Next Article in Journal
Epidemiology, Risk Factors and Seasonal Variation of Scrub Typhus Fever in Central Nepal
Previous Article in Journal
Acute Pulmonary Histoplasmosis Outbreak in A Documentary Film Crew Travelling from Guatemala to Australia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Resistance to Artemisinin Combination Therapies (ACTs): Do Not Forget the Partner Drug!

by
Christian Nsanzabana
1,2
1
Department of Medicine, Swiss Tropical and Public Health Institute, CH-4051 Basel, Switzerland
2
University of Basel, P.O. Box, CH-4003 Basel, Switzerland
Trop. Med. Infect. Dis. 2019, 4(1), 26; https://doi.org/10.3390/tropicalmed4010026
Submission received: 27 December 2018 / Revised: 30 January 2019 / Accepted: 30 January 2019 / Published: 1 February 2019
(This article belongs to the Special Issue Drug Resistance in the Malaria Parasite: Biology and Epidemiology)

Abstract

:
Artemisinin-based combination therapies (ACTs) have become the mainstay for malaria treatment in almost all malaria endemic settings. Artemisinin derivatives are highly potent and fast acting antimalarials; but they have a short half-life and need to be combined with partner drugs with a longer half-life to clear the remaining parasites after a standard 3-day ACT regimen. When introduced, ACTs were highly efficacious and contributed to the steep decrease of malaria over the last decades. However, parasites with decreased susceptibility to artemisinins have emerged in the Greater Mekong Subregion (GMS), followed by ACTs’ failure, due to both decreased susceptibility to artemisinin and partner drug resistance. Therefore, there is an urgent need to strengthen and expand current resistance surveillance systems beyond the GMS to track the emergence or spread of artemisinin resistance. Great attention has been paid to the spread of artemisinin resistance over the last five years, since molecular markers of decreased susceptibility to artemisinin in the GMS have been discovered. However, resistance to partner drugs is critical, as ACTs can still be effective against parasites with decreased susceptibility to artemisinins, when the latter are combined with a highly efficacious partner drug. This review outlines the different mechanisms of resistance and molecular markers associated with resistance to partner drugs for the currently used ACTs. Strategies to improve surveillance and potential solutions to extend the useful therapeutic lifespan of the currently available malaria medicines are proposed.

1. Background

The emergence and spread of Plasmodium falciparum parasites with decreased susceptibility to artemisinin derivatives, and subsequent treatment failures after treatment with artemisinin-based combination therapies (ACTs) in the Greater Mekong Subregion (GMS) have raised concerns about the loss of the only highly-effective treatment currently available to treat malaria [1,2]. Artemisinin resistance, defined as delayed parasite clearance following treatment with conventional ACT regimens, has been associated with pfKelch13 mutations in the GMS, and several mutations have been validated as molecular markers of artemisinin resistance in that region [3,4]. Artemisinin resistance may have spread or emerged in Eastern India, where delayed parasite clearance combined with a high rate of parasite survival in the ring survival assay (RSA) and the presence of certain pfKelch13 mutations have been observed [5]. Moreover, some pfKelch13 mutations associated with delayed parasite clearance have been also reported in South America [6,7] and Papua New Guinea [8]. Nevertheless, different pfKelch13 mutations have been observed in Africa, but these were not associated with artemisinin resistance [9,10,11]. The mode of action of artemisinins is still not fully understood, even though artemisinin derivatives have been shown to cause oxidative stress and are probably impacting on multiple targets in the parasite, definitely implying a more complex resistance mechanism associated with different determinants other than pfKelch13 [12,13]. Therefore, there is a possibility that parasites with decreased artemisinin susceptibility are already present in some places in Africa, and are circulating at low levels, due to the still high multiplicity of infections in this region, and the related semi-immunity in older individuals [14]. Moreover, partner drugs do play a role in parasite clearance, even though to a lesser extent than artemisinin derivatives, but their decreased efficacy is probably affecting parasite clearance, as well [15]. In fact, even though artemisinin derivatives are highly potent antimalarials that can reduce parasite biomass very quickly [16]; due to their short half-life, they need to be combined with partner drugs with long half-life to clear the remaining parasites after a 3-day ACT treatment course [17]. Indeed, ACT failure is not only due to artemisinin resistance, but also to the failure of partner drugs [18,19]. There are currently five ACTs recommended by the World Health Organization (WHO): artesunate-sulfadoxine-pyrimethamine (ASSP), artemether-lumefantrine (AL), dihydroartemisinin-piperaquine (DP), artesunate-amodiaquine (ASAQ), and artesunate-mefloquine (ASMQ) [20]. Artesunate-pyronaridine (ASPY) is now listed under the WHO’s Model List of Essential Medicines, and previous restrictions due to concerns about its hepatotoxicity effects have been removed [21,22]. In this review, the development of resistance to ACTs is discussed and strategies to improve antimalarial drug resistance surveillance systems that should pay the same attention to partner drug resistance are proposed, as partner drug resistance may exist or occur even before resistance to artemisinins emerges in high endemic settings.

2. Mechanisms of Resistance to Partner Drugs

2.1. Lumefantrine

Artemether-lumefantrine (AL) is the most widely used antimalarial in endemic countries. In 2017, it was estimated that it accounted for almost 75% of all procured quality-assured ACTs [23]. The mode of action of lumefantrine is not well understood, and its main target remains unresolved, even though it is thought to interfere with hem detoxification [24], or to directly inhibit pfmdr1 [25]. Some recent reports have shown a decreased efficacy of AL in Angola [26,27], Gambia and Malawi [28]; however, so far no convincing evidence of lumefantrine resistance has been reported from the field [29]. Moreover, it has been shown recently in Angola that efficacy was higher when the full treatment course was directly observed [30]. Decreased susceptibility to lumefantrine (LUM) has been associated with gene copy number variations (CNV) in pfmdr1 [31] and the wild type pfcrt (K76) and pfmdr1 (N86) that confer increased susceptibility to chloroquine [32,33,34]. However, large meta-analyses failed to find a correlation between pfmdr1 CNV and treatment failure, but confirmed the association with of pfcrt K76 and pfmdr-1 N86 polymorphisms [35,36]. Moreover, laboratory selection of LUM resistant parasites has shown that multiple protein transporters may be involved in LUM resistance [37], and other investigations have shown that pfmrp1 was associated with decreased susceptibility to LUM in vitro [38].

2.2. Amodiaquine

The combination of artesunate and amodiaquine (ASAQ) is the second most used ACT in malaria endemic settings. In 2017, it was estimated to account for >20 % of all good quality delivered ACTs [23]. Furthermore, amodiaquine (AQ) is used in combination with sulfadoxine-pyrimethamine (SP) for seasonal malaria chemoprevention in the Sahel region [39]. The mode of action of AQ is similar to chloroquine; the drug accumulates in the digestive vacuole where it binds the toxic hem, preventing its formation into the inert form hemozoin [40]. AQ resistance has been associated with point mutations in the pfcrt and pfmdr1 genes, and the same mutations as those for chloroquine resistance (pfcrt 76T and pfmdr1 86Y) have been shown to be the main determinant of decrease susceptibility to AQ in vitro and in vivo [34,41,42]. However, a specific pfcrt haplotype of codons 72 to 76 (SVMNT) has been associated with resistance to AQ, but to a lesser degree of resistance to chloroquine [43,44,45].

2.3. Piperaquine

The combination of dihydroartemisinin and piperaquine (DP) has proven to be an efficacious treatment in most malaria endemic settings [46], and is used for treatment in some countries in Southeast Asia and Africa [47]. Dihydroartemisinin and Piperaquine are also increasingly considered for malaria prevention in pregnancy [48,49], and for mass drug administration (MDA) in near-elimination settings in Africa [50,51,52]. However, DP is failing in the sub-Mekong region due to resistance to piperaquine (PQ) and decreased susceptibility to artemisinin derivatives [53,54]. Treatment failure with DP was first observed in Western Cambodia, where the cumulative risk of treatment failure increased from 9.2 % to 24.1% between 2008 and 2010 [18]. The mode of action of PQ was initially linked to the inhibition of one or more steps in the hemoglobin degradation pathway, but it is not yet fully understood [55,56]. More recently, PQ resistance has been associated with CNV in plasmepsin II and III [57] and point mutations in pfcrt [56,58,59]. As the classic in vivo assays have shown limitations in the assessment of phonotypical resistance to PQ, a piperaquine survival assay (PSA) has been established to assess changes in parasite strains’ susceptibility to PQ [57]. The presence of different parasite populations with different mechanisms of resistance to PQ is intriguing and warrants further investigations to elucidate this pleiotropic mechanism of resistance [59].

2.4. Sulfadoxine-Pyrimethamine

Despite widespread resistance to sulfadoxine-pyrimethamine (SP) in most malaria endemic settings, the combination artesunate and SP (ASSP) is still used in a few countries for malaria treatment [47] and intermittent preventive treatment in pregnancy (IPTp) [49], and in combination with amodiaquine for seasonal malaria chemoprevention (SMC) [39]. Sulfadoxine and pyrimethamine are inhibiting two enzymes involved the folate biosynthesis pathway; dihydropteroate synthase DHPS and dihydrofolate reductase DHFR, respectively [60,61]. Resistance to sulfadoxine and pyrimethamine is by far the best characterized mechanism of resistance; it is associated with point mutations in the pfdhps and pfdhfr genes, respectively [62,63]. Indeed, the accumulation of point mutations in both genes is associated with increasing levels of resistance to the combination of the two drugs, with the combination of the triple mutant in pfdhfr (51I, 59R, 108N) and the double mutant in pfdhps (437G and 540E) being associated with an increased risk of in vivo treatment failure. This quintuple mutant has been shown to have emerged in Southeast Asia before it spread to other malaria endemic areas, notably Africa [64,65]. The presence of the quintuple mutation is used to guide treatment policy for intermittent preventive treatment in infants (IPTi): WHO is recommending not to use IPTi in areas where the pfdhps 540E mutation (a surrogate marker for the presence of the pfdhfr/pfdhps quintuple mutant) does exceed 50% [66].

2.5. Mefloquine

The combination of artesunate and mefloquine (ASMQ) was the first ACT that has been introduced in Southeast Asia to stop resistance to mefloquine (MQ) monotherapy in the early 1990s [67]. Since, the use of this combination has historically been restricted to Southeast Asia, the Pacific region and South America, but was not used in Africa. With increasing levels of resistance to MQ and decreased susceptibility to artemisinin derivatives, the combination is now used only in a few countries [47]. The mode of action of MQ is still unclear, but the drug may inhibit hem detoxification [24,68], or directly inhibit pfmdr1 [69]. Mefloquine resistance has been associated with CNV in pfmdr1 [70,71], and polymorphisms in pfmrp1 and pfmrp2 could also potentiate MQ resistance [38,72,73]. More recently, the cytoplasmic ribosome (pf80S) of the asexual blood-stage parasite has been suggested as the main target of MQ [74].

2.6. Pyronaridine

The combination of artesunate and pyronaridine (ASPY) is not yet recommended by the WHO, but has received a positive opinion from the European Medical Agency (EMA) under article 58 [21]. Moreover, the product was added to the WHO’s Model List of Essential Medicines (EML) and Model List of Essential Medicines for Children (EMLc) in 2017, and is the only ACT indicated for the blood stage treatment of both P. falciparum and P. vivax [22]. The combination has shown high efficacy in Africa where its efficacy was non-inferior to AL and DP [75,76], and in Eastern Cambodia [77]. However, the efficacy of the combination has been shown to be low in Western Cambodia, probably due to artemisinin resistance and potential cross-resistance between pyronaridine (PY) and PQ [78]. The exact mode of action of PY is not well known; however, the drug is thought to interfere with the hemozoin formation [79]. To date, resistance to PY has not been reported, but ex vivo assessment has shown an association between decreased susceptibility to PY and the 76T mutation in pfcrt [80].

3. Discussion

The emergence and spread of parasites with decreased susceptibility to artemisinins is a major threat to malaria control and elimination. Monitoring antimalarial drug efficacy and resistance is of paramount importance to maintain the gains made over the last decades in reducing malaria burden and mortality. Surveillance systems using molecular markers of resistance should become standard practice, as they are easy to implement and can offer more updated information to complement the often sparse and outdated data from therapeutic efficacy studies. Indeed, molecular markers can provide useful information to policymakers, allowing them anticipating treatment efficacy changes over time, and eventually decide on treatment policy change before resistance translates to clinical failures. However, molecular surveillance should not only focus on artemisinin resistance, but also on partner drug resistance.
Our understanding of the mode of action and mechanisms of resistance, even though still incomplete, has substantially improved, and validated molecular markers can be used to track the emergence and spread of resistance to antimalarial drugs, as well as to predict clinical efficacy or failure. For instance, CQ withdrawal in most of Africa lead to the re-emergence of CQ sensitive parasites with the expansion of wild-type pfcrt K76 that ultimately resulted in CQ efficacy restoration [81,82]. The fact that the two major ACTs in Africa (i.e., ASAQ and AL) were selecting in opposite directions on the pfmdr1 and pfcrt genes [35,83] resulted in increasing efficacy of ASAQ in areas of high AL usage, where the efficacy of the latter was declining [34]. However, mechanisms of resistance may differ from one area to another: Whilst the restoration of CQ efficacy was associated with the return of the wild-type pfcrt K76 in Africa [82], restoration of CQ efficacy in French Guiana was associated with a new mutation in pfcrt (350R), with the 76T mutation remaining fixed in the parasite population [84]. Likewise, resistance to PQ has been associated with an elevated copy number of the plasmepsin II and III gens [54,57]; however, other studies have shown that PQ resistance was mediated through single nucleotide polymorphisms (SNPs) in pfcrt [56,58,59] making the molecular surveillance of PQ resistance more complex. Therefore, surveillance systems should not only aim at detecting known and validated molecular markers, but also at tracking any new genotypes that could be associated with antimalarial drug resistance. Whole genome sequencing (WGS) could be used in routine surveillance through regional reference laboratories with respective capacities [85,86] for defining potential new markers that, in turn, would be validated by in vitro phenotypic assays and/or gene editing techniques.
In high transmission settings, parasites are more exposed to sub-therapeutic drug concentrations of the long half-life partner drugs, thus increasing the chances of developing resistance [87]. The introduction of aggressive chemoprevention methods such as seasonal malaria chemoprevention (SMC) and mass drug administration (MDA) will put even more pressure on partner drugs [88,89]. The presence of parasites with decreased susceptibility to artemisinin derivatives does not necessarily lead to ACT treatment failure, as evidenced with a recent report from Myanmar, where despite high prevalence of pfKelch13 mutations associated with delayed parasite clearance, AL retains its high clinical efficacy [90]. However, resistance to the partner drug would immediately lead to ACT failure, as with the current 3-day ACT regimen, the short half-life of artemisinin derivatives could not sustain the efficacy of the combination on its own [91,92]. Hence, molecular surveillance in high transmission settings should aim at using more sensitive techniques such as amplicon deep sequencing that can also detect minority variants potentially present in the parasite population [93,94,95], this could help to earlier predict the emergence and spread of resistant parasites, allowing policymakers to develop alternative treatment strategies, before resistance translates into clinical treatment failures.
Currently, there is no alternative to ACTs, even though the antimalarial drug pipeline is promising [96,97]. Strategies are needed to prolong the useful therapeutic lifespan of the current malaria medicines, including 1) extending the duration of the current 3-day regimen of ACTs [90,98,99]; 2) increasing the dose of the partner drugs [100,101]; 3) using triple combination therapies, with two partner drugs selecting in opposite directions [36,102,103]; and 4) utilizing multiple first-line treatments [36]. However, those may be solutions to preserve the efficacy of ACTs in the short- or medium-term only, as there is already some evidence of parasites developing resistance to two partner drugs and the artemisinin component at the same time [104,105]. The development of nano-based drug formulations is another strategy to fight drug resistance by improving drug targeting and dosing [106,107,108]. More resources should also be allocated to study the mode of action and mechanisms of resistance to new antimalarial drugs. Not only will this allow the discovery of new molecular markers for resistance surveillance, but also pave the way for the development of new drugs with different modes of action.
The emergence and spread of artemisinin resistance are a major threat to the current efforts to control and eliminate malaria. Molecular markers are valuable tools for monitoring antimalarial drug resistance, and need to be fully integrated in routine surveillance, especially for partner drugs in high endemic settings, where resistance to partner drugs may emerge before resistance to artemisinin.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Menard, D.; Dondorp, A. Antimalarial Drug Resistance: A Threat to Malaria Elimination. Cold Spring Harb. Perspect. Med. 2017, 7, a025619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Woodrow, C.J.; White, N.J. The clinical impact of artemisinin resistance in Southeast Asia and the potential for future spread. FEMS Microbiol. Rev. 2017, 41, 34–48. [Google Scholar] [CrossRef] [PubMed]
  3. Ariey, F.; Witkowski, B.; Amaratunga, C.; Beghain, J.; Langlois, A.-C.; Khim, N.; Kim, S.; Duru, V.; Bouchier, C.; Ma, L.; et al. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature 2014, 505, 50–55. [Google Scholar] [CrossRef] [PubMed]
  4. Ménard, D.; Khim, N.; Beghain, J.; Adegnika, A.A.; Shafiul-Alam, M.; Amodu, O.; Rahim-Awab, G.; Barnadas, C.; Berry, A.; Boum, Y.; et al. A Worldwide Map of Plasmodium falciparum K13-Propeller Polymorphisms. N. Eng. J. Med. 2016, 374, 2453–2464. [Google Scholar] [CrossRef] [PubMed]
  5. Das, S.; Saha, B.; Hati, A.K.; Roy, S. Evidence of Artemisinin-Resistant Plasmodium falciparum Malaria in Eastern India. N. Engl. J. Med. 2018, 379, 1962–1964. [Google Scholar] [CrossRef] [PubMed]
  6. Chenet, S.M.; Akinyi Okoth, S.; Huber, C.S.; Chandrabose, J.; Lucchi, N.W.; Talundzic, E.; Krishnalall, K.; Ceron, N.; Musset, L.; Macedo de Oliveira, A.; et al. Independent Emergence of the Plasmodium falciparum Kelch Propeller Domain Mutant Allele C580Y in Guyana. J. Infect. Dis. 2016, 213, 1472–1475. [Google Scholar] [CrossRef] [PubMed]
  7. Mathieu, L.; Cox, H.; Early, A.M.; Ade, M.-P.; Lazrek, Y.; Grant, Q.; Lucchi, N.W.; Udhayakumar, V.; Seme Fils, A.J.; Fidock, D.A.; et al. Artemisinin resistance and the pfk13 C580Y mutation in Guyana: A confirmed link and emergence. In Proceedings of the ASTMH Annual Meeting, New Orleans, LA, USA, 28 October–1 November 2018. [Google Scholar]
  8. Prosser, C.; Meyer, W.; Ellis, J.; Lee, R. Resistance screening and trend analysis of imported falciparum malaria in NSW, Australia (2010 to 2016). PLoS ONE 2018, 13, e0197369. [Google Scholar] [CrossRef]
  9. Taylor, S.M.; Parobek, C.M.; DeConti, D.K.; Kayentao, K.; Coulibaly, S.O.; Greenwood, B.M.; Tagbor, H.; Williams, J.; Bojang, K.; Njie, F.; et al. Absence of putative artemisinin resistance mutations among Plasmodium falciparum in Sub-Saharan Africa: A molecular epidemiologic study. J. Infect. Dis. 2015, 211, 680–688. [Google Scholar] [CrossRef]
  10. Kamau, E.; Campino, S.; Amenga-Etego, L.; Drury, E.; Ishengoma, D.; Johnson, K.; Mumba, D.; Kekre, M.; Yavo, W.; Mead, D.; et al. K13-propeller polymorphisms in Plasmodium falciparum parasites from sub-Saharan Africa. J. Infect. Dis. 2015, 211, 1352–1355. [Google Scholar] [CrossRef]
  11. Apinjoh, T.O.; Mugri, R.N.; Miotto, O.; Chi, H.F.; Tata, R.B.; Anchang-Kimbi, J.K.; Fon, E.M.; Tangoh, D.A.; Nyingchu, R.V.; Jacob, C.; et al. Molecular markers for artemisinin and partner drug resistance in natural Plasmodium falciparum populations following increased insecticide treated net coverage along the slope of mount Cameroon: Cross-sectional study. Infect. Dis. Poverty 2017, 6, 136. [Google Scholar] [CrossRef]
  12. Mbengue, A.; Bhattacharjee, S.; Pandharkar, T.; Liu, H.; Estiu, G.; Stahelin, R.V.; Rizk, S.S.; Njimoh, D.L.; Ryan, Y.; Chotivanich, K.; et al. A molecular mechanism of artemisinin resistance in Plasmodium falciparum malaria. Nature 2015, 520, 683–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Tilley, L.; Straimer, J.; Gnädig, N.F.; Ralph, S.A.; Fidock, D.A. Artemisinin Action and Resistance in Plasmodium falciparum. Trends Parasitol. 2016, 32, 682–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. MalariaGEN Plasmodium falciparum Community Project. Genomic epidemiology of artemisinin resistant malaria. Elife 2016, 5, e08714. [Google Scholar] [CrossRef] [PubMed]
  15. WWARN Parasite Clearance Study Group, W.P.C.S.; Abdulla, S.; Ashley, E.A.; Bassat, Q.; Bethell, D.; Björkman, A.; Borrmann, S.; D’Alessandro, U.; Dahal, P.; Day, N.P.; et al. Baseline data of parasite clearance in patients with falciparum malaria treated with an artemisinin derivative: An individual patient data meta-analysis. Malar. J. 2015, 14, 359. [Google Scholar] [CrossRef] [PubMed]
  16. White, N.J. Clinical pharmacokinetics and pharmacodynamics of artemisinin and derivatives. Trans. R. Soc. Trop. Med. Hyg. 1994, 88 (Suppl. 1), 41–43. [Google Scholar] [CrossRef]
  17. Nosten, F.; White, N.J. Artemisinin-based combination treatment of falciparum malaria. Am. J. Trop. Med. Hyg. 2007, 77, 181–192. [Google Scholar] [CrossRef] [PubMed]
  18. Leang, R.; Barrette, A.; Bouth, D.M.; Menard, D.; Abdur, R.; Duong, S.; Ringwald, P. Efficacy of dihydroartemisinin-piperaquine for treatment of uncomplicated Plasmodium falciparum and Plasmodium vivax in Cambodia, 2008 to 2010. Antimicrob. Agents Chemother. 2013, 57, 818–826. [Google Scholar] [CrossRef] [PubMed]
  19. Saunders, D.L.; Vanachayangkul, P.; Lon, C. Dihydroartemisinin–Piperaquine Failure in Cambodia. N. Engl. J. Med. 2014, 371, 484–485. [Google Scholar] [CrossRef] [Green Version]
  20. WHO. WHO|Guidelines for the Treatment of Malaria, 3rd ed.; World Health Organization: Geneva, Switzerland, 2015; p. 316. [Google Scholar]
  21. EMEA. Assessment Report Pyramax Pyronaridine Tetraphosphate/Artesunate Procedure No.: EMEA/H/W/002319; EMEA: London, UK, 2012; p. 123. [Google Scholar]
  22. MMV Pyramax® (Pyronaridine-Artesunate). Available online: https://www.mmv.org/access/products-projects/pyramax-pyronaridine-artesunate (accessed on 23 January 2019).
  23. UNITAID. Global Malaria Diagnostic and Artemisinin Treatment Commodities Demand Forecast (Phase 2); UNITAID: Geneva, Switzerland, 2018; p. 89. [Google Scholar]
  24. Combrinck, J.M.; Mabotha, T.E.; Ncokazi, K.K.; Ambele, M.A.; Taylor, D.; Smith, P.J.; Hoppe, H.C.; Egan, T.J. Insights into the Role of Heme in the Mechanism of Action of Antimalarials. ACS Chem. Biol. Biol. 2013, 8, 133–137. [Google Scholar] [CrossRef]
  25. Martin, R.E.; Shafik, S.H.; Richards, S.N. Mechanisms of resistance to the partner drugs of artemisinin in the malaria parasite. Curr. Opin. Pharmacol. 2018, 42, 71–80. [Google Scholar] [CrossRef]
  26. Plucinski, M.M.; Talundzic, E.; Morton, L.; Dimbu, P.R.; Macaia, A.P.; Fortes, F.; Goldman, I.; Lucchi, N.; Stennies, G.; MacArthur, J.R.; et al. Efficacy of artemether-lumefantrine and dihydroartemisinin-piperaquine for treatment of uncomplicated malaria in children in Zaire and Uíge Provinces, angola. Antimicrob. Agents Chemother. 2015, 59, 437–443. [Google Scholar] [CrossRef] [PubMed]
  27. Plucinski, M.M.; Dimbu, P.R.; Macaia, A.P.; Ferreira, C.M.; Samutondo, C.; Quivinja, J.; Afonso, M.; Kiniffo, R.; Mbounga, E.; Kelley, J.S.; et al. Efficacy of artemether–lumefantrine, artesunate–amodiaquine, and dihydroartemisinin–piperaquine for treatment of uncomplicated Plasmodium falciparum malaria in Angola, 2015. Malar. J. 2017, 16, 62. [Google Scholar] [CrossRef]
  28. WHO. World Malaria Report 2017; WHO: Geneva, Switzerland, 2017; p. 196. [Google Scholar]
  29. Hamed, K.; Kuhen, K. No robust evidence of lumefantrine resistance. Antimicrob. Agents Chemother. 2015, 59, 5865–5866. [Google Scholar] [CrossRef] [PubMed]
  30. Davlantes, E.; Dimbu, P.R.; Ferreira, C.M.; Florinda Joao, M.; Pode, D.; Félix, J.; Sanhangala, E.; Andrade, B.N.; dos Santos Souza, S.; Talundzic, E.; et al. Efficacy and safety of artemether–lumefantrine, artesunate–amodiaquine, and dihydroartemisinin–piperaquine for the treatment of uncomplicated Plasmodium falciparum malaria in three provinces in Angola, 2017. Malar. J. 2018, 17, 144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Sidhu, A.B.S.; Uhlemann, A.-C.; Valderramos, S.G.; Valderramos, J.-C.; Krishna, S.; Fidock, D.A. Decreasing pfmdr1 copy number in plasmodium falciparum malaria heightens susceptibility to mefloquine, lumefantrine, halofantrine, quinine, and artemisinin. J. Infect. Dis. 2006, 194, 528–535. [Google Scholar] [CrossRef] [PubMed]
  32. Sisowath, C.; Strömberg, J.; Mårtensson, A.; Msellem, M.; Obondo, C.; Björkman, A.; Gil, J.P. In Vivo Selection of Plasmodium falciparum pfmdr1 86N Coding Alleles by Artemether-Lumefantrine (Coartem). J. Infect. Dis. 2005, 191, 1014–1017. [Google Scholar] [CrossRef] [PubMed]
  33. Sisowath, C.; Petersen, I.; Veiga, M.I.; Mårtensson, A.; Premji, Z.; Björkman, A.; Fidock, D.A.; Gil, J.P. In vivo selection of Plasmodium falciparum parasites carrying the chloroquine-susceptible pfcrt K76 allele after treatment with artemether-lumefantrine in Africa. J. Infect. Dis. 2009, 199, 750–757. [Google Scholar] [CrossRef] [PubMed]
  34. Yeka, A.; Kigozi, R.; Conrad, M.D.; Lugemwa, M.; Okui, P.; Katureebe, C.; Belay, K.; Kapella, B.K.; Chang, M.A.; Kamya, M.R.; et al. Artesunate/Amodiaquine Versus Artemether/Lumefantrine for the Treatment of Uncomplicated Malaria in Uganda: A Randomized Trial. J. Infect. Dis. 2016, 213, 1134–1142. [Google Scholar] [CrossRef] [PubMed]
  35. Venkatesan, M.; Gadalla, N.B.; Stepniewska, K.; Dahal, P.; Nsanzabana, C.; Moriera, C.; Price, R.N.; Mårtensson, A.; Rosenthal, P.J.; Dorsey, G.; et al. Polymorphisms in Plasmodium falciparum chloroquine resistance transporter and multidrug resistance 1 genes: Parasite risk factors that affect treatment outcomes for P. falciparum malaria after artemether-lumefantrine and artesunate-amodiaquine. Am. J. Trop. Med. Hyg. 2014, 91, 833–843. [Google Scholar] [CrossRef] [PubMed]
  36. Okell, L.C.; Reiter, L.M.; Ebbe, L.S.; Baraka, V.; Bisanzio, D.; Watson, O.J.; Bennett, A.; Verity, R.; Gething, P.; Roper, C.; et al. Emerging implications of policies on malaria treatment: Genetic changes in the Pfmdr-1 gene affecting susceptibility to artemether-lumefantrine and artesunate-amodiaquine in Africa. BMJ Glob. Health 2018, 3, e000999. [Google Scholar] [CrossRef] [PubMed]
  37. Mwai, L.; Diriye, A.; Masseno, V.; Muriithi, S.; Feltwell, T.; Musyoki, J.; Lemieux, J.; Feller, A.; Mair, G.R.; Marsh, K.; et al. Genome wide adaptations of Plasmodium falciparum in response to lumefantrine selective drug pressure. PLoS ONE 2012, 7, e31623. [Google Scholar] [CrossRef] [PubMed]
  38. Veiga, M.I.; Ferreira, P.E.; Jörnhagen, L.; Malmberg, M.; Kone, A.; Schmidt, B.A.; Petzold, M.; Björkman, A.; Nosten, F.; Gil, J.P. Novel polymorphisms in Plasmodium falciparum ABC transporter genes are associated with major ACT antimalarial drug resistance. PLoS ONE 2011, 6, e20212. [Google Scholar] [CrossRef] [PubMed]
  39. Coldiron, M.E.; Von Seidlein, L.; Grais, R.F. Seasonal malaria chemoprevention: Successes and missed opportunities. Malar. J. 2017, 16, 481. [Google Scholar] [CrossRef] [PubMed]
  40. Kaur, K.; Jain, M.; Reddy, R.P.; Jain, R. Quinolines and structurally related heterocycles as antimalarials. Eur. J. Med Chem. 2010, 45, 3245–3264. [Google Scholar] [CrossRef] [PubMed]
  41. Duraisingh, M.T.; Drakeley, C.J.; Muller, O.; Bailey, R.; Snounou, G.; Targett, G.A.; Greenwood, B.M.; Warhurst, D.C. Evidence for selection for the tyrosine-86 allele of the pfmdr 1 gene of Plasmodium falciparum by chloroquine and amodiaquine. Parasitology 1997, 114 Pt 3, 205–211. [Google Scholar] [CrossRef]
  42. Holmgren, G.; Gil, J.P.; Ferreira, P.M.; Veiga, M.I.; Obonyo, C.O.; Björkman, A. Amodiaquine resistant Plasmodium falciparum malaria in vivo is associated with selection of pfcrt 76T and pfmdr1 86Y. Infect. Genet. Evol. 2006, 6, 309–314. [Google Scholar] [CrossRef] [PubMed]
  43. Sá, J.M.; Twu, O.; Hayton, K.; Reyes, S.; Fay, M.P.; Ringwald, P.; Wellems, T.E. Geographic patterns of Plasmodium falciparum drug resistance distinguished by differential responses to amodiaquine and chloroquine. Proc. Natl. Acad. Sci. USA 2009, 106, 18883. [Google Scholar] [CrossRef] [PubMed]
  44. Sa, J.M.; Twu, O. Protecting the malaria drug arsenal: Halting the rise and spread of amodiaquine resistance by monitoring the PfCRT SVMNT type. Malar. J. 2010, 9, 374. [Google Scholar] [CrossRef]
  45. Beshir, K.; Sutherland, C.J.; Merinopoulos, I.; Durrani, N.; Leslie, T.; Rowland, M.; Hallett, R.L. Amodiaquine resistance in Plasmodium falciparum malaria in Afghanistan is associated with the pfcrt SVMNT allele at codons 72 to 76. Antimicrob. Agents Chemother. 2010, 54, 3714–3716. [Google Scholar] [CrossRef]
  46. WorldWide Antimalarial Resistance Network (WWARN) DP Study Group. The effect of dosing regimens on the antimalarial efficacy of dihydroartemisinin-piperaquine: A pooled analysis of individual patient data. PLoS Med. 2013, 10, e1001564. [Google Scholar]
  47. WHO. World Malaria Report 2018; WHO: Geneva, Switzerland, 2018; p. 210. [Google Scholar]
  48. Kakuru, A.; Jagannathan, P.; Muhindo, M.K.; Natureeba, P.; Awori, P.; Nakalembe, M.; Opira, B.; Olwoch, P.; Ategeka, J.; Nayebare, P.; et al. Dihydroartemisinin-Piperaquine for the Prevention of Malaria in Pregnancy. N. Engl. J. Med. 2016, 374, 928–939. [Google Scholar] [CrossRef] [PubMed]
  49. Desai, M.; Hill, J.; Fernandes, S.; Walker, P.; Pell, C.; Gutman, J.; Kayentao, K.; Gonzalez, R.; Webster, J.; Greenwood, B.; et al. Prevention of malaria in pregnancy. Lancet Infect. Dis. 2018, 18, e119–e132. [Google Scholar] [CrossRef]
  50. Eisele, T.P.; Bennett, A.; Silumbe, K.; Finn, T.P.; Chalwe, V.; Kamuliwo, M.; Hamainza, B.; Moonga, H.; Kooma, E.; Chizema Kawesha, E.; et al. Short-term Impact of Mass Drug Administration with Dihydroartemisinin Plus Piperaquine on Malaria in Southern Province Zambia: A Cluster-Randomized Controlled Trial. J. Infect. Dis. 2016, 214, 1831–1839. [Google Scholar] [CrossRef] [PubMed]
  51. Mwesigwa, J.; Achan, J.; Affara, M.; Wathuo, M.; Worwui, A.; Muhommed, N.I.; Kanuteh, F.; Prom, A.; Dierickx, S.; di Tanna, G.L.; et al. Mass drug administration with dihydroartemisinin-piperaquine and malaria transmission dynamics in The Gambia—A prospective cohort study. Clin. Infect. Dis. 2018. [Google Scholar] [CrossRef] [PubMed]
  52. Guler, J.L.; Rosenthal, P.J. Mass drug administration to control and eliminate malaria in Africa: How do we best utilize the tools at hand? Clin. Infect. Dis. 2018. [Google Scholar] [CrossRef] [PubMed]
  53. Amaratunga, C.; Lim, P.; Suon, S.; Sreng, S.; Mao, S.; Sopha, C.; Sam, B.; Dek, D.; Try, V.; Amato, R.; et al. Dihydroartemisinin–piperaquine resistance in Plasmodium falciparum malaria in Cambodia: A multisite prospective cohort study. Lancet Infect. Dis. 2016, 16, 357. [Google Scholar] [CrossRef]
  54. Amato, D.R.; Lim, P.; Miotto, O.; Amaratunga, C.; Dek, D.; Pearson, R.D.; Almagro-Garcia, J.; Neal, A.T.; Sreng, S.; Suon, S.; et al. Genetic markers associated with dihydroartemisinin–piperaquine failure in Plasmodium falciparum malaria in Cambodia: A genotype-phenotype association study. Lancet Infect. Dis. 2017, 17, 164–173. [Google Scholar] [CrossRef]
  55. Davis, T.M.E.; Hung, T.-Y.; Sim, I.-K.; Karunajeewa, H.A.; Ilett, K.F. Piperaquine. Drugs 2005, 65, 75–87. [Google Scholar] [CrossRef]
  56. Dhingra, S.K.; Redhi, D.; Combrinck, J.M.; Yeo, T.; Okombo, J.; Henrich, P.P.; Cowell, A.N.; Gupta, P.; Stegman, M.L.; Hoke, J.M.; et al. A Variant PfCRT Isoform Can Contribute to Plasmodium falciparum Resistance to the First-Line Partner Drug Piperaquine. MBio 2017, 8, e00303-17. [Google Scholar] [CrossRef]
  57. Witkowski, B.; Duru, V.; Khim, N.; Ross, L.S.; Saintpierre, B.; Beghain, J.; Chy, S.; Kim, S.; Ke, S.; Kloeung, N.; et al. A surrogate marker of piperaquine-resistant Plasmodium falciparum malaria: A phenotype–genotype association study. Lancet Infect. Dis. 2017, 17, 174–183. [Google Scholar] [CrossRef]
  58. Agrawal, S.; Moser, K.A.; Morton, L.; Cummings, M.P.; Parihar, A.; Dwivedi, A.; Shetty, A.C.; Drabek, E.F.; Jacob, C.G.; Henrich, P.P.; et al. Association of a Novel Mutation in the Plasmodium falciparum Chloroquine Resistance Transporter with Decreased Piperaquine Sensitivity. J. Infect. Dis. 2017, 216, 468–476. [Google Scholar] [CrossRef] [PubMed]
  59. Ross, L.S.; Dhingra, S.K.; Mok, S.; Yeo, T.; Wicht, K.J.; Kümpornsin, K.; Takala-Harrison, S.; Witkowski, B.; Fairhurst, R.M.; Ariey, F.; et al. Emerging Southeast Asian PfCRT mutations confer Plasmodium falciparum resistance to the first-line antimalarial piperaquine. Nat. Commun. 2018, 9, 3314. [Google Scholar] [CrossRef] [PubMed]
  60. Cowman, A.F.; Morry, M.J.; Biggs, B.A.; Cross, G.A.; Foote, S.J. Amino acid changes linked to pyrimethamine resistance in the dihydrofolate reductase-thymidylate synthase gene of Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 1988, 85, 9109–9113. [Google Scholar] [CrossRef] [PubMed]
  61. Wang, P.; Read, M.; Sims, P.F.G.; Hyde, J.E. Sulfadoxine resistance in the human malaria parasite Plasmodium falciparum is determined by mutations in dihydropteroate synthetase and an additional factor associated with folate utilization. Mol Microbiol. 1997, 23, 979–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Plowe, C.V.; Cortese, J.F.; Djimde, A.; Nwanyanwu, O.C.; Watkins, W.M.; Winstanley, P.A.; Estrada-Franco, J.G.; Mollinedo, R.E.; Avila, J.C.; Cespedes, J.L.; et al. Mutations in Plasmodium falciparum dihydrofolate reductase and dihydropteroate synthase and epidemiologic patterns of pyrimethamine-sulfadoxine use and resistance. J. Infect. Dis. 1997, 176, 1590–1596. [Google Scholar] [CrossRef]
  63. Wang, P.; Lee, C.S.; Bayoumi, R.; Djimde, A.; Doumbo, O.; Swedberg, G.; Dao, L.D.; Mshinda, H.; Tanner, M.; Watkins, W.M.; et al. Resistance to antifolates in Plasmodium falciparum monitored by sequence analysis of dihydropteroate synthetase and dihydrofolate reductase alleles in a large number of field samples of diverse origins. Mol. Biochem Parasitol. 1997, 89, 161–177. [Google Scholar] [CrossRef]
  64. Naidoo, I.; Roper, C. Mapping “partially resistant”, “fully resistant”, and “super resistant” malaria. Trends Parasitol. 2013, 29, 505–515. [Google Scholar] [CrossRef]
  65. Roper, C.; Pearce, R.; Nair, S.; Sharp, B.; Nosten, F.; Anderson, T. Intercontinental Spread of Pyrimethamine-Resistant Malaria. Science 2004, 305, 1124. [Google Scholar] [CrossRef]
  66. WHO. WHO Policy Recommendation on Intermittent Preventive Treatment during Infancy with Sulphadoxine-Pyrimethamine (IPTi-SP) for Plasmodium falciparum Malaria Control in Africa; World Health Organization: Geneva, Switzerland, 2010; p. 3. [Google Scholar]
  67. Looareesuwan, S.; Kyle, D.E.; Viravan, C.; Vanijanonta, S.; Wilairatana, P.; Charoenlarp, P.; Canfield, C.J.; Webster, H.K. Treatment of patients with recrudescent falciparum malaria with a sequential combination of artesunate and mefloquine. Am. J. Trop. Med. Hyg. 1992, 47, 794–799. [Google Scholar] [CrossRef]
  68. Fitch, C.D. Ferriprotoporphyrin IX, phospholipids, and the antimalarial actions of quinoline drugs. Life Sci. 2004, 74, 1957–1972. [Google Scholar] [CrossRef]
  69. Cowman, A.F.; Galatis, D.; Thompson, J.K. Selection for mefloquine resistance in Plasmodium falciparum is linked to amplification of the pfmdr1 gene and cross-resistance to halofantrine and quinine. Proc. Natl. Acad. Sci. USA 1994, 91, 1143–1147. [Google Scholar] [CrossRef] [PubMed]
  70. Wilson, C.M.; Volkman, S.K.; Thaithong, S.; Martin, R.K.; Kyle, D.E.; Milhous, W.K.; Wirth, D.F. Amplification of pfmdr1 associated with mefloquine and halofantrine resistance in Plasmodium falciparum from Thailand. Mol. Biochem. Parasitol. 1993, 57, 151–160. [Google Scholar] [CrossRef]
  71. Price, R.N.; Uhlemann, A.-C.; Brockman, A.; McGready, R.; Ashley, E.; Phaipun, L.; Patel, R.; Laing, K.; Looareesuwan, S.; White, N.J.; et al. Mefloquine resistance in Plasmodium falciparum and increased pfmdr1 gene copy number. Lancet 2004, 364, 438–447. [Google Scholar] [CrossRef]
  72. Veiga, M.I.; Osório, N.S.; Ferreira, P.E.; Franzén, O.; Dahlstrom, S.; Lum, J.K.; Nosten, F.; Gil, J.P. Complex polymorphisms in the Plasmodium falciparum multidrug resistance protein 2 gene and its contribution to antimalarial response. Antimicrob. Agents Chemother. 2014, 58, 7390–7397. [Google Scholar] [CrossRef] [PubMed]
  73. Woodland, J.G.; Hunter, R.; Smith, P.J.; Egan, T.J. Chemical Proteomics and Super-resolution Imaging Reveal That Chloroquine Interacts with Plasmodium falciparum Multidrug Resistance-Associated Protein and Lipids. ACS Chem. Biol. 2018, 13, 2939–2948. [Google Scholar] [CrossRef]
  74. Wong, W.; Bai, X.-C.; Sleebs, B.E.; Triglia, T.; Brown, A.; Thompson, J.K.; Jackson, K.E.; Hanssen, E.; Marapana, D.S.; Fernandez, I.S.; et al. Mefloquine targets the Plasmodium falciparum 80S ribosome to inhibit protein synthesis. Nat. Microbiol. 2017, 2, 17031. [Google Scholar] [CrossRef]
  75. Sagara, I.; Beavogui, A.H.; Zongo, I.; Soulama, I.; Borghini-Fuhrer, I.; Fofana, B.; Camara, D.; Somé, A.F.; Coulibaly, A.S.; Traore, O.B.; et al. Safety and efficacy of re-treatments with pyronaridine-artesunate in African patients with malaria: A substudy of the WANECAM randomised trial. Lancet Infect. Dis. 2016, 16, 189–198. [Google Scholar] [CrossRef]
  76. West African Network for Clinical Trials of Antimalarial Drugs (WANECAM), T.W.A.N. for C.T. of A.D. Pyronaridine-artesunate or dihydroartemisinin-piperaquine versus current first-line therapies for repeated treatment of uncomplicated malaria: A randomised, multicentre, open-label, longitudinal, controlled, phase 3b/4 trial. Lancet 2018, 391, 1378–1390. [Google Scholar] [CrossRef]
  77. Leang, R.; Mairet-Khedim, M.; Chea, H.; Huy, R.; Khim, N.; Mey Bouth, D.; Dorina Bustos, M.; Ringwald, P.; Witkowski, B. Efficacy and safety of pyronaridine-artesunate plus single-dose primaquine for the treatment of uncomplicated Plasmodium falciparum malaria in eastern Cambodia. Antimicrob. Agents Chemother. 2019. [Google Scholar] [CrossRef]
  78. Leang, R.; Canavati, S.E.; Khim, N.; Vestergaard, L.S.; Borghini Fuhrer, I.; Kim, S.; Denis, M.B.; Heng, P.; Tol, B.; Huy, R.; et al. Efficacy and Safety of Pyronaridine-Artesunate for Treatment of Uncomplicated Plasmodium falciparum Malaria in Western Cambodia. Antimicrob. Agents Chemother. 2016, 60, 3884–3890. [Google Scholar] [CrossRef] [Green Version]
  79. Auparakkitanon, S.; Chapoomram, S.; Kuaha, K.; Chirachariyavej, T.; Wilairat, P. Targeting of hematin by the antimalarial pyronaridine. Antimicrob. Agents Chemother. 2006, 50, 2197–2200. [Google Scholar] [CrossRef]
  80. Madamet, M.; Briolant, S.; Amalvict, R.; Benoit, N.; Bouchiba, H.; Cren, J.; Pradines, B.; French National Centre for Imported Malaria Study Group. The Plasmodium falciparum chloroquine resistance transporter is associated with the ex vivo P. falciparum African parasite response to pyronaridine. Parasit. Vectors 2016, 9, 77. [Google Scholar] [CrossRef] [PubMed]
  81. Laufer, M.K.; Thesing, P.C.; Eddington, N.D.; Masonga, R.; Dzinjalamala, F.K.; Takala, S.L.; Taylor, T.E.; Plowe, C.V. Return of Chloroquine Antimalarial Efficacy in Malawi. N. Engl. J. Med. 2006, 355, 1959–1966. [Google Scholar] [CrossRef] [PubMed]
  82. Laufer, M.K.; Takala-Harrison, S.; Dzinjalamala, F.K.; Stine, O.C.; Taylor, T.E.; Plowe, C. V Return of chloroquine-susceptible falciparum malaria in Malawi was a reexpansion of diverse susceptible parasites. J. Infect. Dis. 2010, 202, 801–808. [Google Scholar] [CrossRef] [PubMed]
  83. Humphreys, G.S.; Merinopoulos, I.; Ahmed, J.; Whitty, C.J.M.; Mutabingwa, T.K.; Sutherland, C.J.; Hallett, R.L. Amodiaquine and artemether-lumefantrine select distinct alleles of the Plasmodium falciparum mdr1 gene in Tanzanian children treated for uncomplicated malaria. Antimicrob. Agents Chemother. 2007, 51, 991–997. [Google Scholar] [CrossRef] [PubMed]
  84. Pelleau, S.; Moss, E.L.; Dhingra, S.K.; Volney, B.; Casteras, J.; Gabryszewski, S.J.; Volkman, S.K.; Wirth, D.F.; Legrand, E.; Fidock, D.A.; et al. Adaptive evolution of malaria parasites in French Guiana: Reversal of chloroquine resistance by acquisition of a mutation in pfcrt. Proc. Natl. Acad. Sci. USA 2015, 112, 11672–11677. [Google Scholar] [CrossRef] [PubMed]
  85. Nsanzabana, C.; Djalle, D.; Guérin, P.J.; Ménard, D.; González, I.J. Tools for surveillance of anti-malarial drug resistance: An assessment of the current landscape. Malar. J. 2018, 17, 75. [Google Scholar] [CrossRef]
  86. Nsanzabana, C.; Ariey, F.; Beck, H.-P.; Ding, X.C.; Kamau, E.; Krishna, S.; Legrand, E.; Lucchi, N.; Miotto, O.; Nag, S.; et al. Molecular assays for antimalarial drug resistance surveillance: A target product profile. PLoS ONE 2018, 13, e0204347. [Google Scholar] [CrossRef]
  87. Hastings, I.M.; Watkins, W.M.; White, N.J. The evolution of drug-resistant malaria: The role of drug elimination half-life. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2002, 357, 505–519. [Google Scholar] [CrossRef]
  88. Maiga, H.; Lasry, E.; Diarra, M.; Sagara, I.; Bamadio, A.; Traore, A.; Coumare, S.; Bahonan, S.; Sangare, B.; Dicko, Y.; et al. Seasonal Malaria Chemoprevention with Sulphadoxine-Pyrimethamine and Amodiaquine Selects Pfdhfr-dhps Quintuple Mutant Genotype in Mali. PLoS ONE 2016, 11, e0162718. [Google Scholar] [CrossRef]
  89. Zuber, J.A.; Takala-Harrison, S. Multidrug-resistant malaria and the impact of mass drug administration. Infect. Drug Resist. 2018, 11, 299–306. [Google Scholar] [CrossRef] [PubMed]
  90. Tun, K.M.; Jeeyapant, A.; Myint, A.H.; Kyaw, Z.T.; Dhorda, M.; Mukaka, M.; Cheah, P.Y.; Imwong, M.; Hlaing, T.; Kyaw, T.H.; et al. Effectiveness and safety of 3 and 5 day courses of artemether–lumefantrine for the treatment of uncomplicated falciparum malaria in an area of emerging artemisinin resistance in Myanmar. Malar. J. 2018, 17, 258. [Google Scholar] [CrossRef] [PubMed]
  91. Priotto, G.; Kabakyenga, J.; Pinoges, L.; Ruiz, A.; Eriksson, T.; Coussement, F.; Ngambe, T.; Taylor, W.R.J.; Perea, W.; Guthmann, J.-P.; et al. Artesunate and sulfadoxine-pyrimethamine combinations for the treatment of uncomplicated Plasmodium falciparum malaria in Uganda: A randomized, double-blind, placebo-controlled trial. Trans. R. Soc. Trop. Med. Hyg. 2003, 97, 325–330. [Google Scholar] [CrossRef]
  92. Mutabingwa, T.K.; Anthony, D.; Heller, A.; Hallett, R.; Ahmed, J.; Drakeley, C.; Greenwood, B.M.; Whitty, C.J.M. Amodiaquine alone, amodiaquine+sulfadoxine-pyrimethamine, amodiaquine+artesunate, and artemether-lumefantrine for outpatient treatment of malaria in Tanzanian children: A four-arm randomised effectiveness trial. Lancet 2005, 365, 1474–1480. [Google Scholar] [CrossRef]
  93. Taylor, S.M.; Parobek, C.M.; Aragam, N.; Ngasala, B.E.; Mårtensson, A.; Meshnick, S.R.; Juliano, J.J. Pooled deep sequencing of Plasmodium falciparum isolates: An efficient and scalable tool to quantify prevailing malaria drug-resistance genotypes. J. Infect. Dis. 2013, 208, 1998–2006. [Google Scholar] [CrossRef] [PubMed]
  94. Nag, S.; Dalgaard, M.D.; Kofoed, P.-E.; Ursing, J.; Crespo, M.; Andersen, L.O.; Aarestrup, F.M.; Lund, O.; Alifrangis, M. High throughput resistance profiling of Plasmodium falciparum infections based on custom dual indexing and Illumina next generation sequencing-technology. Sci. Rep. 2017, 7, 2398. [Google Scholar] [CrossRef] [PubMed]
  95. Talundzic, E.; Ravishankar, S.; Kelley, J.; Patel, D.; Plucinski, M.; Schmedes, S.; Ljolje, D.; Clemons, B.; Madison-Antenucci, S.; Arguin, P.M.; et al. Next-Generation Sequencing and Bioinformatics Protocol for Malaria Drug Resistance Marker Surveillance. Antimicrob. Agents Chemother. 2018, 62, e02474-17. [Google Scholar] [CrossRef]
  96. Hooft van Huijsduijnen, R.; Wells, T.N. The antimalarial pipeline. Curr. Opin. Pharmacol. 2018, 42, 1–6. [Google Scholar] [CrossRef]
  97. Ashley, E.A.; Phyo, A.P. Drugs in Development for Malaria. Drugs 2018, 78, 861–879. [Google Scholar] [CrossRef]
  98. Worldwide Antimalarial Resistance Network (WWARN) AL Dose Impact Study Group. The effect of dose on the antimalarial efficacy of artemether-lumefantrine: A systematic review and pooled analysis of individual patient data. Lancet Infect. Dis. 2015, 15, 692–702. [Google Scholar] [CrossRef]
  99. Kloprogge, F.; Workman, L.; Borrmann, S.; Tékété, M.; Lefèvre, G.; Hamed, K.; Piola, P.; Ursing, J.; Kofoed, P.E.; Mårtensson, A.; et al. Artemether-lumefantrine dosing for malaria treatment in young children and pregnant women: A pharmacokinetic-pharmacodynamic meta-analysis. PLoS Med. 2018, 15, e1002579. [Google Scholar] [CrossRef] [PubMed]
  100. Ursing, J.; Kofoed, P.-E.; Rodrigues, A.; Blessborn, D.; Thoft-Nielsen, R.; Björkman, A.; Rombo, L. Similar efficacy and tolerability of double-dose chloroquine and artemether-lumefantrine for treatment of Plasmodium falciparum infection in Guinea-Bissau: A randomized trial. J. Infect. Dis. 2011, 203, 109–116. [Google Scholar] [CrossRef] [PubMed]
  101. Ursing, J.; Rombo, L.; Bergqvist, Y.; Rodrigues, A.; Kofoed, P.-E. High-Dose Chloroquine for Treatment of Chloroquine-Resistant Plasmodium falciparum Malaria. J. Infect. Dis. 2016, 213, 1315–1321. [Google Scholar] [CrossRef] [PubMed]
  102. Dipanjan, B.; Shivaprakash, G.; Balaji, O. Triple Combination Therapy and Drug Cycling—Tangential Strategies for Countering Artemisinin Resistance. Curr. Infect. Dis Rep. 2017, 19, 25. [Google Scholar] [CrossRef] [PubMed]
  103. Dini, S.; Zaloumis, S.; Cao, P.; Price, R.N.; Fowkes, F.J.I.; van der Pluijm, R.W.; McCaw, J.M.; Simpson, J.A. Investigating the Efficacy of Triple Artemisinin-Based Combination Therapies for Treating Plasmodium falciparum Malaria Patients Using Mathematical Modeling. Antimicrob. Agents Chemother. 2018, 62, e01068-18. [Google Scholar] [CrossRef] [PubMed]
  104. Rossi, G.; De Smet, M.; Khim, N.; Kindermans, J.-M.; Menard, D. Emergence of Plasmodium falciparum triple mutant in Cambodia. Lancet Infect. Dis. 2017, 17, 1233. [Google Scholar] [CrossRef] [Green Version]
  105. Wojnarski, M.; Lin, J.; Gosi, P.; Spring, M.; Vanachayangkul, P.; Boonyalai, N.; Kuntawunginn, W.; Chaisatit, C.; Kirativanich, K.; Saingam, P.; et al. The emergence of multidrug resistant malaria parasites in Southeast Asia and implications on future malaria treatment Itinerary. In Proceedings of the ASTMH Annual Meeting, New Orleans, LA, USA, 28 October–1 November 2018. [Google Scholar]
  106. Najer, A.; Palivan, C.G.; Beck, H.-P.; Meier, W. Challenges in Malaria Management and a Glimpse at Some Nanotechnological Approaches. Adv. Exp. Med. Biol. 2018, 1052, 103–112. [Google Scholar] [PubMed]
  107. Walvekar, P.; Gannimani, R.; Govender, T. Combination drug therapy via nanocarriers against infectious diseases. Eur. J. Pharm. Sci. 2019, 127, 121–141. [Google Scholar] [CrossRef] [PubMed]
  108. Bakshi, R.P.; Tatham, L.M.; Savage, A.C.; Tripathi, A.K.; Mlambo, G.; Ippolito, M.M.; Nenortas, E.; Rannard, S.P.; Owen, A.; Shapiro, T.A. Long-acting injectable atovaquone nanomedicines for malaria prophylaxis. Nat. Commun. 2018, 9, 315. [Google Scholar] [CrossRef] [Green Version]

Share and Cite

MDPI and ACS Style

Nsanzabana, C. Resistance to Artemisinin Combination Therapies (ACTs): Do Not Forget the Partner Drug! Trop. Med. Infect. Dis. 2019, 4, 26. https://0-doi-org.brum.beds.ac.uk/10.3390/tropicalmed4010026

AMA Style

Nsanzabana C. Resistance to Artemisinin Combination Therapies (ACTs): Do Not Forget the Partner Drug! Tropical Medicine and Infectious Disease. 2019; 4(1):26. https://0-doi-org.brum.beds.ac.uk/10.3390/tropicalmed4010026

Chicago/Turabian Style

Nsanzabana, Christian. 2019. "Resistance to Artemisinin Combination Therapies (ACTs): Do Not Forget the Partner Drug!" Tropical Medicine and Infectious Disease 4, no. 1: 26. https://0-doi-org.brum.beds.ac.uk/10.3390/tropicalmed4010026

Article Metrics

Back to TopTop