Open access peer-reviewed chapter

Elimination of Plasmodium vivax Malaria: Problems and Solutions

Written By

Liwang Cui, Awtum Brashear, Lynette Menezes and John Adams

Submitted: 19 December 2020 Reviewed: 12 February 2021 Published: 08 March 2021

DOI: 10.5772/intechopen.96604

From the Edited Volume

Current Topics and Emerging Issues in Malaria Elimination

Edited by Alfonso J. Rodriguez-Morales

Chapter metrics overview

603 Chapter Downloads

View Full Metrics

Abstract

Malaria is caused by multiple parasitic species of the genus Plasmodium. Although P. falciparum accounts for the highest mortality, P. vivax is the most geographically dispersed and the most common species outside of Africa. Several unique biological features make P. vivax less responsive to conventional control measures and allow it to persist even after elimination of P. falciparum. The ability of P. vivax to develop in diverse vectors at lower ambient temperatures bestows it a greater distribution range and resilience to ecological changes. Its tropism for reticulocytes often causes low-density infections below the levels detectable by routine diagnostic tests, demanding the development of more sensitive diagnostics. P. vivax produces gametocytes early enabling transmission before the manifestation of clinical symptoms, thus emphasizing the need for an integrated vector control strategy. More importantly, its dormant liver stage which engenders relapse is difficult to diagnose and treat. The deployment of available treatments for the liver hypnozoites, including primaquine and the recent U.S. Food and Drug Administration-approved tafenoquine, requires point-of-care diagnostics to detect glucose-6-phosphate dehydrogenase deficiency among endemic human populations. Here we review the continued challenges to effectively control P. vivax and explore integrated technologies and targeted strategies for the elimination of vivax malaria.

Keywords

  • Plasmodium vivax
  • relapse
  • transmission
  • G6PD
  • CYP2D6
  • radical cure

1. Introduction

Malaria has been an ancient scourge of humankind and efforts to mitigate the harm from malaria have been relentless. In 1955, the World Health Organization (WHO) launched the Global Malaria Eradication Program (GMEP), relying heavily on two essential tools: chloroquine (CQ), a safe and effective drug for malaria prevention and treatment, and the insecticide DDT for vector control. Despite the GMEP’s enormous success in reducing malaria burden in many countries outside of sub-Saharan Africa, its failure to sustain the program resulted in malaria resurgence and discontinuation of this global campaign in 1969 [1]. The considerable gains achieved in many areas were soon lost and the world witnessed a sharp rise in malaria incidence in the following two decades. In India, for example, malaria prevalence reduced from an estimated 75 million cases to about 100,000 cases annually during the GMEP, only to rapidly expand to 6.5 million in 1976 [2, 3]. In recognition of this huge malaria burden, the Roll Back Malaria Partnership launched in 1998, marking a renewed attack on this disease resulting in a declining incidence of malaria globally. Empowered by a strong political will and enabled by financial commitment and new interventions, many national malaria control programs (NMCPs) now consider malaria elimination an attainable goal. WHO’s “Global Technical Strategy for Malaria 2016-2030” provided goals for the next 15 years and specific guidelines for achieving these goals. Among them, “eliminating malaria in at least 35 countries” and “preventing the re-establishment of malaria in all countries that are malaria-free” specifically address the tasks to attain and sustain malaria elimination. Significant strides have been made toward malaria elimination in the past two decades with 19 countries attaining zero indigenous cases for 3 years or more between 2000 and 2018. These countries include Sri Lanka, Paraguay, and Uzbekistan, which were recently certified as malaria-free [4]. Despite these laudable achievements, formidable challenges still lie ahead for many endemic nations to achieve malaria elimination.

Of the six Plasmodium species naturally infecting humans, P. falciparum is usually considered the most virulent and is associated with the vast majority of deaths, while P. vivax is the most geographically widespread. In comparison, P. ovale curtisi, P. ovale wallikeri, and P. malariae are much less common, whereas the monkey malaria parasite P. knowlesi is primarily associated with zoonotic infections [5]. Since malaria elimination is the interruption of local malaria transmission (zero indigenous cases) in a defined geographic area, it is time to target the elimination of all malaria parasite species simultaneously to set the final stage for malaria eradication [6].

At the same time that malaria incidence is continually declining [4], malaria epidemiology is rapidly changing [7]. In countries pursuing elimination, structural changes in at-risk populations have resulted in malaria becoming geographically clustered in hard-to-reach pockets. “Border malaria” has become a shared phenomenon and malaria is increasingly an imported disease. Additionally, because of a divergent response by each species to control interventions, P. vivax has become the predominant parasite in malaria-endemic countries outside of Africa. Most pre-elimination countries—such as the members of the Asian Pacific Malaria Elimination Network (APMEN, www.apmen.org) — must be ready to face the ultimate challenge of eliminating vivax malaria, a potentially long and arduous process. In fact, because of the possibility of relapse, the WHO malaria-free certification requires no cases for three years [1]. Here we review the changing malaria epidemiology and discuss the challenges associated with vivax malaria elimination and solutions to address them.

Advertisement

2. Geographic distribution and epidemiology of vivax malaria

Outside of Africa, P. vivax is the most common parasite causing malaria. It accounts for 75, 50, and 29% of the malaria burden in the Americas, SE Asia, and the East Mediterranean, respectively [4]. The parasite’s ability to complete its sporogonic development in mosquitoes at ambient temperatures as low as 16 °C and to lie dormant for seasonal transmission has extended its geographical range deep into the temperate zones. There is considerable spatial heterogeneity in P. vivax distribution at the global and local scales. SE Asia carries more than half of the global P. vivax burden (Table 1). In the Asian continent, India, Cambodia, and Myanmar have higher endogenous P. vivax burden, and transmission is concentrated along international borders [8]. Similarly, the southern part of South America has a relatively low burden with Paraguay and Argentina recently achieving malaria-free status, whereas the northern part of the continent has a substantial P. vivax burden (Figure 1). In Africa, until recently, P. vivax was documented only in the Horn of Africa, and considered extremely rare or “absent” in Central and West Africa because of the dominance of the Duffy-negative blood group [10], a required receptor for erythrocyte invasion by P. vivax [11]. Increasing reports of P. vivax in Duffy-negative individuals suggests its capability to exploit Duffy-independent invasion pathways [12, 13]. In the last decade, the growing evidence of P. vivax transmission in all regions of Africa, including acute and asymptomatic cases, infected vectors, serological indicators, and infected international travelers, indicates more P. vivax transmission than previously thought [14, 15]. Although malaria control programs in Africa are justifiably focused on P. falciparum (given the striking morbidity and mortality associated with this species), P. vivax is becoming an emerging concern for malaria elimination from African nations.

AfricaAmericaEastern MediterraneanSE AsiaWestern PacificTotal
P. vivax7047001,4143,9476907,500
All213,0009294,9007,9001,980228,000

Table 1.

Estimated cases of P. vivax and all malaria (×1,000) by WHO region [4].

Figure 1.

Global distribution of P. vivax malaria. Shading represents incidence in cases per 1000 people per year [8, 9]. Very low incidence areas are shaded in gray.

Throughout history, P. vivax has shown extreme resilience to control measures [16], and in many areas where P. falciparum and P. vivax co-exist, P. vivax is becoming predominant [17, 18]. With this shift in species predominance come changes in the at-risk populations. In areas of P. falciparum and P. vivax sympatry, clinical episodes of vivax malaria rapidly decrease around 12 months of age, whereas P. falciparum cases continue to rise until about 3 years of age [19, 20]. Since exposure undoubtedly plays a role in the acquisition and maintenance of immunity, P. vivax recurrence may allow for repeated exposure from fewer infection events which may contribute to this age discrepancy. Additionally, primaquine (PQ) is not commonly given to children below 5 years, while chloroquine (CQ) underdosing is not unusual [21], resulting in repeated P. vivax attacks in young children. Recent studies in SE Asia showed that school-aged children had significantly increased odds of acquiring P. vivax infections [18, 22]. Because PQ is contraindicated for pregnant women, radical treatment of P. vivax remains difficult in this group. Consequentially, relapsing episodes of malaria during pregnancy can lead to congenital malaria [23, 24, 25]. Certain occupations such as soldiers and forest workers are also more vulnerable to malaria infections [18, 26]. A better understanding of local malaria epidemiology will be essential for implementing targeted control measures.

Advertisement

3. Morbidity of vivax malaria

Biologically, P. vivax exhibits key differences from P. falciparum influencing its transmission, presentation and outcome [27]. Historically, P. vivax malaria has been mistakenly described as “benign tertian malaria”. In fact, P. vivax infection causes a full spectrum of disease symptoms ranging from uncomplicated febrile illness to severe and fatal malaria. Severe P. vivax malaria is often associated with severe anemia, a common complication, as well as thrombocytopenia, acute respiratory distress, hepatic dysfunction, renal failure, seizures or coma, and shock [28, 29]. Severe anemia is the most common complication associated with P. vivax malaria [30, 31, 32]. P. vivax has a strong preference for CD71high reticulocytes [33] and aggravates anemia by targeting cells immediately after replacement [34, 35]. Recurrent P. vivax parasitemia further elevates the risk of severe anemia [36]. Although the risk of thrombocytopenia is prevalent in all forms of malaria, evidence suggests that it is more common in P. vivax than P. falciparum patients [37, 38]. In pregnant women, P. vivax infection is associated with a higher risk of anemia, abortion and low birth weight [39, 40]. Furthermore, the presence of co-morbidities may exacerbate P. vivax infections resulting in severe and life-threatening complications.

Advertisement

4. Relapse

One distinctive feature of P. vivax that enables the parasite to evade conventional control measures designed for P. falciparum is the formation of a dormant liver stage, termed hypnozoite [41]. Hypnozoites persist in a non-dividing fashion within the liver where they may be awakened weeks and months later causing relapse. Recent detections of P. vivax parasites in the bone marrow and spleen have raised the possibility that the extravascular merozoites might be an additional source of recurrence in addition to the relapse from hypnozoite activation [42, 43]. Two latency forms of P. vivax strains are recognized. The long-latency strains (e.g., the St. Elizabeth strain) prevalent in temperate zones have either a ~ 9-month latency period or a 2-week incubation time for the primary infection followed by a ~ 9-month interval before the relapse, allowing for parasite survival through the long winter season when mosquito vectors are absent [44, 45]. In contrast, for the short-latency tropical strains (e.g., the Chesson strain) from SE Asia and Oceania, relapses typically occur ~3 weeks after the primary infection [46]. Some areas, such as the Greater Mekong subregion host both types of strains [46, 47]. Besides, the sporozoite inoculation load can impact the latency period. The ratio of hypnozoites to sporozoites could vary by strain, and parasites with higher proportions of hypnozoites may be more inclined toward frequent relapses [48, 49]. Mechanisms of hypnozoite reactivation are elusive and may involve external stimuli such as drugs, another malaria infection, or other infectious diseases [50]. A meta-analysis of P. falciparum drug efficacy trials reveals a high risk of P. vivax parasitemia after treatment of falciparum malaria [51].

Relapse increases the morbidity associated with P. vivax infections. Relapses can contribute to 50–80% of the overall vivax infections in high transmission areas [52, 53, 54, 55]; sometimes occurring repeatedly from the same infection, such as a case from Eritrea where one patient’s three episodes of vivax malaria were caused by meiotic siblings suggesting the same infection event [56]. Additionally, P. vivax is frequently found in highly polyclonal infections, even in areas with relatively low malaria endemicity. One study from Cambodia found that around half of polyclonal infections might have resulted from relapse [57]. Long-distance parasite migration and introduction also favor hypnozoites. When introduced by an asymptomatic human host to an area with suitable Anopheles vector species, relapsed parasites could spark autochthonous infections and establish new transmission foci, as observed in Greece [58].

Advertisement

5. Diagnosis

Accurate and timely diagnosis of malaria by species is essential for the delivery of appropriate treatments. In clinical settings, acute vivax malaria is diagnosed by microscopy or rapid diagnostic tests (RDTs). Routine microscopy has a limit of detection (LOD) of around 50 parasites/μl of blood [59, 60]. Specific training is required, as misidentification of the infecting species is fairly common [59, 61]. RDTs are a fast, affordable and efficient method for malaria diagnosis with a LOD of ~200 parasites/μl, but the sensitivity varies among brands [62]. The tropism of P. vivax for reticulocytes can result in parasite densities much lower than the LODs of the conventional diagnostic methods. Recently, the creation of ultrasensitive RDTs has lowered the LOD for P. falciparum infection [63, 64] but one targeting P. vivax is still lacking.

In malaria elimination settings, malaria prevalence is often assessed through active case detection in the form of cross-sectional surveillance. Most available clinical diagnostic tools are inadequate for detecting P. vivax asymptomatic reservoirs with very low parasitemia. Molecular methods although sensitive to low-density infections and useful in epidemiological surveillance are not feasible in field applications [19]. The presence of microscopically subpatent infections in endemic populations may render a mass screening and treatment-based strategy ineffective if screening is based on low-sensitivity tools [65].

Hypnozoites pose a significant challenge for the elimination of vivax malaria because they defy detection by any diagnostic methods. Recently, a screen for IgG responses to a panel of 342 P. vivax antigens in longitudinal clinical cohorts established that antibody responses to eight proteins detected P. vivax infections in the previous 9 months with 80% sensitivity and specificity [66]. Modeling demonstrates that treating a serologically positive population could potentially reduce P. vivax prevalence by 59–69%. While this new development still awaits prospective evaluation, it offers a promising surrogate marker for hypnozoite detection and treatment.

Advertisement

6. Chemotherapy and drug resistance

Most antimalarial drugs in use are blood schizontocides that kill asexual blood-stage parasites, which are associated with clinical symptoms. The ability of P. vivax (and also P. ovale spp.) to form liver hypnozoites capable of causing relapses requires the addition of a hypnozoitocide to prevent relapses. For the radical cure of P. vivax malaria, CQ and PQ have been the companion therapies of choice for the treatment of uncomplicated vivax malaria since the 1950s. Due to the development of CQ resistance in the island of New Guinea, CQ was abandoned and replaced with an artemisinin combination therapy (ACT) there [67].

6.1 Treatment of P. vivax blood-stage infections

For the treatment of blood-stage uncomplicated P. vivax malaria, WHO recommends the use of either an ACT or CQ in areas where parasites remain CQ sensitive (CQS) or an ACT in areas where P. vivax is known to be CQ resistant (CQR) [68]. ACTs are contraindicated in pregnant women in their first trimester; thus for this patient population, uncomplicated vivax malaria is treated with either CQ for CQS malaria or quinine for CQR malaria. WHO recommends parenteral therapy for severe malaria with either artesunate, artemether, or quinine (listed here in the order of preference) for at least 24 h regardless of the causative Plasmodium species [68]. No additional drugs are needed to block transmission (as compared to the recommended low-dose PQ for blocking the transmission of P. falciparum) because P. vivax gametocytes are sensitive to most antimalarial drugs.

6.1.1 Chloroquine and unified treatment with ACTs

CQ remains the mainstay treatment for P. vivax malaria in most endemic countries. If low-grade or sporadic CQ resistance is identified, optimizing the treatment regimen can improve the therapeutic efficacy of CQ. A recent meta-analysis of CQ efficacy studies indicates underdosing (<25 mg of CQ/kg) among a substantial proportion (>30%) of patients [21]. Increasing the recommended dose to 30 mg/kg, especially in children under 5 years, could reduce the risk of early recurrence by more than 40% if CQ is used alone. The safety and tolerability of the increased CQ dose are substantiated by earlier studies where CQ doses of 50 mg/kg were used to treat CQR P. falciparum [69]. In addition to underdosing, there is accumulating evidence of emerging CQR parasites in endemic sites [70, 71]. A meta-analysis of 129 clinical trials on CQ efficacy identified CQR P. vivax parasites in most vivax-endemic areas, though the prevalence of resistance varied geographically [72]. The epicenter of CQR P. vivax is located on the island of New Guinea, where the CQR parasite was first reported in 1989 [73]. Reports of high rates of recurrent parasitemia within 28 days in subsequent years [74, 75, 76]—consistent with the WHO definition for RI resistance [77]—led to the ultimate withdrawal of CQ from treating vivax malaria in New Guinea [67]. ACTs have shown high efficacy as a treatment replacement of CQ for uncomplicated vivax malaria in many endemic sites [78, 79, 80]. Dihydroartemisinin-piperaquine treatment had a significantly lower risk of P. vivax recurrence at day 42 than artemether-lumefantrine [81]. These higher rates of recurrence are probably due to different pharmacokinetic profiles of the partner drugs: lumefantrine has a much shorter half-life (~4 days) than piperaquine (28–35 days) and thus offers less protection against early relapse and/or reinfection. This can be mitigated by the inclusion of PQ in the treatment [81]. In areas co-endemic for both P. vivax and P. falciparum, the deployment of a unified ACT-based strategy for both parasites provides several advantages [78]. First, the excellent clinical efficacy of ACTs against vivax malaria makes them highly suitable for areas of known or suspected CQR vivax. Second, it offers operational ease in routine practice where species misdiagnosis is a frequent issue [18]. The World Malaria Report 2020 indicated an increasing number of countries adopting ACTs as first-line therapy for P. vivax [82]. The reluctance to change the treatment may be due to the perceived ease of treating vivax malaria and the economic burden associated with the switch to a much more expensive drug.

6.1.2 Chloroquine resistance

To ensure the high efficacy of first-line therapy, close monitoring is essential. For P. falciparum malaria, this is typically done by clinical efficacy studies, in vitro drug assays, and molecular surveillance of resistance-conferring genetic markers. Surveillance for P. vivax resistance relies heavily on in vivo assessment of the schizontocidal therapy, conducted through follow-ups of recurrent P. vivax parasitemia after initial treatment for 28 days. Extending the follow-up to 42 days will allow the identification of late recrudescence [21]. After the standard treatment with CQ (3-day regimen of 25 mg/kg CQ base), the blood concentration of the active drugs (CQ and desethyl CQ) reaches the minimum inhibitory concentration (MIC, ~100 ng/ml) around 28 days, and thus recurrent parasitemia before day 28, regardless of the origin of the parasites (recrudescence, relapse or new infection), is likely due to CQR parasites [83]. Given that drug resistance is defined as the growth of the parasite in the presence of the drug above the MIC, CQ resistance must be confirmed by measurement of residual blood CQ and desethyl CQ levels on the day of recurrence.

Ex vivo measurement of drug sensitivity has been conducted in many endemic regions, but it is not ideal for routinely monitoring antimalarial drug resistance in P. vivax because of the difficulties in setting up the ex vivo assays [84, 85]. Since a long-term in vitro culture system is not available for P. vivax, ex vivo assays are restricted to one-time assays using fresh field isolates, making further validation of results difficult. The P. vivax tropism for reticulocytes means that reinvasion does not happen frequently under field conditions. As a result, ex vivo drug exposure is limited to one intraerythrocytic cycle. The most commonly used method to quantify parasite growth is the modified Rieckman’s microtest that compares schizont maturation rates [86, 87]. Another method quantifies the production of lactate dehydrogenase by the parasites [88]. Unlike P. falciparum clinical samples where parasites are all at the ring stage, P. vivax clinical isolates contain mixed stages with various degrees of synchronicity. Since P. vivax trophozoites are highly tolerant to CQ [89, 90], ex vivo assays for CQ need to be done using isolates with no less than 80% ring stages. Despite the variability of assay results between labs, ex vivo assays can complement in vivo studies to follow temporal changes of drug sensitivities in an endemic area [91].

Molecular surveillance of putative CQR markers in P. vivax populations, though conducted in multiple endemic sites, is hindered by the lack of understanding of the genetic basis of resistance [71]. Initial studies focused on the orthologs of the pfcrt and pfmdr1, the main determinants of CQ resistance in P. falciparum. Most studies fail to show a strong correlation between pvcrt-o mutations and the CQR phenotype. Some studies from the Brazilian Amazon indicated an association of CQ resistance with higher expression level and gene amplification of pvcrt-o [92, 93], whereas such a link was not validated in Papua Indonesia with high-grade CQ resistance [94]. The relationship between the upregulation of pvcrt-o expression and CQ resistance was recently supported by a genetic cross of P. vivax strains [95]. There are also considerable controversies about the main pfmdr1 mutations Y976F and F1076L as potentially conferring CQ resistance [96, 97, 98, 99], suggesting that pvmdr1 may not be a major determinant for CQ resistance in P. vivax. Population genomics studies of P. vivax populations from areas with drastically different CQ resistance have identified genomic sites under strong selection [100, 101], but their significance in mediating drug resistance remains to be determined. When using Plasmodium knowlesi as an in vitro model, some of the markers did not seem to change the drug sensitivity phenotypes in transgenic parasites [102].

6.2 Treatment of P. vivax liver stages

6.2.1 Primaquine and tafenoquine in anti-relapse therapy

Relapses from hypnozoite reactivation are preventable by anti-relapse therapy with 8-aminoquinoline drugs. For the prevention of relapse, WHO recommends a dose of 0.25–0.5 mg/kg body weight of PQ daily for 14 days [68]. A high dose (0.5 mg/kg/day) is needed for tropical, frequently relapsing P. vivax strains such as the Chesson strain that is prevalent in East Asia and Oceania, whereas a lower dose (0.25 mg/kg/day) is recommended for temperate strains. Many nations adopt low-dose PQ for fear of possible harm to unscreened patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency, but suboptimal dosing may fail to prevent relapses in many endemic sites [46, 103]. Conversely, the high dose requires more detailed clinical investigations to document its efficacy [104].

The 14-day PQ treatment regimen is inevitably associated with poor adherence, which seriously compromises its public health benefit. It is estimated that the effectiveness of unsupervised PQ regimens in vivax patients from Papua, Indonesia, could be as low as 12% [105]. Unfortunately, a shortened 5-day regimen of 15 mg daily PQ did not efficaciously prevent relapse [106, 107]. However, a 7-day high PQ dose (1.0 mg/kg/day) regimen in Asia and Africa recently performed comparably to the 14-day PQ regimen (0.5 mg/kg/day), providing a possible solution to poor adherence [108, 109]. In 2018, another 8-aminoquinoline, tafenoquine (TQ), was approved by the U.S. Food and Drug Administration for radical cure of vivax malaria and malaria prophylaxis [110, 111]. TQ, administered as a single 300 mg dose, showed similar tolerability and efficacy to PQ in preventing relapse in vivax malaria [112, 113]. While TQ appears to be the best choice for travel medicine in people with normal G6PD activity, further clinical studies are needed before seeing its deployment in endemic regions.

In patients with known G6PD deficiency, PQ may be given at 0.75 mg/kg for eight weeks [68], but this should be under close medical supervision with ready access to blood transfusion services. This dosing regimen leverages the “total dose effect”, discovered in the relapsing monkey malaria parasite P. cynomolgi [114]—it posits the same efficacious dose of PQ can be delivered in a range of schedules to achieve the same therapeutic effect. For the temperate and tropical strains, the total dose equals 210 and 420 mg PQ, respectively [115]. This regimen was tested in those carrying the G6PD A- variant experimentally infected with the Chesson strain and found to be safe and efficacious [116]. A recent trial of this regimen found that 5/18 (27.7%) G6PD deficient patients experienced >25% fractional drops in their hemoglobin concentrations, including one patient requiring transfusion [117, 118]. This study precludes the use of unsupervised weekly PQ in Cambodia (and perhaps other parts of the Greater Mekong subregion), where the regional prevalence of Viangchan (a class II G6PD variant) and other hemoglobinopathies such as hemoglobin E and β-thalassemia may predispose G6PD deficient patients to a greater risk of acute hemolytic anemia (AHA) when treated with PQ.

6.2.2 G6PD deficiency and point-of-care testing

The root problem of PQ and TQ is hemolytic toxicity in patients with G6PD deficiency [115, 119, 120]. The G6PD gene is extraordinarily polymorphic with at least 217 known mutations, and their effects on the stability and catalytic efficiency of the enzyme vary greatly [121, 122, 123]. The residual enzyme activity varies from 5–10% of the normal levels in the G6PD A- variant from Africa to less than 1% of the normal levels in the G6PD Mediterranean variant. As a result, the clinical spectrum of PQ toxicity can range from relatively mild and self-limiting in G6PD A- individuals to severe AHA in the G6PD Mediterranean individuals, while most variants from Southeast Asia (the Mahidol, Viangchan, and Canton variants) typically have intermediate levels of enzyme activity [124]. Evidence supporting protective advantages of the G6PD A- against P. falciparum [125, 126, 127, 128] and G6PD Mahidol against P. vivax [129, 130] is consistent with the wide geographic distribution of G6PD deficiency and its overlap with malaria distribution [124]. G6PD deficiency affects around 8% of the global population, but its distribution is geographically heterogeneous and can range from 3 to 30% in tropical areas [131, 132, 133]. G6PD is X-linked; thus, male hemizygotes and female homozygotes have full expression of the G6PD deficiency, whereas heterozygous females display varying degrees of G6PD activity due to random X-chromosome inactivation (lyonization) resulting in red cell mosaicism. As a result, the male population displays two distinctive phenotypes, whereas the female population shows a full spectrum of G6PD activity, which has significant ramifications for the treatment with the 8-aminoquinoline drugs [134]. Because of this, cases of severe AHA have been identified in female heterozygotes receiving the high daily dose of PQ (1.0 mg/kg) even though these subjects tested as G6PD normal after screening with the qualitative fluorescent spot test (FST) [108, 109, 135]. Furthermore, even in vivax patients with a class III G6PD variant (e.g., the Mahidol variant considered mildly deficient), a low-dose PQ treatment (15 mg/kg/day) for three days could lead to AHA, requiring blood transfusions or even renal failure [117, 136, 137]. Therefore, for the goal of malaria elimination in areas with P. vivax, the deployment of point-of-care G6PD deficiency diagnostics is urgent [138]. Currently, FST is the most common method to screen for G6PD deficiency, which has minimum lab requirements of cold chain and electricity as well as trained personnel. The CareStart™ G6PD RDT (Access Bio) is a point-of-care screen for G6PD deficiency, but the cost (~15 USD) is prohibitive for large-scale implementation in low-resource endemic areas [139]. Of note, qualitative screening with the FST or RDT can detect G6PD deficiency below 30% of normal activity, but cannot reliably diagnose female heterozygotes with an intermediate deficiency (30–70% normal activity). Fortunately, rapidly eliminating PQ with its half-life of 6 h can be prescribed to patients with G6PD activity above 30% of normal activity [111]. However, TQ has a long half-life of 14 days, and the recommended threshold of G6PD activity is set at 70% of normal activity. Thus, for rolling out TQ in endemic areas, more stringent screening of G6PD activity with quantitative tests is needed [111, 140]. A recent cost-effectiveness analysis suggests that TQ may be deployed in endemic areas outside sub-Saharan Africa using a gender-specific strategy where G6PD-normal females can be prescribed a low-dose PQ for 14 days [141]. This approach again centers on the availability of a qualitative G6PD test.

6.2.3 Host cytochrome P450 (CYP) 2D6 activity

Another problem identified recently is that PQ efficacy depends on the host activity of the hepatic cytochrome P450 (CYP) 2D6. Failures of PQ to radically cure have been linked to reduced activity of CYP2D6 [142], which mediates activation of PQ to its active metabolite(s) [143, 144]. Follow-up studies in Indonesia have established CYP2D6-dependent metabolism of PQ as a key determinant of the efficacy against relapse [145]. Studies in Brazil similarly identified an association of the diplotype-based CYP2D6 activity score of ≤1.0 with increased risks of P. vivax recurrence within 180 days after PQ treatment [146, 147]. There are also cases of patients with impaired CYP2D6 activity suffering from multiple relapse attacks despite receiving adequate anti-relapse therapy with PQ [148, 149]. Even for the single-low-dose PQ used as a transmission-reducing strategy for P. falciparum, the genetic variation in CYP2D6 affects the pharmacokinetics of PQ [150], and CYP2D6 poor/intermediate metabolism is associated with prolonged gametocyte carriage [151]. Thus, it is important to determine the extent to which reduced CYP2D6 activity is responsible for PQ failures in the radical cure of vivax malaria [152].

CYP2D6 is involved in the metabolism of as many as 25% of drugs in clinical use and is also highly polymorphic [153, 154]. Over 150 CYP2D6 allelic variants have been found and grouped into four phenotypic classes of non-functional, low, normal, and increased metabolizers, with respective activity scores of 0, 0.5, 1.0, and 2.0 per allele, corresponding to diplotype activity scores of 0, 0.5, 1.0, 1.5, 2.0, and > 2.0 [155]. Individuals with diplotype activity scores of ≤1.0 are considered to be poor PQ metabolizers [156]. The proportion of poor PQ metabolizers varies geographically. In the Brazilian Amazon, ~25% of the population was found to have reduced CYP2D6 activity [146, 147]. In Cambodia, 52 and 1% of subjects were found to have intermediate and poor metabolism, respectively [157]. Most surveys are based on genotyping results, whereas direct measurement of the CYP2D6 activity using CYP2D6 substrate metabolism (dextromethorphan to dextrorphan conversion) could more accurately determine the phenotype [145]. While CYP2D6 genotypes are not routinely screened in malaria-endemic areas, knowledge of the extent of this problem will help plan for vivax elimination.

6.2.4 Primaquine resistance

PQ-resistant P. vivax hypnozoites have never been unequivocally demonstrated. PQ efficacy studies are complicated to conduct and possibly one reason PQ resistant parasites have not been identified. PQ alone has excellent anti-relapse activity [158], but co-administration of a schizontocide (e.g., quinine, CQ, or an ACT) has been shown to significantly potentiate PQ’s anti-relapse activity [159]. This effect has been recently verified using an in vitro P. cynomolgi hepatic system, wherein CQ could enhance PQ’s activity against schizonts and hypnozoites by ~18-fold [160]. Any therapeutic failures of PQ in P. vivax radical cure could plausibly result from reasons other than PQ resistance. For example, treatment may fail because of improper PQ dose, short duration of treatment, or poor adherence to the treatment regimen [80, 161, 162]. Further, with the current genotyping strategy, it is not possible to reliably determine whether a recurrent infection after day 28 is due to relapse or reinfection. In endemic areas, patients can harbor multiple genotypically different hypnozoites and a relapse infection may be from reactivation of a heterologous hypnozoite clone [163, 164]. Likewise, a genotype different from that of the primary infection may be from either relapse or reinfection. PQ efficacy studies require longer follow-up, making it difficult to exclude the possibility of reinfection in endemic areas. In studies where the possibility of reinfection can be excluded [158, 165], PQ failure requires further scrutiny, especially with the newly identified CYP2D6 effect. In 21 Indonesian patients who experienced therapeutic failure of PQ against P. vivax relapse, 20 were classified as CYP2D6 impaired, whereas only one with normal CYP2D6 activity and adequate PQ exposure may represent true resistance to PQ [145]. Ultimately, PQ resistance may still be rare in most endemic areas, though continued surveillance is recommended.

Advertisement

7. Vector control

P. vivax produces transmissible gametocytes early in infection before the development of clinical symptoms [166, 167, 168], allowing ready transmission through mosquito vectors with more efficient transmission in certain species [169, 170]. There are 71 Anopheles species/species complexes that are potential vectors for vivax malaria [171], and vector control is a critical component for integrated control of vivax malaria [172]. Long-lasting insecticide-treated nets (LLINs) and indoor residual spraying (IRS) are the key vector-based malaria interventions that are highly effective in sub-Saharan Africa [173, 174]. However, these measures are either under-utilized with low coverage or less effective in certain regions [82, 175]. Since many vector species exhibit early evening and outdoor biting preferences, LLINs and IRS alone are not sufficient for interrupting malaria transmission [176]. In malaria elimination settings, residual transmission often occurs in a forested habitat that lacks core mosquito control coverage [177, 178], requiring targeted vector control efforts for special populations. Further, the emergence and spread of insecticide resistance compromising the efficacy of mosquito control measures needs continual monitoring [179, 180, 181]. Successful malaria elimination programs in various regions of the world have all included vector control as one of their pillar strategies [16]. Thus, novel vector control approaches are desperately needed including larval control strategies [182, 183], incorporation of ivermectin in the mass drug administration (MDA) program to reduce the life span of mosquitoes [184, 185], topical and spatial repellents [186, 187], genetically manipulated mosquitoes for population replacement [188], and next-generation LLINs and IRS [189].

Advertisement

8. Technologies and strategies for supporting elimination

8.1 Experience gleaned from successful stories

Despite the unique challenges posed by P. vivax, elimination is achievable with integrated control measures. There are nearly 40 countries and territories that have been WHO certified as malaria-free, with 10 of those achieving certification since the turn of the century. Although they all used a combination of strategies including vector control, case management, and mass drug administration, different regions emphasized specific sets of tools at different stages of elimination. The Maldives, the first country in SE Asia to reach malaria-free status, relied heavily on vector control [190]. For Sri Lanka’s battle against malaria, strong surveillance, case detection, and patient isolation with treatment were key to its highly targeted elimination strategy [191]. Sri Lanka had anti-relapse treatment as a component of its elimination plan, especially for highly mobile military members, engendering the elimination of P. vivax almost simultaneously with P. falciparum [191]. In the republics of the former Soviet Union, preventive therapy and MDA with PQ, seasonal CQ chemoprophylaxis, and IRS, were instrumental for malaria elimination [16, 192]. China eliminated indigenous malaria cases in 2017 after the declaration of the National Malaria Elimination Action Plan in 2010 [193]. In the final stage of malaria elimination, China adopted targeted MDA to eliminate vivax transmission in central provinces and developed a rigorous 1–3-7 malaria surveillance strategy [194, 195].

8.2 Strategies for vivax elimination

Management of clinical vivax malaria. Accurate diagnosis using sensitive methods is critical for proper treatment of vivax cases. More sensitive diagnostics such as the uRDTs under development may fill such a need. For the treatment of blood stages, a unified treatment with ACTs is highly recommended. The deployment of point-of-care diagnostics for G6PD deficiency will ensure the wider prescription of the anti-relapse drug PQ. For those patients with G6PD deficiency, monthly presumptive treatment or prophylaxis with a drug with a long half-life such as naphthoquine may be adopted, as it has proven to be safe and 100% effective for preventing relapse malaria parasite [196, 197].

Targeted MDA. As mass screenings and treatment-based strategies are ineffective for the final elimination phase [65], residual transmission requires targeted MDA to eliminate asymptomatic and submicroscopic parasite reservoirs. For the success of MDA, better knowledge of malaria epidemiology and strong community engagement are needed. In areas such as the Greater Mekong subregion, G6PD deficiency and CYP2D6 poor metabolizers are prevalent and may account for 30% of the population. In addition, point-of-care diagnostics for G6PD deficiency are not available, which seriously undermines the feasibility of PQ-based MDA [156]. In these regions, periodic MDA with an ACT or prophylaxis drug combination with a long eliminating half-life may be an effective alternative [196, 197, 198, 199]. Incorporation of ivermectin in the MDA program can reduce the life span of adult mosquitoes and in turn, the transmission of the parasite [184, 185].

Vector control. Traditional control methods such as LLIN and IRS need to be implemented with high coverage. This can be supplemented with novel vector control approaches such as larval control [183]. Topical and spatial repellents [186, 187] may be used for populations at higher risk of outdoor transmission.

Surveillance system. The establishment of a stringent malaria surveillance system in the NMCPs that allows timely responses to new malaria incidence plays a crucial role in malaria elimination. This has proved highly important for many, if not all, successful malaria elimination stories. Within this system, training and capacity building are necessary to establish a malaria control network responding effectively to emerging malaria cases.

8.3 Sustaining elimination

With increased international and cross-border travel, imported malaria cases re-introduce malaria in countries where malaria has been eliminated [193, 200], potentially leading to local transmission [58]. Weakening malaria control programs have been linked to almost all resurgence events such as one that occurred in central China [201], and resource concerns are a large contributing factor [202]. Targeted elimination programs (including regular screening and extensive vector management) can be costly, and there remains a concern that countries who have achieved elimination status may be tempted to reduce their targeted vigilance in order to prioritize funding for other endeavors [203]. However, vector control programs are vital to multiple infectious disease programs, which makes them key components of an integrated response. Additionally, countries should continue to train medical workers for the diagnosis and treatment of malaria and remain vigilant to malaria re-introduction from international travelers or mobile communities. Experience from the malaria program in South Korea demonstrated the significance of good case management practice combined with stringent surveillance for reducing the resurgent malaria threat [204]. In many malaria-free nations, chemoprophylaxis is suggested for international travelers [191, 205], and introduced cases are met with an investigation to eliminate the possibility of endemic spread [206].

Advertisement

9. Conclusions

Several unique biological characters of the P. vivax parasite are responsible for its wide distribution and persistence in the face of escalating control efforts. Table 2 summarizes the challenges in controlling and eliminating vivax malaria and potential solutions. Because of the geographical variation of the vivax malaria situation, different endemic countries are likely to emphasize one or a few control strategies. As an infectious disease involving the human-parasite-vector triad, it requires integrated approaches targeting all components of these interactions for the ultimate elimination of vivax malaria.

ProblemsSolutions
Relapse from reactivation of dormant hypnozoites; long-latency strains allow for parasite survival beyond winter seasonTreatment of hypnozoite stage; sustained vector control to prevent transmission
Anti-relapse treatment with PQ or TQ increases risk of acute hemolytic anemia in G6PD deficient patients with varying degrees in male vs. femaleDeployment of point-of-care G6PD RDTs and gender-sensitive treatment strategies that account for differing phenotypic presentation of G6PD deficiency.
Impaired activity of CYP2D6 is associated with poor PQ efficacyScreening for impairment in CYP2D6 activity in regions of PQ treatment failure can inform targeted treatment strategy
Lack of radical treatment for patients with G6PD deficiency, low CYP2D6 activity, and PQ contraindicationProphylaxis with a safe drug with a long half-life (e.g., naphthoquine)
Suboptimal dosing in CQ treatment of uncomplicated malariaStandardizing dose to 30 mg/kg, especially in children under five years can prevent early recurrence
CQ resistance in many endemic sitesTreatment with ACTs
Lack of RDTs for detecting asymptomatic reservoirs with low parasitemiaDetecting antibodies to specific P. vivax proteins followed by treatment may reduce P. vivax prevalence
Residual transmission from asymptomatic and submicroscopic parasite reservoirsTargeted MDA following epidemiological assessment and community engagement
Vector control strategies such as LLINs and IRS have low coverage, decreased adherence and limited efficacy in some regionsIncreasing coverage of LLINs and IRS. Implementing novel vector control approaches to decrease mosquito density, lifespan, and outdoor transmission
Single vertical strategy to eliminate malaria remains unsuccessfulCombined strategies of strong surveillance, early case detection, patient isolation with treatment, sustained vector control.

Table 2.

Summary of the problems and solutions in eliminating P. vivax malaria.

Advertisement

Acknowledgments

We thank the National Institute of Allergy and Infectious Diseases, NIH, for financial support (U19AI089672).

References

  1. 1. Li XH, Kondrashin A, Greenwood B, et al. A Historical Review of WHO Certification of Malaria Elimination. Trends Parasitol. 2019;35:163-171.
  2. 2. Pattanayak S, Sharma VP, Kalra NL, et al. Malaria paradigms in India and control strategies. Indian J Malariol. 1994;31:141-199.
  3. 3. Baird JK. Essential guidance on malaria elimination in its history. J Vector Borne Dis. 2019;56:11-14.
  4. 4. WHO. World Malaria Report 2019. https://www.who.int/publications-detail/world-malaria-report-2019. 2019.
  5. 5. Millar SB, Cox-Singh J. Human infections with Plasmodium knowlesi--zoonotic malaria. Clin Microbiol Infect. 2015;21:640-648.
  6. 6. Lover AA, Baird JK, Gosling R, et al. Malaria elimination: time to target all species. Am J Trop Med Hyg. 2018;99:17-23.
  7. 7. Cotter C, Sturrock HJ, Hsiang MS, et al. The changing epidemiology of malaria elimination: new strategies for new challenges. Lancet. 2013;382:900-911.
  8. 8. Battle KE, Lucas TCD, Nguyen M, et al. Mapping the global endemicity and clinical burden of Plasmodium vivax, 2000-17: a spatial and temporal modelling study. Lancet. 2019;394:332-343.
  9. 9. Kahle D, Wickham H. ggmap: Spatial Visualization with ggplot2. The R Journal. 2013;5:144-161.
  10. 10. Howes RE, Patil AP, Piel FB, et al. The global distribution of the Duffy blood group. Nat Commun. 2011;2:266.
  11. 11. Miller LH, Mason SJ, Clyde DF, et al. The resistance factor to Plasmodium vivax in blacks. The Duffy-blood-group genotype, FyFy. N Engl J Med. 1976;295:302-304.
  12. 12. Popovici J, Roesch C, Rougeron V. The enigmatic mechanisms by which Plasmodium vivax infects Duffy-negative individuals. PLoS Pathog. 2020;16:e1008258.
  13. 13. Gunalan K, Niangaly A, Thera MA, et al. Plasmodium vivax infections of Duffy-negative erythrocytes: Historically undetected or a recent adaptation? Trends Parasitol. 2018;34:420-429.
  14. 14. Howes RE, Reiner RC, Jr., Battle KE, et al. Plasmodium vivax Transmission in Africa. PLoS Negl Trop Dis. 2015;9:e0004222.
  15. 15. Twohig KA, Pfeffer DA, Baird JK, et al. Growing evidence of Plasmodium vivax across malaria-endemic Africa. PLoS Negl Trop Dis. 2019;13:e0007140.
  16. 16. Shanks GD. Control and elimination of Plasmodium vivax. Adv Parasitol. 2012;80:301-341.
  17. 17. Cui L, Cao Y, Kaewkungwal J, et al. Malaria Elimination in the Greater Mekong Subregion: Challenges and Prospects. In: Manguin S, Dev V, editors. Towards Malaria Elimination: A Leap Forward: IntechOpen; 2018. p. 179-200.
  18. 18. Geng J, Malla P, Zhang J, et al. Increasing trends of malaria in a border area of the Greater Mekong Subregion. Malar J. 2019;18:309.
  19. 19. Lin E, Kiniboro B, Gray L, et al. Differential patterns of infection and disease with P. falciparum and P. vivax in young Papua New Guinean children. PLoS One. 2010;5:e9047.
  20. 20. Mueller I, Galinski MR, Tsuboi T, et al. Natural acquisition of immunity to Plasmodium vivax: epidemiological observations and potential targets. Adv Parasitol. 2013;81:77-131.
  21. 21. Commons RJ, Simpson JA, Thriemer K, et al. The effect of chloroquine dose and primaquine on Plasmodium vivax recurrence: a WorldWide Antimalarial Resistance Network systematic review and individual patient pooled meta-analysis. Lancet Infect Dis. 2018;18:1025-1034.
  22. 22. Li N, Parker DM, Yang Z, et al. Risk factors associated with slide positivity among febrile patients in a conflict zone of north-eastern Myanmar along the China-Myanmar border. Malar J. 2013;12:361.
  23. 23. Corder RM, de Lima ACP, Khoury DS, et al. Quantifying and preventing Plasmodium vivax recurrences in primaquine-untreated pregnant women: An observational and modeling study in Brazil. PLOS Neglected Tropical Diseases. 2020;14:e0008526.
  24. 24. Brummaier T, Gilder ME, Gornsawun G, et al. Vivax malaria in pregnancy and lactation: a long way to health equity. Malar J. 2020;19:40.
  25. 25. Tao ZY, Fang Q, Liu X, et al. Congenital malaria in China. PLoS Negl Trop Dis. 2014;8:e2622.
  26. 26. Sandfort M, Vantaux A, Kim S, et al. Forest malaria in Cambodia: the occupational and spatial clustering of Plasmodium vivax and Plasmodium falciparum infection risk in a cross-sectional survey in Mondulkiri province, Cambodia. Malar J. 2020;19:413.
  27. 27. Adams JH, Mueller I. The Biology of Plasmodium vivax. Cold Spring Harb Perspect Med. 2017;7: a025585.
  28. 28. Anstey NM, Douglas NM, Poespoprodjo JR, et al. Plasmodium vivax: clinical spectrum, risk factors and pathogenesis. Adv Parasitol. 2012;80:151-201.
  29. 29. Baird JK. Evidence and implications of mortality associated with acute Plasmodium vivax malaria. Clin Microbiol Rev. 2013;26:36-57.
  30. 30. Kotepui M, Kotepui KU, Milanez GJ, et al. Prevalence and risk factors related to poor outcome of patients with severe Plasmodium vivax infection: a systematic review, meta-analysis, and analysis of case reports. BMC Infect Dis. 2020;20:363.
  31. 31. Tjitra E, Anstey NM, Sugiarto P, et al. Multidrug-resistant Plasmodium vivax associated with severe and fatal malaria: a prospective study in Papua, Indonesia. PLoS Med. 2008;5:e128.
  32. 32. Rahimi BA, Thakkinstian A, White NJ, et al. Severe vivax malaria: a systematic review and meta-analysis of clinical studies since 1900. Malar J. 2014;13:481.
  33. 33. Malleret B, Li A, Zhang R, et al. Plasmodium vivax: restricted tropism and rapid remodeling of CD71-positive reticulocytes. Blood. 2015;125:1314-1324.
  34. 34. McQueen PG, McKenzie FE. Age-structured red blood cell susceptibility and the dynamics of malaria infections. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:9161-9166.
  35. 35. Cromer D, Stark J, Davenport MP. Low red cell production may protect against severe anemia during a malaria infection—Insights from modeling. Journal of Theoretical Biology. 2009;257:533-542.
  36. 36. Douglas NM, Lampah DA, Kenangalem E, et al. Major burden of severe anemia from non-falciparum malaria species in Southern Papua: a hospital-based surveillance study. PLoS Med. 2013;10:e1001575; discussion e1001575.
  37. 37. Kochar DK, Das A, Kochar A, et al. Thrombocytopenia in Plasmodium falciparum, Plasmodium vivax and mixed infection malaria: a study from Bikaner (Northwestern India). Platelets. 2010;21:623-627.
  38. 38. Tanwar GS, Khatri PC, Chahar CK, et al. Thrombocytopenia in childhood malaria with special reference to P. vivax monoinfection: A study from Bikaner (Northwestern India). Platelets. 2012;23:211-216.
  39. 39. Nosten F, McGready R, Simpson JA, et al. Effects of Plasmodium vivax malaria in pregnancy. Lancet. 1999;354:546-549.
  40. 40. Pincelli A, Neves PAR, Lourenco BH, et al. The hidden burden of Plasmodium vivax malaria in pregnancy in the Amazon: an observational study in northwestern Brazil. Am J Trop Med Hyg. 2018.
  41. 41. Markus MB. Dormancy in mammalian malaria. Trends Parasitol. 2012;28:39-45.
  42. 42. Obaldia N, 3rd, Meibalan E, Sa JM, et al. Bone marrow is a major parasite reservoir in Plasmodium vivax infection. MBio. 2018;9.
  43. 43. Markus MB. Biological concepts in recurrent Plasmodium vivax malaria. Parasitology. 2018:1-7.
  44. 44. Lover AA, Coker RJ. Quantifying effect of geographic location on epidemiology of Plasmodium vivax malaria. Emerg Infect Dis. 2013;19:1058-1065.
  45. 45. Kondrashin AV, Morozova LF, Stepanova EV, et al. On the epidemiology of Plasmodium vivax malaria: past and present with special reference to the former USSR. Malar J. 2018;17:346.
  46. 46. White NJ. Determinants of relapse periodicity in Plasmodium vivax malaria. Malar J. 2011;10:297.
  47. 47. Yang BL, Wan WJ, Wang WR, et al. [Experimental studies on the biological characteristics of Plasmodium vivax in south Yunnan]. Ji Sheng Chong Xue Yu Ji Sheng Chong Bing Za Zhi. 1986;4:101-105.
  48. 48. Hollingdale MR, Collins WE, Campbell CC, et al. In vitro culture of two populations (dividing and nondividing) of exoerythrocytic parasites of Plasmodium vivax. Am J Trop Med Hyg. 1985;34:216-222.
  49. 49. Hollingdale MR, Collins WE, Campbell CC. In vitro culture of exoerythrocytic parasites of the North Korean strain of Plasmodium vivax in hepatoma cells. Am J Trop Med Hyg. 1986;35:275-276.
  50. 50. Shanks GD, White NJ. The activation of vivax malaria hypnozoites by infectious diseases. Lancet Infect Dis. 2013;13:900-906.
  51. 51. Commons RJ, Simpson JA, Thriemer K, et al. Risk of Plasmodium vivax parasitaemia after Plasmodium falciparum infection: a systematic review and meta-analysis. Lancet Infect Dis. 2019;19:91-101.
  52. 52. Robinson LJ, Wampfler R, Betuela I, et al. Strategies for understanding and reducing the Plasmodium vivax and Plasmodium ovale hypnozoite reservoir in Papua New Guinean children: a randomised placebo-controlled trial and mathematical model. PLoS Med. 2015;12:e1001891.
  53. 53. Adekunle AI, Pinkevych M, McGready R, et al. Modeling the dynamics of Plasmodium vivax infection and hypnozoite reactivation in vivo. PLoS Negl Trop Dis. 2015;9:e0003595.
  54. 54. Betuela I, Rosanas-Urgell A, Kiniboro B, et al. Relapses contribute significantly to the risk of Plasmodium vivax infection and disease in Papua New Guinean children 1-5 years of age. J Infect Dis. 2012;206:1771-1780.
  55. 55. Commons RJ, Simpson JA, Watson J, et al. Estimating the proportion of Plasmodium vivax recurrences caused by relapse: a systematic review and meta-analysis. Am J Trop Med Hyg. 2020;103:1094-1099.
  56. 56. Bright AT, Manary MJ, Tewhey R, et al. A high resolution case study of a patient with recurrent Plasmodium vivax infections shows that relapses were caused by meiotic siblings. PLoS Negl Trop Dis. 2014;8:e2882.
  57. 57. Lin JT, Hathaway NJ, Saunders DL, et al. Using Amplicon Deep Sequencing to Detect Genetic Signatures of Plasmodium vivax Relapse. J Infect Dis. 2015;212:999-1008.
  58. 58. Spanakos G, Snounou G, Pervanidou D, et al. Genetic Spatiotemporal Anatomy of Plasmodium vivax Malaria Episodes in Greece, 2009-2013. Emerg Infect Dis. 2018;24:541-548.
  59. 59. Mekonnen SK, Aseffa A, Medhin G, et al. Re-evaluation of microscopy confirmed Plasmodium falciparum and Plasmodium vivax malaria by nested PCR detection in southern Ethiopia. Malar J. 2014;13:48.
  60. 60. WHO. Control and elimination of Plasmodium vivax malaria. 2015.
  61. 61. Barber BE, William T, Grigg MJ, et al. Limitations of microscopy to differentiate Plasmodium species in a region co-endemic for Plasmodium falciparum, Plasmodium vivax and Plasmodium knowlesi. Malar J. 2013;12:8.
  62. 62. Agarwal R, Choi L, Johnson S, et al. Rapid diagnostic tests for Plasmodium vivax malaria in endemic countries. Cochrane Database Syst Rev. 2020;11:CD013218.
  63. 63. Unwin VT, Ahmed R, Noviyanti R, et al. Use of a highly-sensitive rapid diagnostic test to screen for malaria in pregnancy in Indonesia. Malar J. 2020;19:28.
  64. 64. Yeung S, McGregor D, James N, et al. Performance of Ultrasensitive Rapid Diagnostic Tests for Detecting Asymptomatic Plasmodium falciparum. Am J Trop Med Hyg. 2020;102:307-309.
  65. 65. Sutanto I, Kosasih A, Elyazar IRF, et al. Negligible Impact of Mass Screening and Treatment on Mesoendemic Malaria Transmission at West Timor in Eastern Indonesia: A Cluster-Randomized Trial. Clin Infect Dis. 2018;67:1364-1372.
  66. 66. Longley RJ, White MT, Takashima E, et al. Development and validation of serological markers for detecting recent Plasmodium vivax infection. Nat Med. 2020;26:741-749.
  67. 67. Baird JK. Resistance to chloroquine unhinges vivax malaria therapeutics. Antimicrob Agents Chemother. 2011;55:1827-1830.
  68. 68. WHO. Guidelines for the treatment of malaria. Third edition. 2015:pp. 316.
  69. 69. Ursing J, Rombo L, Bergqvist Y, et al. High-Dose Chloroquine for Treatment of Chloroquine-Resistant Plasmodium falciparum Malaria. J Infect Dis. 2016;213:1315-1321.
  70. 70. Baird JK. Resistance to therapies for infection by Plasmodium vivax. Clin Microbiol Rev. 2009;22:508-534.
  71. 71. Price RN, Auburn S, Marfurt J, et al. Phenotypic and genotypic characterisation of drug-resistant Plasmodium vivax. Trends Parasitol. 2012;28:522-529.
  72. 72. Price RN, von Seidlein L, Valecha N, et al. Global extent of chloroquine-resistant Plasmodium vivax: a systematic review and meta-analysis. Lancet Infect Dis. 2014;14:982-991.
  73. 73. Rieckmann KH, Davis DR, Hutton DC. Plasmodium vivax resistance to chloroquine? Lancet. 1989;2:1183-1184.
  74. 74. Sumawinata IW, Bernadeta, Leksana B, et al. Very high risk of therapeutic failure with chloroquine for uncomplicated Plasmodium falciparum and P. vivax malaria in Indonesian Papua. Am J Trop Med Hyg. 2003;68:416-420.
  75. 75. Baird JK, Basri H, Purnomo, et al. Resistance to chloroquine by Plasmodium vivax in Irian Jaya, Indonesia. Am J Trop Med Hyg. 1991;44:547-552.
  76. 76. Ratcliff A, Siswantoro H, Kenangalem E, et al. Therapeutic response of multidrug-resistant Plasmodium falciparum and P. vivax to chloroquine and sulfadoxine-pyrimethamine in southern Papua, Indonesia. Trans R Soc Trop Med Hyg. 2007;101:351-359.
  77. 77. WHO. Chemotherapy of malaria: Report of the WHO Scientific Group. WHO Technical Report Series No. 375. 1967. Available from: https://apps.who.int/iris/bitstream/handle/10665/40671/WHO_TRS_375.pdf.
  78. 78. Douglas NM, Anstey NM, Angus BJ, et al. Artemisinin combination therapy for vivax malaria. Lancet Infect Dis. 2010;10:405-416.
  79. 79. Gogtay N, Kannan S, Thatte UM, et al. Artemisinin-based combination therapy for treating uncomplicated Plasmodium vivax malaria. Cochrane Database Syst Rev. 2013;10:CD008492.
  80. 80. Baird JK, Valecha N, Duparc S, et al. Diagnosis and Treatment of Plasmodium vivax Malaria. Am J Trop Med Hyg. 2016;95:35-51.
  81. 81. Commons RJ, Simpson JA, Thriemer K, et al. The efficacy of dihydroartemisinin-piperaquine and artemether-lumefantrine with and without primaquine on Plasmodium vivax recurrence: A systematic review and individual patient data meta-analysis. PLoS Med. 2019;16:e1002928.
  82. 82. WHO. World Malaria Report 2020 2020. Available from: https://www.who.int/publications/i/item/9789240015791.
  83. 83. Baird JK, Leksana B, Masbar S, et al. Diagnosis of resistance to chloroquine by Plasmodium vivax: timing of recurrence and whole blood chloroquine levels. Am J Trop Med Hyg. 1997;56:621-626.
  84. 84. Wernsdorfer WH, Tasanor O, Wernsdorfer G. In vitro drug sensitivity testing in Plasmodium vivax. Wien Klin Wochenschr. 2008;120:30-33.
  85. 85. Russell B, Suwanarusk R, Malleret B, et al. Human ex vivo studies on asexual Plasmodium vivax: the best way forward. Int J Parasitol. 2012;42:1063-1070.
  86. 86. Renapurkar DM, Pradhan VR, Sutar NK, et al. Micro test for assaying sensitivity of Plasmodium vivax in vitro. Chemotherapy. 1989;35:160-163.
  87. 87. Russell BM, Udomsangpetch R, Rieckmann KH, et al. Simple in vitro assay for determining the sensitivity of Plasmodium vivax isolates from fresh human blood to antimalarials in areas where P. vivax is endemic. Antimicrob Agents Chemother. 2003;47:170-173.
  88. 88. Druilhe P, Brasseur P, Blanc C, et al. Improved assessment of Plasmodium vivax response to antimalarial drugs by a colorimetric double-site plasmodium lactate dehydrogenase antigen capture enzyme-linked immunosorbent assay. Antimicrob Agents Chemother. 2007;51:2112-2116.
  89. 89. Russell B, Chalfein F, Prasetyorini B, et al. Determinants of in vitro drug susceptibility testing of Plasmodium vivax. Antimicrob Agents Chemother. 2008;52:1040-1045.
  90. 90. Sharrock WW, Suwanarusk R, Lek-Uthai U, et al. Plasmodium vivax trophozoites insensitive to chloroquine. Malar J. 2008;7:94.
  91. 91. Li J, Zhang J, Li Q, et al. 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. 2020;14:e0008255.
  92. 92. Melo GC, Monteiro WM, Siqueira AM, et al. Expression levels of pvcrt-o and pvmdr-1 are associated with chloroquine resistance and severe Plasmodium vivax malaria in patients of the Brazilian Amazon. PLoS One. 2014;9:e105922.
  93. 93. Silva SR, Almeida ACG, da Silva GAV, et al. Chloroquine resistance is associated to multi-copy pvcrt-o gene in Plasmodium vivax malaria in the Brazilian Amazon. Malar J. 2018;17:267.
  94. 94. Pava Z, Handayuni I, Wirjanata G, et al. Expression of Plasmodium vivax crt-o is related to parasite stage but not ex vivo chloroquine susceptibility. Antimicrob Agents Chemother. 2015;60:361-367.
  95. 95. Sa JM, Kaslow SR, Moraes Barros RR, et al. Plasmodium vivax chloroquine resistance links to pvcrt transcription in a genetic cross. Nat Commun. 2019;10:4300.
  96. 96. Suwanarusk R, Russell B, Chavchich M, et al. Chloroquine resistant Plasmodium vivax: in vitro characterisation and association with molecular polymorphisms. PLoS One. 2007;2:e1089.
  97. 97. Orjuela-Sanchez P, de Santana Filho FS, Machado-Lima A, et al. Analysis of single-nucleotide polymorphisms in the crt-o and mdr1 genes of Plasmodium vivax among chloroquine-resistant isolates from the Brazilian Amazon region. Antimicrob Agents Chemother. 2009;53:3561-3564.
  98. 98. Marfurt J, de Monbrison F, Brega S, et al. Molecular markers of in vivo Plasmodium vivax resistance to amodiaquine plus sulfadoxine-pyrimethamine: mutations in pvdhfr and pvmdr1. J Infect Dis. 2008;198:409-417.
  99. 99. Shalini S, Chaudhuri S, Sutton PL, et al. Chloroquine efficacy studies confirm drug susceptibility of Plasmodium vivax in Chennai, India. Malar J. 2014;13:129.
  100. 100. Pearson RD, Amato R, Auburn S, et al. Genomic analysis of local variation and recent evolution in Plasmodium vivax. Nat Genet. 2016;48:959-964.
  101. 101. Brashear AM, Fan Q, Hu Y, et al. Population genomics identifies a distinct Plasmodium vivax population on the China-Myanmar border of Southeast Asia. PLoS Negl Trop Dis. 2020;14:e0008506.
  102. 102. Verzier LH, Coyle R, Singh S, et al. Plasmodium knowlesi as a model system for characterising Plasmodium vivax drug resistance candidate genes. PLoS Negl Trop Dis. 2019;13:e0007470.
  103. 103. John GK, Douglas NM, von Seidlein L, et al. Primaquine radical cure of Plasmodium vivax: a critical review of the literature. Malar J. 2012;11:280.
  104. 104. Milligan R, Daher A, Villanueva G, et al. Primaquine alternative dosing schedules for preventing malaria relapse in people with Plasmodium vivax. Cochrane Database Syst Rev. 2020;8:CD012656.
  105. 105. Douglas NM, Poespoprodjo JR, Patriani D, et al. Unsupervised primaquine for the treatment of Plasmodium vivax malaria relapses in southern Papua: A hospital-based cohort study. PLoS Med. 2017;14:e1002379.
  106. 106. Gogtay NJ, Desai S, Kamtekar KD, et al. Efficacies of 5- and 14-day primaquine regimens in the prevention of relapses in Plasmodium vivax infections. Ann Trop Med Parasitol. 1999;93:809-812.
  107. 107. Yadav RS, Ghosh SK. Radical curative efficacy of five-day regimen of primaquine for treatment of Plasmodium vivax malaria in India. J Parasitol. 2002;88:1042-1044.
  108. 108. Chu CS, Phyo AP, Turner C, et al. Chloroquine versus dihydroartemisinin-piperaquine with standard high-dose primaquine given either for 7 days or 14 days in Plasmodium vivax malaria. Clin Infect Dis. 2019;68:1311-1319.
  109. 109. Taylor WRJ, Thriemer K, von Seidlein L, et al. Short-course primaquine for the radical cure of Plasmodium vivax malaria: a multicentre, randomised, placebo-controlled non-inferiority trial. Lancet. 2019.
  110. 110. Hounkpatin AB, Kreidenweiss A, Held J. Clinical utility of tafenoquine in the prevention of relapse of Plasmodium vivax malaria: a review on the mode of action and emerging trial data. Infect Drug Resist. 2019;12:553-570.
  111. 111. Baird JK. Tafenoquine for travelers' malaria: evidence, rationale and recommendations. J Travel Med. 2018;25.
  112. 112. Lacerda MVG, Llanos-Cuentas A, Krudsood S, et al. Single-dose tafenoquine to prevent relapse of Plasmodium vivax malaria. N Engl J Med. 2019;380:215-228.
  113. 113. Llanos-Cuentas A, Lacerda MVG, Hien TT, et al. Tafenoquine versus primaquine to prevent relapse of Plasmodium vivax malaria. N Engl J Med. 2019;380:229-241.
  114. 114. Schmidt LH, Fradkin R, Vaughan D, et al. Radical cure of infections with Plasmodium cynomolgi: a function of total 8-aminoquinoline dose. Am J Trop Med Hyg. 1977;26:1116-1128.
  115. 115. Baird JK. 8-Aminoquinoline therapy for latent malaria. Clin Microbiol Rev. 2019;32.
  116. 116. Alving AS, Johnson CF, Tarlov AR, et al. Mitigation of the haemolytic effect of primaquine and enhancement of its action against exoerythrocytic forms of the Chesson strain of Piasmodium vivax by intermittent regimens of drug administration: a preliminary report. Bull World Health Organ. 1960;22:621-631.
  117. 117. Kheng S, Muth S, Taylor WR, et al. Tolerability and safety of weekly primaquine against relapse of Plasmodium vivax in Cambodians with glucose-6-phosphate dehydrogenase deficiency. BMC Med. 2015;13:203.
  118. 118. Taylor WRJ, Kheng S, Muth S, et al. Hemolytic dynamics of weekly primaquine antirelapse therapy among Cambodians with acute Plasmodium vivax malaria with or without glucose-6-phosphate dehydrogenase deficiency. J Infect Dis. 2019;220:1750-1760.
  119. 119. Baird JK. Primaquine toxicity forestalls effective therapeutic management of the endemic malarias. Int J Parasitol. 2012;42:1049-1054.
  120. 120. Ashley EA, Recht J, White NJ. Primaquine: the risks and the benefits. Malar J. 2014;13:418.
  121. 121. Gomez-Manzo S, Marcial-Quino J, Vanoye-Carlo A, et al. Glucose-6-Phosphate Dehydrogenase: Update and Analysis of New Mutations around the World. Int J Mol Sci. 2016;17.
  122. 122. Minucci A, Moradkhani K, Hwang MJ, et al. Glucose-6-phosphate dehydrogenase (G6PD) mutations database: review of the "old" and update of the new mutations. Blood cells, molecules & diseases. 2012;48:154-165.
  123. 123. Luzzatto L, Nannelli C, Notaro R. Glucose-6-Phosphate Dehydrogenase Deficiency. Hematol Oncol Clin North Am. 2016;30:373-393.
  124. 124. Howes RE, Dewi M, Piel FB, et al. Spatial distribution of G6PD deficiency variants across malaria-endemic regions. Malar J. 2013;12:418.
  125. 125. Ruwende C, Khoo SC, Snow RW, et al. Natural selection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. Nature. 1995;376:246-249.
  126. 126. Shah SS, Rockett KA, Jallow M, et al. Heterogeneous alleles comprising G6PD deficiency trait in West Africa exert contrasting effects on two major clinical presentations of severe malaria. Malar J. 2016;15:13.
  127. 127. Guindo A, Fairhurst RM, Doumbo OK, et al. X-linked G6PD deficiency protects hemizygous males but not heterozygous females against severe malaria. PLoS Med. 2007;4:e66.
  128. 128. Bienzle U, Ayeni O, Lucas AO, et al. Glucose-6-phosphate dehydrogenase and malaria. Greater resistance of females heterozygous for enzyme deficiency and of males with non-deficient variant. Lancet. 1972;1:107-110.
  129. 129. Louicharoen C, Patin E, Paul R, et al. Positively selected G6PD-Mahidol mutation reduces Plasmodium vivax density in Southeast Asians. Science. 2009;326:1546-1549.
  130. 130. Yi H, Li H, Liang L, et al. 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. 2019;75:103980.
  131. 131. Howes RE, Dewi M, Piel FB, et al. Spatial distribution of G6PD deficiency variants across malaria-endemic regions. Malaria journal. 2013;12:418.
  132. 132. Nkhoma ET, Poole C, Vannappagari V, et al. The global prevalence of glucose-6-phosphate dehydrogenase deficiency: a systematic review and meta-analysis. Blood Cells Mol Dis. 2009;42:267-278.
  133. 133. Howes RE, Piel FB, Patil AP, et al. G6PD deficiency prevalence and estimates of affected populations in malaria endemic countries: a geostatistical model-based map. PLoS Med. 2012;9:e1001339.
  134. 134. Chu CS, Bancone G, Nosten F, et al. Primaquine-induced haemolysis in females heterozygous for G6PD deficiency. Malar J. 2018;17:101.
  135. 135. Chu CS, Bancone G, Moore KA, et al. Haemolysis in G6PD heterozygous females treated with primaquine for Plasmodium vivax malaria: a nested cohort in a trial of radical curative regimens. PLoS Med. 2017;14:e1002224.
  136. 136. Karwacki JJ, Shanks GD, Kummalue T, et al. Primaquine induced hemolysis in a Thai soldier. Southeast Asian J Trop Med Public Health. 1989;20:555-556.
  137. 137. Chen X, He Y, Miao Y, et al. A young man with severe acute haemolytic anaemia. BMJ. 2017;359:j4263.
  138. 138. Domingo GJ, Satyagraha AW, Anvikar A, et al. G6PD testing in support of treatment and elimination of malaria: recommendations for evaluation of G6PD tests. Malar J. 2013;12:391.
  139. 139. Baird JK. Management of Plasmodium vivax risk and illness in travelers. Trop Dis Travel Med Vaccines. 2017;3:7.
  140. 140. Commons RJ, McCarthy JS, Price RN. Tafenoquine for the radical cure and prevention of malaria: the importance of testing for G6PD deficiency. Med J Aust. 2020;212:152-153 e151.
  141. 141. Devine A, Howes RE, Price DJ, et al. Cost-effectiveness analysis of sex-stratified Plasmodium vivax treatment strategies using available G6PD diagnostics to accelerate access to radical cure. Am J Trop Med Hyg. 2020;103:394-403.
  142. 142. Bennett JW, Pybus BS, Yadava A, et al. Primaquine failure and cytochrome P-450 2D6 in Plasmodium vivax malaria. N Engl J Med. 2013;369:1381-1382.
  143. 143. Pybus BS, Marcsisin SR, Jin X, et al. The metabolism of primaquine to its active metabolite is dependent on CYP 2D6. Malar J. 2013;12:212.
  144. 144. Pybus BS, Sousa JC, Jin X, et al. CYP450 phenotyping and accurate mass identification of metabolites of the 8-aminoquinoline, anti-malarial drug primaquine. Malar J. 2012;11:259.
  145. 145. Baird JK, Louisa M, Noviyanti R, et al. Association of Impaired Cytochrome P450 2D6 Activity Genotype and Phenotype With Therapeutic Efficacy of Primaquine Treatment for Latent Plasmodium vivax Malaria. JAMA Netw Open. 2018;1:e181449.
  146. 146. Brasil LW, Rodrigues-Soares F, Santoro AB, et al. CYP2D6 activity and the risk of recurrence of Plasmodium vivax malaria in the Brazilian Amazon: a prospective cohort study. Malar J. 2018;17:57.
  147. 147. Silvino ACR, Kano FS, Costa MA, et al. Novel insights into Plasmodium vivax therapeutic failure: CYP2D6 activity and time of exposure to malaria modulate the risk of recurrence. Antimicrob Agents Chemother. 2020;64.
  148. 148. Ingram RJ, Crenna-Darusallam C, Soebianto S, et al. The clinical and public health problem of relapse despite primaquine therapy: case review of repeated relapses of Plasmodium vivax acquired in Papua New Guinea. Malar J. 2014;13:488.
  149. 149. He X, Pan M, Zeng W, et al. Multiple relapses of Plasmodium vivax malaria acquired from West Africa and association with poor metabolizer CYP2D6 variant: a case report. BMC Infect Dis. 2019;19:704.
  150. 150. Goncalves BP, Pett H, Tiono AB, et al. Age, weight, and CYP2D6 genotype are major determinants of primaquine pharmacokinetics in african children. Antimicrob Agents Chemother. 2017;61:e02590-02516.
  151. 151. Pett H, Bradley J, Okebe J, et al. CYP2D6 polymorphisms and the safety and gametocytocidal activity of single dose primaquine for P. falciparum. Antimicrob Agents Chemother. 2019.
  152. 152. Marcsisin SR, Reichard G, Pybus BS. Primaquine pharmacology in the context of CYP 2D6 pharmacogenomics: Current state of the art. Pharmacol Ther. 2016.
  153. 153. Zhou SF, Liu JP, Lai XS. Substrate specificity, inhibitors and regulation of human cytochrome P450 2D6 and implications in drug development. Curr Med Chem. 2009;16:2661-2805.
  154. 154. Zhou SF. Polymorphism of human cytochrome P450 2D6 and its clinical significance: Part I. Clin Pharmacokinet. 2009;48:689-723.
  155. 155. Gaedigk A, Simon SD, Pearce RE, et al. The CYP2D6 activity score: translating genotype information into a qualitative measure of phenotype. Clin Pharmacol Ther. 2008;83:234-242.
  156. 156. Baird JK, Battle KE, Howes RE. Primaquine ineligibility in anti-relapse therapy of Plasmodium vivax malaria: the problem of G6PD deficiency and cytochrome P-450 2D6 polymorphisms. Malar J. 2018;17:42.
  157. 157. Spring MD, Lon C, Sok S, et al. Prevalence of CYP2D6 genotypes and predicted phenotypes in a cohort of cambodians at high risk for infections with Plasmodium vivax. Am J Trop Med Hyg. 2020;103:756-759.
  158. 158. Nelwan EJ, Ekawati LL, Tjahjono B, et al. Randomized trial of primaquine hypnozoitocidal efficacy when administered with artemisinin-combined blood schizontocides for radical cure of Plasmodium vivax in Indonesia. BMC Med. 2015;13:294.
  159. 159. Alving AS, Arnold J, Hockwald RS, et al. Potentiation of the curative action of primaquine in vivax malaria by quinine and chloroquine. J Lab Clin Med. 1955;46:301-306.
  160. 160. Dembele L, Franetich JF, Soulard V, et al. Chloroquine potentiates primaquine activity against active and latent hepatic plasmodia ex vivo: potentials and pitfalls. Antimicrob Agents Chemother. 2021;65:e01416-20.
  161. 161. Baird JK, Hoffman SL. Primaquine therapy for malaria. Clin Infect Dis. 2004;39:1336-1345.
  162. 162. Thomas D, Tazerouni H, Sundararaj KG, et al. Therapeutic failure of primaquine and need for new medicines in radical cure of Plasmodium vivax. Acta Trop. 2016;160:35-38.
  163. 163. Imwong M, Snounou G, Pukrittayakamee S, et al. Relapses of Plasmodium vivax infection usually result from activation of heterologous hypnozoites. J Infect Dis. 2007;195:927-933.
  164. 164. Chen N, Auliff A, Rieckmann K, et al. Relapses of Plasmodium vivax infection result from clonal hypnozoites activated at predetermined intervals. J Infect Dis. 2007;195:934-941.
  165. 165. Sutanto I, Tjahjono B, Basri H, et al. Randomized, open-label trial of primaquine against vivax malaria relapse in Indonesia. Antimicrob Agents Chemother. 2013;57:1128-1135.
  166. 166. Vallejo AF, Garcia J, Amado-Garavito AB, et al. Plasmodium vivax gametocyte infectivity in sub-microscopic infections. Malar J. 2016;15:48.
  167. 167. Almeida ACG, Kuehn A, Castro AJM, et al. High proportions of asymptomatic and submicroscopic Plasmodium vivax infections in a peri-urban area of low transmission in the Brazilian Amazon. Parasit Vectors. 2018;11:194.
  168. 168. Tadesse FG, Slater HC, Chali W, et al. The Relative Contribution of Symptomatic and Asymptomatic Plasmodium vivax and Plasmodium falciparum Infections to the Infectious Reservoir in a Low-Endemic Setting in Ethiopia. Clin Infect Dis. 2018;66:1883-1891.
  169. 169. Collins WE, Sullivan JS, Nace D, et al. Experimental infection of Anopheles farauti with different species of Plasmodium. J Parasitol. 2002;88:295-298.
  170. 170. Sattabongkot J, Tsuboi T, Zollner GE, et al. Plasmodium vivax transmission: chances for control? Trends Parasitol. 2004;20:192-198.
  171. 171. Battle KE, Gething PW, Elyazar IR, et al. The global public health significance of Plasmodium vivax. Adv Parasitol. 2012;80:1-111.
  172. 172. Hemingway J. The role of vector control in stopping the transmission of malaria: threats and opportunities. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130431.
  173. 173. Bhatt S, Weiss DJ, Cameron E, et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015;526:207-211.
  174. 174. Killeen GF. Control of malaria vectors and management of insecticide resistance through universal coverage with next-generation insecticide-treated nets. Lancet. 2020;395:1394-1400.
  175. 175. Smithuis FM, Kyaw MK, Phe UO, et al. Entomological determinants of insecticide-treated bed net effectiveness in Western Myanmar. Malar J. 2013;12:364.
  176. 176. Smithuis FM, Kyaw MK, Phe UO, et al. The effect of insecticide-treated bed nets on the incidence and prevalence of malaria in children in an area of unstable seasonal transmission in western Myanmar. Malar J. 2013;12:363.
  177. 177. Edwards HM, Chinh VD, Le Duy B, et al. Characterising residual malaria transmission in forested areas with low coverage of core vector control in central Viet Nam. Parasit Vectors. 2019;12:454.
  178. 178. Edwards HM, Sriwichai P, Kirabittir K, et al. 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. 2019;18:221.
  179. 179. Chareonviriyaphap T, Bangs MJ, Suwonkerd W, et al. Review of insecticide resistance and behavioral avoidance of vectors of human diseases in Thailand. Parasit Vectors. 2013;6:280.
  180. 180. Van Bortel W, Trung HD, Thuan le K, et al. The insecticide resistance status of malaria vectors in the Mekong region. Malar J. 2008;7:102.
  181. 181. Verhaeghen K, Van Bortel W, Trung HD, et al. Absence of knockdown resistance suggests metabolic resistance in the main malaria vectors of the Mekong region. Malar J. 2009;8:84.
  182. 182. Derua YA, Kweka EJ, Kisinza WN, et al. Bacterial larvicides used for malaria vector control in sub-Saharan Africa: review of their effectiveness and operational feasibility. Parasit Vectors. 2019;12:426.
  183. 183. Geissbuhler Y, Kannady K, Chaki PP, et al. Microbial larvicide application by a large-scale, community-based program reduces malaria infection prevalence in urban Dar es Salaam, Tanzania. PLoS One. 2009;4:e5107.
  184. 184. Chaccour CJ, Kobylinski KC, Bassat Q, et al. Ivermectin to reduce malaria transmission: a research agenda for a promising new tool for elimination. Malar J. 2013;12:153.
  185. 185. Kobylinski KC, Ubalee R, Ponlawat A, et al. Ivermectin susceptibility and sporontocidal effect in Greater Mekong Subregion Anopheles. Malar J. 2017;16:280.
  186. 186. Sluydts V, Durnez L, Heng S, et al. Efficacy of topical mosquito repellent (picaridin) plus long-lasting insecticidal nets versus long-lasting insecticidal nets alone for control of malaria: a cluster randomised controlled trial. Lancet Infect Dis. 2016;16:1169-1177.
  187. 187. Gryseels C, Uk S, Sluydts V, et al. Factors influencing the use of topical repellents: implications for the effectiveness of malaria elimination strategies. Sci Rep. 2015;5:16847.
  188. 188. Wang S, Jacobs-Lorena M. Genetic approaches to interfere with malaria transmission by vector mosquitoes. Trends Biotechnol. 2013;31:185-193.
  189. 189. Barreaux P, Barreaux AMG, Sternberg ED, et al. Priorities for broadening the malaria vector control tool kit. Trends Parasitol. 2017;33:763-774.
  190. 190. WHO. Malaria-free Maldives. 2016.
  191. 191. Premaratne R, Ortega L, Janakan N, et al. Malaria elimination in Sri Lanka: what it would take to reach the goal. WHO South East Asia J Public Health. 2014;3:85-89.
  192. 192. WHO. Turkmenistan certified malaria-free. Weekly epidemiological record. 2010;85:461-472.
  193. 193. Feng J, Zhang L, Huang F, et al. Ready for malaria elimination: zero indigenous case reported in the People's Republic of China. Malar J. 2018;17:315.
  194. 194. Cao J, Sturrock HJ, Cotter C, et al. Communicating and monitoring surveillance and response activities for malaria elimination: China's "1-3-7" strategy. PLoS Med. 2014;11:e1001642.
  195. 195. Feng J, Liu J, Feng X, et al. Towards malaria elimination: monitoring and evaluation of the "1-3-7" approach at the China-Myanmar border. Am J Trop Med Hyg. 2016;95:806-810.
  196. 196. Yang H, Wang J, Liu H, et al. Randomized, double-blind, placebo-controlled studies to assess safety and prophylactic efficacy of naphthoquine-azithromycin combination for malaria prophylaxis in Southeast Asia. Antimicrob Agents Chemother. 2018;62:e00793-00718.
  197. 197. Yang H, Wang J, Liu H, et al. Efficacy and safety of a naphthoquine-azithromycin co-formulation for malaria prophylaxis in Southeast Asia: A phase 3, double-blind, randomized, placebo-controlled trial. Clin Infect Dis. 2020.
  198. 198. Song J, Socheat D, Tan B, et al. Rapid and effective malaria control in Cambodia through mass administration of artemisinin-piperaquine. Malar J. 2010;9:57.
  199. 199. Landier J, Kajeechiwa L, Thwin MM, et al. Safety and effectiveness of mass drug administration to accelerate elimination of artemisinin-resistant falciparum malaria: A pilot trial in four villages of Eastern Myanmar. Wellcome open research. 2017;2:81.
  200. 200. Zhou S, Li Z, Cotter C, et al. Trends of imported malaria in China 2010-2014: analysis of surveillance data. Malar J. 2016;15:39.
  201. 201. Sleigh AC, Liu XL, Jackson S, et al. Resurgence of vivax malaria in Henan Province, China. Bull World Health Organ. 1998;76:265-270.
  202. 202. Cohen JM, Smith DL, Cotter C, et al. Malaria resurgence: a systematic review and assessment of its causes. Malar J. 2012;11:122.
  203. 203. Mendis K. Eliminating malaria should not be the end of vigilance. Nature. 2019;573:7.
  204. 204. Bahk YY, Lee HW, Na BK, et al. Epidemiological characteristics of re-emerging vivax malaria in the Republic of Korea (1993-2017). Korean J Parasitol. 2018;56:531-543.
  205. 205. Fernando SD, Booso R, Dharmawardena P, et al. The need for preventive and curative services for malaria when the military is deployed in endemic overseas territories: a case study and lessons learned. Mil Med Res. 2017;4:19.
  206. 206. Karunasena VM, Marasinghe M, Koo C, et al. The first introduced malaria case reported from Sri Lanka after elimination: implications for preventing the re-introduction of malaria in recently eliminated countries. Malar J. 2019;18:210.

Written By

Liwang Cui, Awtum Brashear, Lynette Menezes and John Adams

Submitted: 19 December 2020 Reviewed: 12 February 2021 Published: 08 March 2021