Skip to main content

Are k13 and plasmepsin II genes, involved in Plasmodium falciparum resistance to artemisinin derivatives and piperaquine in Southeast Asia, reliable to monitor resistance surveillance in Africa?

Abstract

Mutations in the propeller domain of Plasmodium falciparum kelch 13 (Pfk13) gene are associated with artemisinin resistance in Southeast Asia. Artemisinin resistance is defined by increased ring survival rate and delayed parasite clearance half-life in patients. Additionally, an amplification of the Plasmodium falciparum plasmepsin II gene (pfpm2), encoding a protease involved in hemoglobin degradation, has been found to be associated with reduced in vitro susceptibility to piperaquine in Cambodian P. falciparum parasites and with dihydroartemisinin–piperaquine failures in Cambodia. The World Health Organization (WHO) has recommended the use of these two genes to track the emergence and the spread of the resistance to dihydroartemisinin–piperaquine in malaria endemic areas. Although the resistance to dihydroartemisinin–piperaquine has not yet emerged in Africa, few reports on clinical failures suggest that k13 and pfpm2 would not be the only genes involved in artemisinin and piperaquine resistance. It is imperative to identify molecular markers or drug resistance genes that associate with artemisinin and piperaquine in Africa. K13 polymorphisms and Pfpm2 copy number variation analysis may not be sufficient for monitoring the emergence of dihydroartemisinin–piperaquine resistance in Africa. But, these markers should not be ruled out for tracking the emergence of resistance.

Background

According to the World Health Organization (WHO), artemisinin-based combination therapy (ACT) has been recommended as treatment of uncomplicated falciparum malaria since 2001. However, Plasmodium falciparum parasites resistant to artemisinin derivatives emerged in Southeast Asia, and more particularly in western Cambodia, Myanmar, Thailand and Laos [1,2,3,4,5,6]. More recently, the emergence of P. falciparum resistance to dihydroartemisinin–piperaquine was observed in Cambodia, where recrudescent infections had rapidly increased [7,8,9], and then in Vietnam [10, 11]. However, dihydroartemisinin–piperaquine is little-used in African countries for the treatment of uncomplicated malaria, where artemether–lumefantrine and/or artesunate–amodiaquine are currently used. Only Senegal has adopted dihydroartemisinin–piperaquine as a third alternative first-line regimen. Dihydroartemisinin–piperaquine has emerged as a potential combination for chemoprevention in pregnant women and children in Africa [12,13,14].

According to the WHO, the resistance to dihydroartemisinin–piperaquine has not yet emerged in Africa. There is currently no evidence of failing efficacy of dihydroartemisinin–piperaquine in Africa. The latest published studies showed that PCR-corrected adequate clinical and parasitological response (APCR) at day 42 ranged between 94.6 and 100% for the treatment of uncomplicated P. falciparum malaria in children treated between 2011 and 2017 in Africa (Tanzania, Rwanda, Mali, Guinea, Burkina Faso, Angola, Niger) [15,16,17,18,19,20]. However, there are some rare cases of clinical failures with dihydroartemisinin–piperaquine in Africa [15]. Two cases of late treatment failure after 30 and 32 days were reported in Italian travelers returning from Ethiopia and treated with dihydroartemisinin–piperaquine [21, 22]. Additionally, some clinical failures in travelers returning from Africa, and confirmed by an expected plasmatic level of dihydroartemisinin–piperaquine, were obtained in the French national reference centre for malaria (unpublished personal data). The genes involved in resistance to artemisinin derivatives and piperaquine in Southeast Asia do not properly explain these few clinical failures observed in Africa [22,23,24,25,26,27]. It is imperative to monitor the emergence of dihydroartemisinin–piperaquine resistance in Africa. But, are k13 and plasmepsin II genes reliable to survey resistance in Africa?

Artemisinin derivative resistance

The emergence and spread of resistance to artemisinin derivatives were observed in Southeast Asia [1,2,3,4,5,6]. This resistance was associated with delayed parasite clearance half-lives (> 5 h) after artemisinin-based monotherapy treatment or ACT [1, 4, 28, 29]. Additionally, slow in vivo parasite clearance half-live was correlated with in vitro resistance manifested an increase in the ring-stage survival rate after contact with 700 nM of artemisinin for 6 h, evaluated with a new phenotypic assay, the in vitro ring-stage survival assay or RSA [30,31,32,33].

Different molecular markers, associated with in vitro resistance to artemisinin derivatives measured by standard phenotypic assays, were previously proposed. Polymorphisms in the pfATPase6 gene, encoding the P. falciparum sarco-endoplasmic reticulum calcium-ATPase PfATPase 6 protein, were first associated with in vitro resistance [34], but not with in vivo delayed parasite clearance in P. falciparum parasites from the Thai-Cambodia border [35]. Amplification of the P. falciparum multidrug resistance 1 gene (pfmdr1) was also associated with in vitro reduced susceptibility to artemisinin derivatives [36,37,38], but never with delayed parasite clearance [39]. Additionally, mutations on pfmdr1 genes were shown to be correlated with in vitro reduced susceptibility to artemisinin derivatives [40,41,42]. The involvement of polymorphisms in potential genes was evaluated, such as pfubp-1 encoding the P. falciparum ubiquitin specific protease 1 [43,44,45], the gene encoding the RING E3 protein ubiquitin ligase [46, 47], pfap2mu encoding the P. falciparum adaptor protein complex 2 mu subunit [44, 48], pfmdr5 encoding the P. falciparum multidrug resistance 5 protein [49] or pfmdr6 encoding the P. falciparum multidrug resistance 6 protein [49,50,51]. Only mutations pfap2mu S160N and pfubp1 E1525D/Q were found in cases of African imported P. falciparum malaria with clinical failure with ACT [25]. Whole-genome sequencing of the artemisinin-susceptible F32-Tanzania strain and the artemisinin-resistant F32-ART line, obtained after 5 years of artemisinin pressure, led to identification of several mutations (M476I, C580Y, R539T, Y493H, I543T and P574L) in the propeller domain of the kelch 13 (k13) gene (PF3D7_1343700) that are associated with in vitro resistance to artemisinin [31, 52, 53]. These mutations were associated with artemisinin-resistant (high survival rate) Cambodian isolates evaluated with RSA [1, 31, 32]. Additionally, these mutations were also associated with in vivo delayed parasite clearance half-lives (> 5 h) in Southeast Asia, including Cambodia, Vietnam, Thailand, Myanmar and China [1, 31, 54] or parasitaemia still positive on day 3 after 7 days of artesunate monotherapy or 3 days of ACT [23]. Another mutation, F446I, was predominant in Myanmar and associated with high survival rate and P. falciparum in vivo delayed clearance [55,56,57,58].

According to the WHO, the proportion of patients still parasitaemic on day 3 (10%) or with a parasite slow clearance half-life above 5 h (10%) after artesunate monotherapy or treatment with ACT, or carrying k13 mutations associated with artemisinin resistance in Asia are indicators to identify emergence of suspected artemisinin resistance [59]. Resistance to artemisinin is confirmed when at least 5% of the patients carry parasites with k13 resistance-associated mutations are still parasitaemic on day 3 or show slow parasite clearance [59]. The WHO has recommended evaluate k13 resistance-associated mutations to track emergence and spread of artemisinin resistance in Africa.

The main k13 mutations involved in artemisinin resistance in Southeast Asia are not yet reported in Africa certainly due to an absence of artemisinin resistance in Africa [23, 60,61,62,63,64,65,66,67,68]. Artemisinin resistance due to k13 mutations has not disseminated to African countries yet. However, clinical failures with ACT, although rare, were reported in Africa (Angola, Senegal, Zaire) or in imported falciparum cases from Africa (Angola, Ethiopia, Liberia, Uganda) and were not associated with k13 resistance-associated mutations [21,22,23,24,25, 69,70,71]. In some cases, pharmacokinetic data were associated and allowed to exclude sub-therapeutic drug exposure to dihydroartemisinin [21, 22]. On Senegalese patients, parasites were still detected on day 3 after ACT treatment and were wild-type for K13 [24]. An isolate from Equatorial Guinea collected from patient with early treatment failure after artemisinin–piperaquine showed in vitro survival rate higher than the rate observed in the control strains but lower than rate in Asian artemisinin-resistant strain with a C580Y mutation [72]. However, none of the mutations described in artemisinin resistance in Asia was detected. A new mutation (M579I) was identified.

Additionally, 98.5% of Cambodian patients with isolates carrying C580Y or Y493H mutations on day 1 were negative on day 3 after dihydroartemisinin–piperaquine treatment [73]. Cambodian parasites with in vitro survival rates above the cut-off of 1% can lack the k13 mutations involved in artemisinin resistance in Cambodia [74]. Chinese patients with R539T mutant parasites imported from Angola and P574L mutant parasites from Equatorial Guinea all recovered after treatment with dihydroartemisinin–piperaquine [75].

These data suggest that other mechanisms than k13 mutations may explain artemisinin resistance, and more particularly in Africa. Mutations on falcipain 2a gene, encoding a cysteine protease and haemoglobinase and atg18 gene, encoding the autophagy-related protein 18, might be associated with artemisinin resistance in parasites from the China Myanmar-border [76,77,78]. Additionally, mutations in the actin-binding protein coronin (R100K, E107V or G50E) conferred high in vitro survival rate in Senegalese P. falciparum strains, and this in the absence of mutation on the k13 propeller gene [79].

Piperaquine resistance

Emergence of P. falciparum resistance to dihydroartemisinin–piperaquine was observed in Cambodia, where the prevalence of recrudescent infections rapidly increased [7,8,9], and then in Vietnam [10, 11]. Additionally, in vitro resistance to piperaquine was detected in Cambodia and increased rapidly between 2013 and 2015 [80]. Duplication of the Plasmodium falciparum plasmepsin II gene (pfpm2) (PF3D7_1408000), encoding a protease involved in haemoglobin degradation, has been found to be associated with reduced in vitro susceptibility to piperaquine in Cambodian P. falciparum parasites and with dihydroartemisinin–piperaquine failures in Cambodia [81, 82]. A new in vitro test, the piperaquine survival assay (PSA), was developed to follow piperaquine resistance [83]. Plasmodium falciparum dihydroartemisinin–piperaquine failures in Cambodia were associated with piperaquine survival rate above 10% or high piperaquine IC50 above 90 nM estimated by in vitro standard assay [81,82,83].

However, the involvement of pfpm2 in piperaquine resistance seems controversial in Africa. In Mali, the presence of P. falciparum isolates with pfpm2 duplications was confirmed in only 7 out of 65 clinical failures with dihydroartemisinin–piperaquine [26]. Three patients harbouring parasites with two copies of pfpm2 in Tanzania were successfully treated with dihydroartemisinin–piperaquine [84]. Additionally, only a single copy of pfpm2 was detected in two isolates collected in imported malaria cases from Ethiopia and Cameroon after dihydroartemisinin–piperaquine failures [22, 27]. The use of dihydroartemisinin–piperaquine as intermittent preventive treatment during pregnancy did not select for pfpm2 duplication in Uganda [85]. Ex vivo susceptibility to piperaquine in imported P. falciparum parasites from Africa, in Ugandan and Senegalese isolates was not associated with variation in pfpm2 copy number ([86,87,88], unpublished personal data). Additionally, a recent publication showed that overexpression of pfpm2 did not change the susceptibility of the 3D7 P. falciparum strain to piperaquine [89].

All these data suggest that pfpm2 would not be the only gene that explains the resistance to piperaquine in Africa. The P. falciparum chloroquine resistance transporter gene (pfcrt) may be a causal gene because piperaquine is a dimer of chloroquine. Mutations in pfcrt could be involved in piperaquine resistance. However, the K76T mutation involved in chloroquine resistance was not associated with in vitro and ex vivo resistance to piperaquine [90, 91]. Novel mutations in pfcrt, like H97Y, F145I, M343L, C350R or G353V, seem to confer in vitro resistance to piperaquine in P. falciparum parasites [92,93,94]. However, there is no direct evidence of piperaquine inhibiting PfCRT.

Conclusion

Mutations in K13 (C580Y, R539T, Y493H, I543T and P574L) and pfpm2 duplications in P. falciparum are associated with in vitro resistance and clinical failures with dihydroartemisinin–piperaquine in Southeast Asia. Although the resistance to dihydroartemisinin–piperaquine has not yet emerged in Africa, the first data on clinical failures and in vitro reduced susceptibility suggest that k13 and pfpm2 would be not the only genes involved in artemisinin and piperaquine resistance. It is imperative to identify new genes to explain resistance to artemisinin and piperaquine in Africa. It is necessary to maintain tracking of the emergence and spread of k13 and pfpm2 mutant parasites in Africa, which could be imported from Asia. This surveillance must be associated with the tracking of dihydroartemisinin–piperaquine clinical failures in Africa due to resistant parasites. Too few studies associate drug plasmatic measures to verify good compliance and pharmacokinetic to confirm resistance. African parasites may have their own genetic background preference to select dihydroartemisinin–piperaquine resistance which surely differs from Southeast Asian parasites. These isolates should be characterized by assessing k13 polymorphisms, pfpm2 copy number variation, but also other potential marker of resistance. The identification of new genes involved in dihydroartemisinin–piperaquine resistance in Africa could be performed by systematic analysis of African resistant parasites by genome wide association study (GWAS).

Availability of data and materials

Not applicable.

Abbreviations

ACT:

artemisinin-based combination therapy

ACPR:

adequate clinical and parasitological response

GWAS:

genome wide association study

PCR:

polymerase chain reaction

pfap2mu :

P. falciparum adaptor protein complex 2 mu gene

pfatg18 :

P. falciparum autophagy-related gene 18

pfATPase6 :

P. falciparum sarco-endoplasmic reticulum calcium-ATPase 6 gene

pfcrt :

P. falciparum chloroquine resistance transporter gene

pfmdr1 :

P. falciparum multidrug resistance 1 gene

pfmdr5 :

P. falciparum multidrug resistance 5 gene

pfmdr6 :

P. falciparum multidrug resistance 6 gene

pfubp-1 :

P. falciparum ubiquitin specific protease 1 gene

PSA:

in vitro piperaquine survival assay

RSA:

in vitro ring-stage survival assay

WHO:

World Health Organization

References

  1. Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, et al. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2014;371:411–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Noedl H, Se Y, Schaecher K, Smith BL, Socheat D, Fukuda MM. Evidence of artemisinin-resistant malaria in western Cambodia. New Engl J Med. 2008;359:2619–20.

    Article  CAS  PubMed  Google Scholar 

  3. Dondorp AM, Nosten F, Yi P, Das D, Phyo AP, Tarning J, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361:455–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Phyo AP, Nkhoma S, Stepniewska K, Ashley EA, Nair S, McGready R, et al. Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet. 2012;379:1960–6.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Bouillé M, Witkowski B, Duru V, Sriprawat K, Nair SK, McDew-White M, et al. Artemisinin-resistant Plasmodium falciparum K13 mutant alleles, Thailand-Myanmar border. Emerg Infect Dis. 2016;22:1503–5.

    Article  Google Scholar 

  6. Imwong M, Suwannasin K, Kunasol C, Sutawong K, Mayxay M, Rekol H, et al. The spread of artemisinin-resistant Plasmodium falciparum in the Greater Mekong subregion: a molecular epidemiology observational study. Lancet Infect Dis. 2017;17:491–7.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Amaratunga C, Lim P, Suon S, Sreng S, Mao S, Sopha C, et al. Dihydroartemisinin–piperaquine resistance in Plasmodium falciparum malaria in Cambodia: a multisite prospective cohort study. Lancet Infect Dis. 2016;16:357–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Leang R, Taylor WRJ, Bouth DM, Song L, Tarning J, Char MC, et al. Evidence of Plasmodium falciparum malaria multidrug resistance to artemisinin and piperaquine in Western Cambodia: dihydroartemisinin–piperaquine open-label multicenter clinical assessment. Antimicrob Agents Chemother. 2015;59:4719–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Spring MD, Lin JT, Manning JE, Vanachayangkul P, Somethy S, Bun R, et al. Dihydroartemisinin–piperaquine failure associated with a triple mutant including kelch13 C580Y in Cambodia: an observational cohort study. Lancet Infect Dis. 2015;15:683–91.

    Article  CAS  PubMed  Google Scholar 

  10. Phuc BQ, Rasmussen C, Duong TT, Dong LT, Loi MA, Ménard D, et al. Treatment failure of dihydroartemisinin/piperaquine for Plasmodium falciparum malaria, Vietnam. Emerg Infect Dis. 2017;23:715–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Thanh NV, Thuy-Nhien N, Tuyen NT, Tong NT, Nha-Ca NT, Dong LT, et al. Rapid decline in the susceptibility of Plasmodium falciparum to dihydroartemisinin–piperaquine in the south of Vietnam. Malar J. 2017;16:27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Kwambai TK, Dhabangi A, Idro R, Opoka R, Kariuki S, Samuels AM, et al. Malaria chemoprevention with monthly dihydroartemisinin–piperaquine for the post-discharge management of severe anaemia in children aged less than 5 years in Uganda and Kenya: study protocol for a multi-centre, two-arm, randomised, placebo-controlled, superiority trial. Trials. 2018;19:610.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Kajubi R, Ochieng T, Kakuru A, Jagannathan P, Nakalembe M, Ruel T, et al. Monthly sulfadoxine-pyrimethamine versus dihydroartemisinin–piperaquine for intermittent preventive treatment of malaria in pregnancy: a double-blind, randomized, controlled, superiority trial. Lancet. 2019;393:1428–39.

    Article  CAS  PubMed  Google Scholar 

  14. Jagannathan P, Kakuru A, Okiring J, Muhindo MK, Natureeba P, Nakalembe M, et al. Dihydroartemisinin–piperaquine for intermittent preventive treatment of malaria during pregnancy and risk of malaria in early childhood: a randomized controlled trial. PLoS Med. 2018;15:e1002606.

    Article  PubMed  PubMed Central  Google Scholar 

  15. The West African Network for Clinical Trials of Antimalarial Drugs (WANECAM). Pyronaridine–artesunate or dihydroartemisinin–piperaquine versus current first-line therapies for repeated treatment of uncomplicated malaria: a randomized, multicentre, open-label, longitudinal, controlled, phase 3b/4 trial. Lancet. 2018;391:1378–90.

    Article  PubMed Central  Google Scholar 

  16. Mandara CI, Francis F, Chiduo MG, Ngasala B, Mandike R, Mkude S, et al. High cure rates and tolerability of artesunate-amodiaquine and dihydroartemisinin–piperaquine for the treatment of uncomplicated falciparum malaria in Kibaha and Kigoma, Tanzania. Malar J. 2019;18:99.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Madara CI, Kavishe RA, Gesase S, Mghamba J, Ngadaya E, Mmbuji P, et al. High efficacy of artemether-lumefantrine and dihydroartemisinin–piperaquine for the treatment of uncomplicated falciparum malaria in Muheza and Kigoma Districts, Tanzania. Malar J. 2018;17:261.

    Article  Google Scholar 

  18. Uwimana A, Penkunas MJ, Nisingizwe MP, Warsame M, Umalissa N, Uyizeye D, et al. Efficacy of artemether-lumefantrine versus dihydroartemisinin–piperaquine for the treatment of uncomplicated malaria among children in Rwanda: an open-label, randomized controlled trial. Trans R Soc Trop Med Hyg. 2019;113:312–9.

    Article  PubMed  Google Scholar 

  19. Davlantes E, Dimbu PR, Ferreira CM, Joao MF, Pode D, Félix J, 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.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Grandesso F, Guindo O, Messe LW, Makarimi R, Traore A, Dama S, et al. Efficacy of artesunate–amodiaquine, dihydroartemisinin–piperaquine and artemether–lumefantrine for the treatment of uncomplicated Plasmodium falciparum malaria in Maradi, Niger. Malar J. 2018;17:52.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Gobbi F, Buonfrate D, Menegon M, Lunardi G, Angheben A, Severini C, et al. Failure of dihydroartemisinin–piperaquine treatment of uncomplicated Plasmodium falciparum malaria in a traveller coming from Ethiopia. Malar J. 2016;15:525.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Russo G, L’Episcopia M, Menegon M, Souza SS, Dongho BGD, Vullo V, et al. Dihydroartemisinin–piperaquine treatment failure in uncomplicated Plasmodium falciparum malaria case imported from Ethiopia. Infection. 2018;46:867–70.

    Article  CAS  PubMed  Google Scholar 

  23. Ménard D, Khim N, Beghain J, Adegnika AA, Alam MS, Amodu O, et al. A worldwide map of Plasmodium falciparum K13-propeller polymorphisms. N Engl J Med. 2016;374:2453–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Madamet M, Kouta MB, Wade KA, Lo G, Diawara S, Fall M, et al. Absence of association between polymorphisms in the K13 gene and the presence of Plasmodium falciparum parasites at day 3 after treatment with artemisinin derivatives in Senegal. Int J Antimicrob Agents. 2017;49:754–6.

    Article  CAS  PubMed  Google Scholar 

  25. Sutherland CJ, Lansdell P, Sanders M, Muwanguzi J, van Schalkwyk DA, Kaur H, et al. pfk13-independent treatment failure in four imported cases of Plasmodium falciparum malaria treated with artemether–lumefantrine in the United Kingdom. Antimicrob Agents Chemother. 2017;61:e02382-16.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Inoue J, Silva M, Fofana B, Sanogo K, Martensson A, Sagara I, et al. Plasmodium falciparum plasmepsin 2 duplications, West Africa. Emerg Infect Dis. 2018;24:1591–3.

    Article  CAS  PubMed Central  Google Scholar 

  27. Malvy D, Torrentino-Madamet M, L’Ollivier C, Receveur MC, Jeddi F, Delhaes L, et al. Plasmodium falciparum recrudescence two years after treatment of an uncomplicated infection without return to an area where malaria is endemic. Antimicrob Agents Chemother. 2018;62:e01892-17.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Flegg JA, Guerin PJ, White NJ, Stepniewska K. Standardizing the measurement of parasite clearance in falciparum malaria: the parasite clearance estimator. Malar J. 2011;10:339.

    Article  PubMed  PubMed Central  Google Scholar 

  29. WWARN Parasite Clearance Study Group, Abdulla S, Ashley EA, Bassat Q, Bethell D, Björkman A, et al. Baseline data of parasite clearance in patients with falciparum malaria treated with artemisinin derivative: an individual patient data meta-analysis. Malar J. 2015;14:359.

    Article  PubMed Central  CAS  Google Scholar 

  30. Witkowski B, Amaratunga C, Khim N, Sreng S, Chim P, Kim S, et al. Novel phenotypic assays for the detection of artemisinin-resistant Plasmodium falciparum malaria in Cambodia: in vitro and ex vivo drug-response studies. Lancet Infect Dis. 2013;13:1043–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois AC, Khim N, et al. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature. 2014;505:50–5.

    Article  PubMed  CAS  Google Scholar 

  32. Amaratunga C, Witkowski B, Dek D, Try V, Khim N, Miotto O, et al. Plasmodium falciparum founder populations in western Cambodia have reduced artemisinin sensitivity in vitro. Antimicrob Agents Chemother. 2014;58:4935–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Amaratunga C, Witkowski B, Khim N, Ménard D, Fairhurst RM. Artemisinin resistance in Plasmodium falciparum. Lancet Infect Dis. 2014;14:449–50.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Jambou R, Legrand E, Niang M, Khim N, Lim P, Volney B, et al. Resistance of Plasmodium falciparum field isolates to in vitro artemether and point mutations of the SERCA-type PfATPase6. Lancet. 2005;366:1960–3.

    Article  CAS  PubMed  Google Scholar 

  35. Noedl H, Se Y, Sriwichai S, Schaecher K, Teja-Isavadharm P, Smith B, et al. Artemisinin resistance in Cambodia: a clinical trial designed to address an emerging problem in Southeast Asia. Clin Infect Dis. 2010;51:82–9.

    Article  Google Scholar 

  36. Sidhu AB, Uhleman AC, Valderramos SG, Valderramos JC, Krishna S, Fidock DA. Decreasing pfmdr1 copy number in Plasmodium falciparum malaria heightens susceptibility to mefloquine, lumefantrine, halofantrine, quinine, and artemisinin. J Infect Dis. 2006;194:528–35.

    Article  PubMed  Google Scholar 

  37. Pascual A, Fall B, Wurtz N, Fall M, Camara C, Nakoulima A, et al. Plasmodium falciparum with multidrug resistance 1 gene duplications, Senegal. Emerg Infect Dis. 2013;19:814–5.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Chavchich M, Gerena L, Peters J, Chen N, Cheng Q, Kyle DE. Role of pfmdr1 amplification and expression in induction of resistance to artemisinin derivatives in Plasmodium falciparum. Antimicrob Agents Chemother. 2010;54:2455–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ngalah BS, Ingasia LA, Cheruiyot AC, Chebon LJ, Juma DW, Muiruri P, et al. Analysis of major genome loci underlying artemisinin resistance and pfmdr1 copy number in pre- and post-ACTs in western Kenya. Sci Rep. 2015;5:8308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Duraisingh MT, Roper C, Walliker D, Warhurst DC. Increased sensitivity to the antimalarials mefloquine and artemisinin is conferred by mutations in the pfmdr1 gene of Plasmodium falciparum. Mol Microbiol. 2000;36:955–61.

    Article  CAS  PubMed  Google Scholar 

  41. Wurtz N, Fall B, Pascual A, Fall M, Baret E, Camara C, et al. Role of Pfmdr1 in in vitro Plasmodium falciparum susceptibility to chloroquine, quinine, monodesethylamodiaquine, mefloquine, lumefantrine, and dihydroartemisinin. Antimicrob Agents Chemother. 2014;58:7032–40.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Njokah MJ, Kangethe JN, Kinyua J, Kariuki D, Kimani FT. In vitro selection of Plasmodium falciparum Pfcrt and Pfmdr1 variants by artemisinin. Malar J. 2016;15:381.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Imwong M, Dondorp AM, Nosten F, Yi P, Mungthin M, Hanchana S, et al. Exploring the contribution of candidate genes to artemisinin resistance in Plasmodium falciparum. Antimicrob Agents Chemother. 2010;54:2886–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Henriques G, Hallett RL, Beshir KB, Gadalla NB, Johnson RE, Burrow R, et al. Directional selection at the pfmdr1, pfcrt, pfubp1, and pfap2mu loci of Plasmodium falciparum in Kenya children treated with ACT. J Infect Dis. 2014;201:2001–8.

    Article  CAS  Google Scholar 

  45. Borrman S, Straimer J, Mwai L, Abdi A, Rippert A, Okombo J, et al. Genome-wide screen identifies new candidate genes associated with artemisinin susceptibility in Plasmodium falciparum in Kenya. Sci Rep. 2013;3:3318.

    Article  Google Scholar 

  46. Gendrot M, Fall B, Madamet M, Fall M, Wade KA, Amalvict R, et al. Absence of association between polymorphisms in the RING E3 ubiquitin protein ligase gene and ex vivo susceptibility to conventional antimalarial drugs in Plasmodium falciparum isolates from Dakar, Senegal. Antimicrob Agents Chemother. 2016;60:5010–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gendrot M, Foguim FT, Robert MG, Amalvict R, Mosnier J, Benoit M, et al. The D113N mutation in the RING E3 ubiquitin protein ligase gene is not associated with ex vivo susceptibility to common anti-malarial drugs in African Plasmodium falciparum isolates. Malar J. 2018;17:108.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Adams T, Ennuson NAA, Quashie NB, Futagbi G, Matrevi S, Hagan OCK, et al. Prevalence of Plasmodium falciparum delayed clearance associated polymorphisms in adaptor protein complex 2 mu subunit (pfap2mu) and ubiquitin specific protease 1 (pfubp1) genes in Ghanaian isolates. Parasit Vectors. 2018;11:175.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Okombo J, Abdi AI, Kiara SM, Mwai L, Pole L, Sutherland CJ, et al. Repeat polymorphisms in the low-complexity regions of Plasmodium falciparum ABC transporters and associations with in vitro antimalarial responses. Antimicrob Agents Chemother. 2013;57:6196–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang Z, Parker D, Meng H, Wu L, Li J, Zhao Z, et al. In vitro sensitivity of Plasmodium falciparum from China-Myanmar border area to major ACT drugs and polymorphisms in potential genes. PLoS ONE. 2012;7:e30927.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gendrot M, Diawara S, Madamet M, Kounta MB, Briolant S, Wade KA, et al. Association between polymorphisms in the Pfmdr6 gene and ex vivo susceptibility to quinine in Plasmodium falciparum parasites from Dakar, Senegal. Antimicrob Agents Chemother. 2017;61:e01183-16.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Witkowski B, Lelièvre J, Barragan MJ, Laurent V, Su XZ, Berry A, et al. Increased tolerance to artemisinin in Plasmodium falciparum is mediated by a quiescence mechanism. Antimicrob Agents Chemother. 2010;54:1872–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Straimer J, Gnädig NF, Witkowski B, Amaratunga C, Duru V, Pramundita Ramadani A, et al. K-13propeller mutations confer artemisinin resistance in Plasmodium falciparum clinical isolates. Science. 2015;347:428–31.

    Article  CAS  PubMed  Google Scholar 

  54. Takala-Harrison S, Jacob CG, Arze C, Cummings MP, Silva JC, Dondorp AM, et al. Independent emergence of artemisinin resistance mutations among Plasmodium falciparum in Southeast Asia. J Infect Dis. 2015;211:670–9.

    Article  CAS  PubMed  Google Scholar 

  55. Huang F, Takala-Harrison S, Jacob CG, Liu H, Sun X, Yang H, et al. A single mutation in K13 predominates in Southern China and is associated with delayed clearance of Plasmodium falciparum following artemisinin treatment. J Infect Dis. 2015;212:1629–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wang Z, Wang Y, Cabrera M, Zhang Y, Gupta B, Wu Y, et al. Artemisinin resistance at the China-Myanmar border and association with mutations in the K13 gene. Antimicrob Agents Chemother. 2015;59:6952–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bonnington CA, Phyo AP, Ashley EA, Imwong M, Sriprawat K, Parker DM, et al. Plasmodium falciparum Kelch 13 mutations and treatment response in patients in Hpa-Pun District, Northern Kayin State, Myanmar. Malar J. 2017;16:480.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Wang J, Huang Y, Zhao Y, Zhang D, Pan W. Introduction of F446I mutation in the K13 propeller gene leads to increased ring survival rates in Plasmodium falciparum isolates. Malar J. 2018;17:248.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. WHO. Status report on artemisinin resistance (September 2014). Geneva: World Health Organization; 2014.

    Google Scholar 

  60. Torrentino-Madamet M, Fall B, Benoit N, Camara C, Amalvict R, Fall M, et al. Limited polymorphisms in k13 gene in Plasmodium falciparum isolates from Dakar, Senegal in 2012–2013. Malar J. 2014;13:472.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Boussaroque A, Fall A, Madamet M, Camara C, Benoit N, Fall M, et al. Emergence of mutations in the K13 Propeller gene of Plasmodium falciparum isolates from Dakar, Senegal in 2013–2014. Antimicrob Agents Chemother. 2015;60:624–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Hawkes M, Conroy AL, Opoka RO, Namasopo S, Zhong K, Liles WC, et al. Slow clearance of Plasmodium falciparum in severe pediatric malaria, Uganda, 2011–2013. Emerg Infect Dis. 2015;21:1237–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kamau E, Campino S, Amenga-Etego L, Drury E, Ishengoma D, Johnson K, et al. K13-propeller polymorphisms in Plasmodium falciparum parasites from sub-saharan Africa. J Infect Dis. 2015;211:1352–5.

    CAS  PubMed  Google Scholar 

  64. Ouattara A, Kone A, Adams M, Fofana B, Maiga AW, Hampton S, et al. Polymorphisms in the K13-propeller gene in artemisinin-susceptible Plasmodium falciparum parasites from Bougoula-Hameau and Bandiagara, Mali. Am J Trop Med Hyg. 2015;92:1202–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Taylor SM, Parobek CM, DeConti DK, Kayentao K, Coulibaly SO, Greenwood BM, et al. Absence of putative artemisinin resistance mutations among Plasmodium falciparum in sub-Saharan Africa: a molecular epidemiologic study. J Infect Dis. 2015;211:680–8.

    Article  CAS  PubMed  Google Scholar 

  66. Torrentino-Madamet M, Collet L, Lepère JF, Benoit N, Amalvict R, Ménard D, et al. K13-propeller polymorphisms in Plasmodium falciparumisolates from patients in Mayotte in 2013 and 2014. Antimicrob Agents Chemother. 2015;59:7878–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Muwanguzi J, Henriques G, Sawa P, Bousema T, Sutherland CJ, Beshir KB. Lack of K13 mutations in Plasmodium falciparum persisting after artemisinin combination therapy treatment of Kenyan children. Malar J. 2016;15:36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Voumbo-Matoumona DF, Kouna LC, Madamet M, Maghendji-Nzondo S, Pradines B, Lekana-Douki JB. Prevalence of Plasmodium falciparum antimalarial drug resistance genes in Southeastern Gabon from 2011 to 2014. Infect Drug Resist. 2018;11:1329–38.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Dieye B, Affara M, Sangare L, Joof F, Ndiaye YD, Gomis JF, et al. West Africa international centers of excellence for malaria research: drug resistance patterns to artemether-lumefantrine in Senegal, Mali, and The Gambia. Am J Trop Med Hyg. 2016;95:1054–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Plucinski MM, Talundzic E, Morton L, Dimbu PR, Macaia AP, Fortes F, 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;59:437–43.

    Article  PubMed  CAS  Google Scholar 

  71. Plucinski MM, Dimbu PR, Macaia AP, Ferreira CM, Samutondo C, Quivinja J, 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.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Lu F, Culleton R, Zhang M, Ramaprasad A, von Seidlein L, Zhou H, et al. Emergence of indigenous artemisinin-resistant Plasmodium falciparum in Africa. New Engl J Med. 2017;376:991–3.

    Article  PubMed  Google Scholar 

  73. Kheang ST, Sovannaroth S, Ek S, Chy S, Chhun P, Mao S, et al. Prevalence of k13 mutation and day-3 positive parasitaemia in artemisinin-resistant malaria endemic area of Cambodia: a cross-sectional study. Malar J. 2017;16:372.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Mukherjee A, Bopp S, Magistrado P, Wong W, Daniels R, Demas A, et al. Artemisinin resistance without pfkelch13 mutations in Plasmodium falciparum isolates from Cambodia. Malar J. 2017;16:195.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Yang C, Zhang H, Zhou R, Qian D, Liu Y, Zhao Y, et al. Polymorphisms of Plasmodium falciparum k13-propeller gene among migrant workers returning to Henan Province, China from Africa. BMC Infect Dis. 2017;17:560.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Siddiqui FA, Cabrera M, Wang M, Brashear A, Kemirembe K, Wang Z, et al. Plasmodium falciparum Falcipain-2-a polymorphisms in Southeast Asia and their association with artemisinin resistance. J Infect Dis. 2018;218:434–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wang Z, Cabrera M, Yang L, Gupta B, Liang X, Kemirembe K, et al. Genome-wide association analysis identifies genetic loci associated with resistance to multiple antimalarials in Plasmodium falciparum from China-Myanmar border. Sci Rep. 2016;3:33891.

    Article  CAS  Google Scholar 

  78. Breglio KF, Amato R, Eastman R, Lim P, Sa JM, Guha R, et al. A single nucleotide polymorphism in the Plasmodium falciparum atg18 gene associates with artemisinin resistance and confers enhanced parasite survival under nutrient deprivation. Malar J. 2018;17:391.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Demas AR, Sharma AI, Wong W, Early AM, Redmond S, Bopp S, et al. Mutations in Plasmodium falciparum actin-binding protein coronin confer reduced artemisinin susceptibility. Proc Natl Acad Sci USA. 2018;115:12799–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Chaorattanakawee S, Lon C, Jongsakul K, Gawee J, Sok S, Sundrakes S, et al. Ex vivo piperaquine resistance developed rapidly in Plasmodium falciparum isolates in northern Cambodia compared to Thailand. Malar J. 2016;15:519.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Witkowski B, Duru V, Khim N, Ross LS, Saintpierre B, Beghain J, et al. A surrogate marker of piperaquine-resistant Plasmodium falciparum malaria: a phenotype-genotype association study. Lancet Infect Dis. 2016;17:174–83.

    Article  PubMed  CAS  Google Scholar 

  82. Amato R, Lim P, Miotto O, Amaratunga C, Dek D, Pearson RD, et al. Genetic markers associated with dihydroartemisinin-piperaquine failure in Plasmodium falciparum malaria in Cambodia: a genotype-phenotype association study. Lancet Infect Dis. 2016;17:164–73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Duru V, Khim N, Leang R, Kim S, Domergue A, Kloeung N, et al. Plasmodium falciparum dihydroartemisinin-piperaquine failures in Cambodia are associated with mutant K13 parasites presenting high survival rates in novel piperaquine in vitro assays: retrospective and prospective investigations. BMC Med. 2015;13:305.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Kakolwa MA, Mahende MK, Ishengoma DS, Mandara CI, Ngasala B, Kamugisha E, et al. Efficacy and safety of artemisinin-based combination therapy and molecular markers for artemisinin and piperaquine resistance in Mailand Tanzania. Malar J. 2018;17:369.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Conrad MD, Mota D, Foster M, Tukwasibwe S, Legac J, Tumwebase P, et al. Impact of intermittent preventive treatment during pregnancy on Plasmodium falciparum drug resistance-mediating polymorphisms in Uganda. J Infect Dis. 2017;2016:1008–17.

    Article  CAS  Google Scholar 

  86. Rasmussen SA, Ceja FG, Conrad MD, Tumwebase PK, Byaruhanga O, Katairo T, et al. Changing antimalaria drug sensitivities in Uganda. Antimicrob Agents Chemother. 2017;61:e01516-17.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Robert MG, Foguim Tsombeng F, Gendrot M, Mosnier J, Amalvict R, Benoit N, et al. Absence of a high level of duplication of the Plasmepsin II gene in Africa. Antimicrob Agents Chemother. 2018;62:e00374-18.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Robert MG, Foguim Tsombeng F, Gendrot M, Diawara S, Madamet M, Kounta MB, et al. Baseline ex vivo and molecular responses of Plasmodium falciparum isolates to piperaquine before implementation of dihydroartemisinin-piperaquine in Senegal. Antimicrob Agents Chemother. 2019;63:e02445-18.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Loesbanluechai D, Kotanan N, de Cozar C, Kochakarn T, Ansbro MR, Chotivanich K, et al. Overexpression of plasmepsin II and plasmepsin III does not directly cause reduction in Plasmodium falciparum sensitivity to artesunate, chloroquine and piperaquine. Int J Parasitol Drugs Drug Resist. 2019;9:16–22.

    Article  PubMed  Google Scholar 

  90. Briolant S, Henry M, Oeuvray C, Amalvict R, Baret E, Didillon E, et al. Absence of association between in vitro responses and polymorphisms in the pfcrt, pfmdr1, pfmrp, and pfnhe genes in Plasmodium falciparum. Antimicrob Agents Chemother. 2010;54:3537–44.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Pascual A, Madamet M, Bertaux L, Amalvict R, Benoit N, Travers D, et al. In vitro piperaquine susceptibility is not associated with the Plasmodium falciparum chloroquine resistance transporter gene. Malar J. 2013;12:431.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Pelleau S, Moss EL, Dhingra SK, Volney B, Casteras J, Gabryszewski SJ, et al. Adaptative evolution of malaria parasites in French Guiana: reversal of chloroquine resistance by acquisition of a mutation in pfcrt. Proc Natl Aca Sci USA. 2015;112:11672–7.

    Article  CAS  Google Scholar 

  93. Agrawal S, Moser KA, Morton L, Cummings MP, Parihar A, Dwivedi A, et al. A novel mutation in the Plasmodium falciparum chloroquine resistance transporter is associated with decreased piperaquine sensitivity. J Infect Dis. 2017;216:468–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Ross LS, Dhingra SK, Mok S, Yeo T, Wicht KJ, Kümpornsin K, et al. Emerging Southeast Asian PFCRT mutations confer Plasmodium falciparum resistance to the first-line antimalarial piperaquine. Nat Commun. 2018;9:3314.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

FFT, MG, MGR, MM, and BP drafted the manuscript. All the authors read and approved the final manuscript.

Corresponding author

Correspondence to Bruno Pradines.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Foguim Tsombeng, F., Gendrot, M., Robert, M.G. et al. Are k13 and plasmepsin II genes, involved in Plasmodium falciparum resistance to artemisinin derivatives and piperaquine in Southeast Asia, reliable to monitor resistance surveillance in Africa?. Malar J 18, 285 (2019). https://doi.org/10.1186/s12936-019-2916-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12936-019-2916-6

Keywords