. Longitudinal surveillance of drug resistance in Plasmodium falciparum isolates from the China–Myanmar border reveals persistent circulation of multidrug resistant parasites. Int J Parasitol Drugs Drug Resist 8: 320–328.
- 127.↑
Zhang J et al.2019. 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 10: 20–27.
- 128.↑
Wang S et al.2020. Molecular surveillance and in vitro drug sensitivity study of Plasmodium falciparum isolates from the China–Myanmar border. Am J Trop Med Hyg 103: 1100–1106.
- 129.↑
Si Y et al.2021. In vitro susceptibility of Plasmodium falciparum isolates from the China–Myanmar border area to piperaquine and association with candidate markers. Antimicrob Agents Chemother 65 : e02305–20.
- 130.↑
Davis TM , Hung TY , Sim IK , Karunajeewa HA , Ilett KF , 2005. Piperaquine: a resurgent antimalarial drug. Drugs 65: 75–87.
- 131.↑
Guo XB , 1993. Randomised comparison on the treatment of falciparum malaria with dihydroartemisinin and piperaquine. Zhonghua Yi Xue Za Zhi 73: 602–604, 638.
- 132.↑
Yang Z , Zhang Z , Sun X , Wan W , Cui L , Zhang X , Zhong D , Yan G , 2007. Molecular analysis of chloroquine resistance in Plasmodium falciparum in Yunnan Province, China. Trop Med Int Health 12: 1051–1060.
- 133.↑
Liu H et al.2015. In vivo monitoring of dihydroartemisinin-piperaquine sensitivity in Plasmodium falciparum along the China–Myanmar border of Yunnan Province, China from 2007 to 2013. Malar J 14: 47.
- 134.↑
Amato R et al.2017. Genetic markers associated with dihydroartemisinin-piperaquine failure in Plasmodium falciparum malaria in Cambodia: a genotype-phenotype association study. Lancet Infect Dis 17: 164–173.
- 135.↑
Witkowski B et al.2017. A surrogate marker of piperaquine-resistant Plasmodium falciparum malaria: a phenotype-genotype association study. Lancet Infect Dis 17: 174–183.
- 136.↑
Duru V et al.2015. Plasmodium falciparum dihydroartemisinin-piperaquine failures in Cambodia are associated with mutant K13 parasites presenting high survival rates in novel piperaquine in vitro assays: retrospective and prospective investigations. BMC Med 13: 305.
- 137.↑
Agrawal S et al.2017. Association of a novel mutation in the Plasmodium falciparum chloroquine resistance transporter with decreased piperaquine sensitivity. J Infect Dis 216: 468–476.
- 138.↑
Ross LS et al.2018. Emerging Southeast Asian PfCRT mutations confer Plasmodium falciparum resistance to the first-line antimalarial piperaquine. Nat Commun 9: 3314.
- 139.↑
Tun KM et al.2015. Spread of artemisinin-resistant Plasmodium falciparum in Myanmar: a cross-sectional survey of the K13 molecular marker. Lancet Infect Dis 15: 415–421.
- 140.↑
Wang Z , Shrestha S , Li X , Miao J , Yuan L , Cabrera M , Grube C , Yang Z , Cui L , 2015. Prevalence of K13-propeller polymorphisms in Plasmodium falciparum from China–Myanmar border in 2007–2012. Malar J 14: 168.
- 141.↑
Ye R et al.2016. Distinctive origin of artemisinin-resistant Plasmodium falciparum on the China–Myanmar border. Sci Rep 6: 20100.
- 142.↑
Zhao Y et al.2019. Genetic variations associated with drug resistance markers in asymptomatic Plasmodium falciparum infections in Myanmar. Genes (Basel) 10 : 692.
- 143.↑
Rosenthal MR , Ng CL , 2020. Plasmodium falciparum artemisinin resistance: the effect of heme, protein damage, and parasite cell stress response. ACS Infect Dis 6: 1599–1614.
- 144.↑
Xie SC , Ralph SA , Tilley L , 2020. K13, the cytostome, and artemisinin resistance. Trends Parasitol 36: 533–544.
- 145.↑
Wicht KJ , Mok S , Fidock DA , 2020. Molecular mechanisms of drug resistance in Plasmodium falciparum malaria. Annu Rev Microbiol 74: 431–454.
- 146.↑
Sutherland CJ , Henrici RC , Artavanis-Tsakonas K , 2020. Artemisinin susceptibility in the malaria parasite Plasmodium falciparum: propellers, adaptor proteins and the need for cellular healing. FEMS Microbiol Rev 45: fuaa056.
- 147.↑
Rocamora F , Zhu L , Liong KY , Dondorp A , Miotto O , Mok S , Bozdech Z , 2018. Oxidative stress and protein damage responses mediate artemisinin resistance in malaria parasites. PLOS Pathog 14: e1006930.
- 148.↑
Siddiqui FA et al.2018. Plasmodium falciparum falcipain-2a polymorphisms in Southeast Asia and their association with artemisinin resistance. J Infect Dis 218: 434–442.
- 149.↑
Mukherjee A et al.2017. Artemisinin resistance without pfkelch13 mutations in Plasmodium falciparum isolates from Cambodia. Malar J 16: 195.
- 150.↑
Sutherland CJ et al.2017. Pfk13-independent treatment failure in four imported cases of Plasmodium falciparum malaria treated with artemether-lumefantrine in the United Kingdom. Antimicrob Agents Chemother 61: e02382–e16.
- 151.↑
Wang Z et al.2016. Genome-wide association analysis identifies genetic loci associated with resistance to multiple antimalarials in Plasmodium falciparum from China–Myanmar border. Sci Rep 6: 33891.
- 152.↑
Cui L , Brashear A , Menezes L , Adams J , 2021. Elimination of Plasmodium vivax malaria: problems and solutions. Current Topics and Emerging Issues in Malaria Elimination: IntechOpen, London, UK. 159–185.
- 153.↑
Li Q et al.2015. Prevalence and molecular characterization of glucose-6-phosphate dehydrogenase deficiency at the China–Myanmar border. PLOS ONE 10: e0134593.
- 154.↑
Matsuoka H , Wang J , Hirai M , Arai M , Yoshida S , Kobayashi T , Jalloh A , Lin K , Kawamoto F , 2004. Glucose-6-phosphate dehydrogenase (G6PD) mutations in Myanmar: G6PD Mahidol (487G>A) is the most common variant in the Myanmar population. J Hum Genet 49: 544–547.
- 155.↑
Bancone G , Chu CS , Somsakchaicharoen R , Chowwiwat N , Parker DM , Charunwatthana P , White NJ , Nosten FH , 2014. Characterization of G6PD genotypes and phenotypes on the Northwestern Thailand–Myanmar border. PLOS ONE 9: e116063.
- 156.↑
Nuchprayoon I , Sanpavat S , Nuchprayoon S , 2002. Glucose-6-phosphate dehydrogenase (G6PD) mutations in Thailand: G6PD Viangchan (871G>A) is the most common deficiency variant in the Thai population. Hum Mutat 19: 185.
- 157.↑
Louicharoen C et al.2009. Positively selected G6PD-Mahidol mutation reduces Plasmodium vivax density in Southeast Asians. Science 326: 1546–1549.
- 158.↑
Yi H et al.2019. The glucose-6-phosphate dehydrogenase Mahidol variant protects against uncomplicated Plasmodium vivax infection and reduces disease severity in a Kachin population from northeast Myanmar. Infect Genet Evol 75: 103980.
- 159.↑
Minucci A , Moradkhani K , Hwang MJ , Zuppi C , Giardina B , Capoluongo E , 2012. Glucose-6-phosphate dehydrogenase (G6PD) mutations database: review of the “old” and update of the new mutations. Blood Cells Mol Dis 48: 154–165.
- 160.↑
Charoenlarp P , Areekul S , Pholpothi T , Harinasuta T , 1973. The course of primaquine-induced haemolysis in G-6-PD-deficient Thais. J Med Assoc Thai 56: 392–397.
- 161.↑
Charoenlarp P , Areekul S , Harinasuta T , Sirivorasarn P , 1972. The haemolytic effect of a single dose of 45 mg of primaquine in G-6-PD deficient Thais. J Med Assoc Thai 55: 631–638.
- 162.↑
Deng Z et al.2017. Co-inheritance of glucose-6-phosphate dehydrogenase deficiency mutations and hemoglobin E in a Kachin population in a malaria-endemic region of Southeast Asia. PLOS ONE 12: e0177917.
- 163.↑
Chen X , He Y , Miao Y , Yang Z , Cui L , 2017. A young man with severe acute haemolytic anaemia. BMJ 359: j4263.
- 164.↑
Bennett JW , Pybus BS , Yadava A , Tosh D , Sousa JC , McCarthy WF , Deye G , Melendez V , Ockenhouse CF , 2013. Primaquine failure and cytochrome P-450 2D6 in Plasmodium vivax malaria. N Engl J Med 369: 1381–1382.
- 165.↑
Pybus BS et al.2013. The metabolism of primaquine to its active metabolite is dependent on CYP 2D6. Malar J 12: 212.
- 166.↑
Pybus BS et al.2012. CYP450 phenotyping and accurate mass identification of metabolites of the 8-aminoquinoline, anti-malarial drug primaquine. Malar J 11: 259.
- 167.↑
Baird JK et al., 2018. Association of impaired cytochrome P450 2D6 activity genotype and phenotype with therapeutic efficacy of primaquine treatment for latent Plasmodium vivax malaria. JAMA Netw Open 1: e181449.
- 168.↑
Sutanto I et al.2013. Randomized, open-label trial of primaquine against vivax malaria relapse in Indonesia. Antimicrob Agents Chemother 57: 1128–1135.
- 169.↑
Nelwan EJ et al.2015. Randomized trial of primaquine hypnozoitocidal efficacy when administered with artemisinin-combined blood schizontocides for radical cure of Plasmodium vivax in Indonesia. BMC Med 13: 294.
- 170.↑
Liu P et al.2021. Increasing proportions of relapsing parasite species among imported malaria in China’s Guangxi Province from Western and Central Africa. Travel Med Infect Dis 43: 102130.
- 171.↑
He X et al.2021. Unraveling the complexity of imported malaria infections by amplicon deep sequencing. Front Cell Infect Microbiol 11: 725859.
- 172.↑
He X et al.2019. Multiple relapses of Plasmodium vivax malaria acquired from West Africa and association with poor metabolizer CYP2D6 variant: a case report. BMC Infect Dis 19: 704.
- 173.↑
Douglas NM , Anstey NM , Angus BJ , Nosten F , Price RN , 2010. Artemisinin combination therapy for vivax malaria. Lancet Infect Dis 10: 405–416.
- 174.↑
Myat-Phone-Kyaw M-O , Myint-Lwin T-Z , Kyin-Hla-Aye N-N-Y , 1993. Emergence of chloroquine-resistant Plasmodium vivax in Myanmar (Burma). Trans R Soc Trop Med Hyg 87: 687.
- 175.↑
Marlar T , Myat Phone K , Aye Yu S , Khaing Khaing G , Ma S , Myint O , 1995. Development of resistance to chloroquine by Plasmodium vivax in Myanmar. Trans R Soc Trop Med Hyg 89: 307–308.
- 176.↑
Guthmann JP , Pittet A , Lesage A , Imwong M , Lindegardh N , Min Lwin M , Zaw T , Annerberg A , de Radigues X , Nosten F , 2008. Plasmodium vivax resistance to chloroquine in Dawei, Southern Myanmar. Trop Med Int Health 13: 91–98.
- 177.↑
Htun MW et al.2017. Chloroquine efficacy for Plasmodium vivax in Myanmar in populations with high genetic diversity and moderate parasite gene flow. Malar J 16: 281.
- 178.↑
Yuan L et al.2015. Therapeutic responses of Plasmodium vivax malaria to chloroquine and primaquine treatment in Northeastern Myanmar. Antimicrob Agents Chemother 59: 1230–1235.
- 179.↑
Xu S et al.2020. Efficacy of directly-observed chloroquine-primaquine treatment for uncomplicated acute Plasmodium vivax malaria in northeast Myanmar: a prospective open-label efficacy trial. Travel Med Infect Dis 36: 101499.
- 180.↑
Li J et al.2020. Ex vivo susceptibilities of Plasmodium vivax isolates from the China–Myanmar border to antimalarial drugs and association with polymorphisms in Pvmdr1 and Pvcrt-o genes. PLOS Negl Trop Dis 14: e0008255.
- 181.↑
Suwanarusk R et al.2007. Chloroquine resistant Plasmodium vivax: in vitro characterisation and association with molecular polymorphisms. PLOS ONE 2: e1089.
- 182.↑
Zeng W et al.2021. Molecular surveillance and ex vivo drug susceptibilities of Plasmodium vivax isolates from the China–Myanmar border. Front Cell Dev Biol 11 : 738075.
- 183.↑
Price RN , Marfurt J , Chalfein F , Kenangalem E , Piera KA , Tjitra E , Anstey NM , Russell B , 2010. In vitro activity of pyronaridine against multidrug-resistant Plasmodium falciparum and Plasmodium vivax. Antimicrob Agents Chemother 54: 5146–5150.
- 184.↑
Schousboe ML et al.2015. Multiple origins of mutations in the mdr1 gene—a putative marker of chloroquine resistance in P. vivax. PLOS Negl Trop Dis 9: e0004196.
- 185.↑
Kittichai V , Nguitragool W , Ngassa Mbenda HG , Sattabongkot J , Cui L , 2018. Genetic diversity of the Plasmodium vivax multidrug resistance 1 gene in Thai parasite populations. Infect Genet Evol 64: 168–177.
- 186.↑
Ngassa Mbenda HG et al.2020. Evolution of the Plasmodium vivax multidrug resistance 1 gene in the Greater Mekong subregion during malaria elimination. Parasit Vectors 13: 67.
- 187.↑
Noisang C , Prosser C , Meyer W , Chemoh W , Ellis J , Sawangjaroen N , Lee R , 2019. Molecular detection of drug resistant malaria in southern Thailand. Malar J 18: 275.
- 188.↑
Zhao Y et al.2020. Molecular surveillance for drug resistance markers in Plasmodium vivax isolates from symptomatic and asymptomatic infections at the China–Myanmar border. Malar J 19: 281.
- 189.↑
Lin JT , Patel JC , Kharabora O , Sattabongkot J , Muth S , Ubalee R , Schuster AL , Rogers WO , Wongsrichanalai C , Juliano JJ , 2013. Plasmodium vivax isolates from Cambodia and Thailand show high genetic complexity and distinct patterns of P. vivax multidrug resistance gene 1 (pvmdr1) polymorphisms. Am J Trop Med Hyg 88: 1116–1123.
- 190.↑
Lu F et al.2011. Genetic polymorphism in pvmdr1 and pvcrt-o genes in relation to in vitro drug susceptibility of Plasmodium vivax isolates from malaria-endemic countries. Acta Trop 117: 69–75.
- 191.↑
Wang X , Ruan W , Zhou S , Feng X , Yan H , Huang F , 2020. Prevalence of molecular markers associated with drug resistance of Plasmodium vivax isolates in western Yunnan Province, China. BMC Infect Dis 20: 307.
- 192.↑
Iwagami M et al.2018. Heterogeneous distribution of k13 mutations in Plasmodium falciparum in Laos. Malar J 17: 483.
- 193.↑
Thuy-Nhien N et al.2017. K13 propeller mutations in Plasmodium falciparum populations in regions of malaria endemicity in Vietnam from 2009 to 2016. Antimicrob Agents Chemother 61 : e01578–16.
- 194.↑
Imwong M et al.2017. The spread of artemisinin-resistant Plasmodium falciparum in the Greater Mekong subregion: a molecular epidemiology observational study. Lancet Infect Dis 17: 491–497.
- 195.↑
Kobasa T et al.2018. Emergence and spread of kelch13 mutations associated with artemisinin resistance in Plasmodium falciparum parasites in 12 Thai provinces from 2007 to 2016. Antimicrob Agents Chemother 62: e02141–e17.