Skip to main content

Failure of dihydroartemisinin plus piperaquine treatment of falciparum malaria by under-dosing in an overweight patient

Abstract

Background

Artemisinin-based combination therapy (ACT) introduced in the mid-1990s has been recommended since 2005 by the World Health Organization as first-line treatment against Plasmodium falciparum in all endemic countries. In 2010, the combination dihydroartemisinin–piperaquine (DP) was recommended for the treatment of uncomplicated P. falciparum malaria. DP is one of the first-line treatments used by the French army since 2013.

Case presentation

A case of P. falciparum clinical failure with DP at day 20 was described in a 104 kg French soldier deployed in Djibouti. He was admitted to hospital for supervision of oral treatment with DP [40 mg dihydroartemisinin (DHA) plus 320 mg piperaquine tetraphosphate (PPQ)]. This corresponded to a cumulative dose of 4.6 mg/kg DHA and 37 mg/kg PPQ in the present patient, which is far below the WHO recommended ranges. No mutation was found in the propeller domain of the Kelch 13 (k13) gene, which is associated with artemisinin resistance in Southeast Asia. Pfmdr1 N86, 184F, S1034 and N1042 polymorphisms and haplotype 72–76 CVIET for the pfcrt gene were found in the present case. There was no evidence of resistance to DP.

Conclusion

This case confirms the risk of therapeutic failure with dihydroartemisinin–piperaquine by under-dosing in patients weighing more than 100 kg. This therapeutic failure with DP by under-dosing highlighted the importance of appropriate dosing guidelines and the need of research data (efficacy, pharmacokinetics and pharmacodynamics) in over-weight patient group.

Background

Artemisinin-based combination therapy (ACT) introduced in the mid-1990s has been recommended since 2005 by the World Health Organization (WHO) as first-line treatment against Plasmodium falciparum in all endemic countries. Combined with wide-spread use of insecticide-treated bed nets, ACT has led to a significant reduction in malaria incidence and mortality, especially in sub-Saharan Africa [1]. However, with 214 million cases globally leading to an estimated 438,000 deaths worldwide in 2015, malaria remains a public health concern [2]; moreover, ACT therapeutic failures are increasing in Cambodia, where resistance to previous first-line anti-malarial drugs also first emerged [3]. Today, artemisinin resistance has been documented in Southeast Asia and must be closely monitored [4].

Clinical resistance, defined by a slow parasite clearance [3], is in vitro correlated with an increase in the ring-stage survival rate after contact with artemisinin [5]. Recently, mutations in the propeller domain of the Kelch 13 (k13) gene (PF3D71343700), especially the Y493H, R539T, I543T, and C580Y mutations, have been associated with in vivo and in vitro resistance to artemisinin in Southeast Asia P. falciparum strains [6–8]. Currently, these mutations have not been identified in African P. falciparum isolates [9–15]. K13 mutations as well as patients with slow clearing parasites were found in and some African countries (Kenya, Democratic Republic of Congo and Nigeria) [4]. However, there was no confirmation about spreading artemisinin resistance to Africa.

Nevertheless, addressing ACT-failure requires a systematic approach, and the conclusion of drug resistance is based on a body of clinical and scientific arguments. Poor compliance (insufficient oversight or intolerance) and under-dosing (insufficient dosage, poor absorption, drug interactions or defects in drug metabolism) may also contribute.

In this report, a failure of dihydroartemisinin–piperaquine (DP) anti-malarial treatment was identified in a patient weighing 104 kg.

Case presentation

A 35-year-old French soldier was deployed to Djibouti. He had no medical history, no recent trips except a return to France during summer holidays and intermittent observance of doxycycline (100 mg/day) malaria prophylaxis. He presented with fever, headaches, and diarrhoea for 4 days with paracetamol self-medication. His weight was 104 kg. His blood pressure was 143/78 mmHg, and his pulse rate was 84 bpm. A rapid diagnostic test (RDT) for malaria was positive for P. falciparum (Core Malaria™ pan/pv/pf, Fumouze diagnostics). Therefore, he was referred to a military hospital. Laboratory tests revealed a few abnormalities: lymphopenia (0.6 × 109/l lymphocyte count) and thrombocytopenia (30 × 109/l platelet count) without anaemia and moderate elevation in hepatic enzymes (ASAT 125 UI/l and ALAT 147 UI/l) and in bilirubin (30 µmol/l). Parasite detection in blood using quantitative buffy coat (QBC) was positive, and examination of thin blood smear stained by the May-Grünwald-Giemsa method confirmed the diagnosis of P. falciparum infection with parasitaemia <0.01 %. Blood and faecal culture were negative, as were dengue serology and NS1 antigen detection. He met none of the clinical or biological WHO criteria for severe P. falciparum malaria and was admitted to hospital for supervision of oral treatment with DP [40 mg dihydroartemisinin (DHA) plus 320 mg piperaquine tetraphosphate (PPQ); Eurartesim©; Sigma-Tau laboratory] at the maximal dosage of four pills once daily between meals for 3 days, recommended by the Sigma-Tau laboratory in absence of data on which to base a dose recommendation in patients weighing above 100 kg. He was discharged at the third day with no more fever and a negative QBC test. He took the correct doxycycline prophylaxis and used a bed-net. He was asymptomatic at day 12 with a negative QBC test. At day 20, he complained of fever (38.9 °C) and diarrhoea. Laboratory examination revealed recurrence of P. falciparum with 0.5 % parasitaemia and the absence of co-infection. A thin blood smear found late-stage trophozoites without gametocytes. He was successfully cured by the standard oral therapeutic course of a fixed atovaquone–proguanil combination [atovaquone (250 mg) plus proguanil hydrochloride (100 mg), Malarone®], four pills during meals once daily for 3 days. Clinical and parasitological follow-up were completed at day 22, 27, and 48 without recurrence.

Though ACT failure is infrequent in Africa, the genome of the recurrent P. falciparum isolate was sequenced and searched for polymorphisms involved in antimalarial drug resistance. The k13 gene was amplified by polymerase chain reaction (PCR) with the nested PCR method previously described [13]. The following primers were used for PCR: 3′ GGG AAT CTG GTG GTA ACA GC 5′ and 3′ CGG AGT GAC CAA ATC TGG GA 5′; 3′ GCC TTG TTG AAA GAA GCA GA 5′ and 3′ GCC AAG CTG CCA TTC ATT TG 5′. The K13 propeller gene was successfully sequenced, and the result was compared to the reference 3D7 strain. No mutation was identified in the propeller domain of the k13 gene. Two fragments of the pfmdr1 gene (P. falciparum multidrug resistance 1) were also amplified according to the description by Wurtz et al., revealing N86, 184F, S1034 and N1042 alleles [16]. The pfcrt gene (P. falciparum chloroquine resistance transporter) was also sequenced, as previously described [17], and the 72–76 haplotype found was CVIET. Finally, the copy number of the two genes pfmdt and pftetQ, which in vitro reduced susceptibility to doxycycline in Africa, was determined by finding one copy of pfmdt and two copies of pftetQ [18–20].

Discussion

Artemisinin derivatives clear parasitaemia more rapidly than all other currently available anti-malarial agents, including atovaquone–proguanil [21]. In 2010, DP was recommended by WHO [22] for the treatment of uncomplicated P. falciparum malaria, providing a promising alternative to other artemisinin-based combinations on the basis of its high efficacy, excellent safety profile, once-daily dosing scheme, and prolonged post-treatment prophylactic protection [23]. In the French army, DP is the available first-line treatment.

In this case, P. falciparum reinfection cannot be excluded because of the lack of comparative genome sequencing between the two isolates. However, the low P. falciparum transmission in Djibouti [24–26] and the 19-day delay both indicated that malaria relapse had occurred; this was further supported by the fact that this occurred despite prophylaxis, and only late trophozoite forms of the parasites without gametocytes appeared on the thin blood smear.

DP was well tolerated, and the whole course of treatment was well completed. Failure more likely resulted from inadequate dosage, because there are no data on which to base a dose recommendation in patients weighing above 100 kg according to the European Medicines Agency (EMA) [27]. In the absence of toxicity data in patients weighing above 100 kg, the DP maximal dosage (i.e., 1280 mg/day PPQ and 160 mg/day DHA indicated for body weight 75 to 100 kg) was administered. This corresponded to a cumulative dose of 4.6 mg/kg DHA and 37 mg/kg PPQ in the present patient, which is far below the WHO recommended ranges (6–20 mg/kg DHA and 48–78 mg/kg PPQ) [22]. Indeed, WHO recommended the use of a PPQ dose of 1600 mg/day for body weigh above 80 kg [22]. In the literature, concerns have been raised over potential DP under-dosing in young children [28], and the 2013 Worldwide Antimalarial Resistance Network (WWARN) has explored the relationship between weight-adjusted DP dosage (mg/kg) and therapeutic efficacy in a meta-analysis [23]. Data of 7072 patients from prospective studies investigating DP efficiency were pooled. Those without molecular evidence for failure or with follow-up less than 28 days were excluded. The primary endpoint was the PCR-adjusted risk of P. falciparum recrudescence at the end of the study follow-up. Secondary endpoints included new P. falciparum infections, parasitological clearance rates, and gametocyte carriage. No patient was heavier than 100 kg. Twenty-eight patients weighed from 75 to 100 kg, and the median cumulative dose in this group was 37.4 mg/kg (32.8–48.3) for PPQ and 4.7 mg/kg (4.1–6.4) for DHA; these were close to the rates in the present patient and corresponded to 71.4 % (20/28) of patients below the WHO recommended dose [22]. The WWARN DP Study Group clearly showed that a total PPQ dose below 40 mg/kg predicted risk of recrudescence failure [23]. Additionally, in this previous study, six factors were reported to be associated with the risk of malaria recrudescence with PPQ [23]. In the present case, two of these six factors were found: the PPQ total dose and the body weight.

Thus, there is much evidence that DP treatment outcome depends on the PPQ plasma concentration. In DP, the artemisinin derivative DHA is responsible for a rapid initial reduction in parasitemia by targeting the parasitized erythrocytes, while PPQ is the partner drug that has a much longer elimination half-life to complete long-term parasite clearance [29]. Moreover, PPQ is of a lipophilic nature with a large volume of distribution [27]. For all of these reasons, it can assume that DP treatment failure in the present case was more related to PPQ under-dosing.

Interestingly, similar overweight-related failures have been documented with atovaquone–proguanil [30, 31]. In a series of 347 P. falciparum—infected travellers, atovaquone–proguanil therapeutic failures were overrepresented among patients weighing more than 100 kg [3/12 in the group (100–115 kg) receiving a standard atovaquone–proguanil regimen versus 2/335 in patients <100 kg] [32]. These data emphasize the lack of an alternative validated malaria treatment in overweight patients.

However, clinical failure by resistance cannot be excluded. Multidrug resistance to DP is currently emerging in Cambodia, where recrudescent infections have increased from 15.4 % in 2011–2013 to 39 % in 2012–2014; 84 % of treated patients presented with parasite clearance half-lives longer than 5 h in 2012–2104, and 57 % were still parasitaemic at 72 h [33, 34]. All the C580Y k13 mutant parasites collected from recrudescent patients after a 3-day DP treatment were resistant in vitro by PPQ survival assay [35]. DHA resistance is a real threat in African countries [4]. However, no mutation in the k13 gene was found in the present patient’s isolate. However, clinical failure by resistance cannot be excluded even if no mutations were identified in k13. Artemisinin failures seem not to be associated with k13 polymorphisms in Africa [15, 36]. However, in the absence of in vitro phenotyping (ring-stage survival assay) as well as data on parasite clearance rate, there was no evidence of resistance to artemisinin.

Decreased susceptibility or parasite resistance to artemisinin partner drugs has been observed in Africa, where single nucleotide polymorphisms in pfcrt and pfmdr1 have been identified in recurrent infections with ACT partners like lumefantrine, mefloquine or amodiaquine [37–41]. However, there is no data currently supporting the involvement of the pfcrt or pfmdr1 genes in PPQ or DHA resistance [17, 42]. The isolate contained two copies of the pftetQ gene, suggesting that the parasites were resistant to doxycycline [18–20]. This result may explain why, even as he took doxycycline as prophylaxis after his treatment by dihydroartemisinin–piperaquine, the patient had recurrent malaria.

Conclusion

Sub-optimal dosing of either component of ACT can result in incomplete elimination of the parasite biomass and subsequent recrudescence, both of which are important driving forces for the selection of parasites with reduced drug susceptibility. ACT regimens should absolutely be deployed using optimal dosing strategies to maximize the likelihood of rapid clinical and parasitological cure, minimize transmission, and retard the onset and spread of resistances. Overweight patients are at particular risk of under-dosing. In some military units, the soldiers often weigh more than 90–100 kg. Should physicians automatically exclude the use of dihydroartemisinin–piperaquine as an anti-malarial treatment in these patients in the absence of data in patients weighing above 100 kg? Until studies assessing how to adapt treatment are performed (increase the duration or the daily dose or repeat a course later), patients weighing more than 100 kg should be aware of the higher risk of therapeutic failure and should be closely monitored at least until day 28, and beyond if possible. This therapeutic failure with DP by under-dosing highlighted the importance of appropriate dosing guidelines and the need of research data (efficacy, pharmacokinetics and pharmacodynamics) in over-weight patient group.

Abbreviations

ACT:

artemisinin-based combination therapy

DHA:

dihydroartemisinin

DP:

dihydroartemisinin–piperaquine

PCR:

polymerase chain reaction

Pfcrt :

Plasmodium falciparum chloroquine resistance transporter

Pfmdt :

Plasmodium falciparum metabolite drug transporter

Pfmdr1 :

Plasmodium falciparum multidrug resistance

PPQ:

piperaquine

QBC:

quantitative buffy coat

RDT:

rapid diagnostic test

WWARN:

Worldwide Antimalarial Resistance Network

WHO:

World Health Organization

References

  1. Bhattarai A, Ali AS, Kachur SP, MÃ¥rtensson A, Abbas AK, Khatib R, et al. Impact of artemisinin-based combination therapy and insecticide-treated nets on malaria burden in Zanzibar. PLoS Med. 2007;4:309.

    Article  Google Scholar 

  2. WHO. World malaria report 2015. Geneva, World Health Organization. http://apps.who.int/iris/bitstream/10665/200018/1/9789241565158_eng.pdf.

  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. 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  Google Scholar 

  5. 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 

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

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  8. 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  PubMed  Google Scholar 

  9. Fairhurst RM. Understanding artemisinin-resistant malaria: what a difference a year makes. Curr Opin Infect Dis. 2015;28:417–25.

    Article  CAS  PubMed  Google Scholar 

  10. 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.

    PubMed  Google Scholar 

  11. 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  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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  Google Scholar 

  14. Boussaroque A, Fall B, 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. 2016;60:624–7.

    Article  CAS  Google Scholar 

  15. Ménard D, Khim N, Beghain J, Adegnika AA, Kremsner PG, Ramherter M, et al. A worldwide map of Plasmodium falciparum artemisinin resistance. N Engl J Med. 2016;74:2453–64.

    Article  Google Scholar 

  16. 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  Google Scholar 

  17. Briolant S, Henry M, Oeuvray C, Amalvict R, Baret E, Didillon E, et al. Absence of association between piperaquine 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 

  18. Briolant S, Wurtz N, Zettor A, Rogier C, Pradines B. Susceptibility of Plasmodium falciparum isolates to doxycycline is associated with pftetQ sequence polymorphisms and pftetQ and pfmdt copy numbers. J Infect Dis. 2010;201:153–9.

    Article  CAS  PubMed  Google Scholar 

  19. Madamet M, Gaillard T, Velut G, Ficko C, Houzé P, Bylicki C, et al. Malaria prophylaxis failure with doxycycline, Central African Republic, 2014. Emerg Infect Dis. 2015;8:1485–6.

    Article  Google Scholar 

  20. Gaillard T, Briolant S, Houzé S, Baragatti M, Wurtz N, Hubert V, et al. PftetQ and pfmdt copy numbers as predictive molecular markers of decreased ex vivo doxycycline susceptibility in imported Plasmodium falciparum malaria. Malar J. 2013;12:414.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Nosten F, White NJ. Artemisinin-based combination treatment of falciparum malaria. Am J Trop Med Hyg. 2007;77(6 Suppl):191–2.

    Google Scholar 

  22. WHO. Guidelines for the treatment of malaria. Geneva, World Health Organization. http://apps.who.int/iris/bitstream/10665/162441/1/9789241549127_eng.pdf?ua=1&ua=1.

  23. 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;10:1001564.

    Article  Google Scholar 

  24. Khaireh BA, Briolant S, Pascual A, Mokrane M, Machault V, Travaillé C, et al. Plasmodium vivax and Plasmodium falciparum infections in the Republic of Djibouti: evaluation of their prevalence and potential determinants. Malar J. 2012;11:395.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Khaireh BA, Assefa A, Guessod HH, Basco LK, Khairh MA, Pascual A, et al. Population genetics analysis during the elimination process of Plasmodium falciparum in Djibouti. Malar J. 2013;12:201.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Faulde MK, Rueda LM, Khaireh BA. First record of the Asian malaria vector Anopheles stephensi and its possible role in the resurgence of malaria in Djibouti, Horn of Africa. Acta Trop. 2014;139:39–43.

    Article  PubMed  Google Scholar 

  27. Eurartesim, INN-piperaquine & INN dihydroartemisinin—WC500118113.pdf. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/001199/WC500118113.pdf.

  28. Zongo I, Somé FA, Somda SAM, Parikh S, Rouamba N, Rosenthal PJ, et al. Efficacy and day 7 plasma piperaquine concentrations in African children treated for uncomplicated malaria with dihydroartemisinin–piperaquine. PLoS ONE. 2014;9:103200.

    Article  Google Scholar 

  29. Price RN, Hasugian AR, Ratcliff A, Siswantoro H, Purba HLE, Kenangalem E, et al. Clinical and pharmacological determinants of the therapeutic response to dihydroartemisinin–piperaquine for drug-resistant malaria. Antimicrob Agents Chemother. 2007;51:4090–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cordel H, Cailhol J, Matheron S, Bloch M, Godineau N, Consigny P-H, et al. Atovaquone–proguanil in the treatment of imported uncomplicated Plasmodium falciparum malaria: a prospective observational study of 553 cases. Malar J. 2013;12:399.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Durand R, Prendki V, Cailhol J, Hubert V, Ralaimazava P, Massias L, et al. Plasmodium falciparum malaria and atovaquone–proguanil treatment failure. Emerg Infect Dis. 2008;14:320–2.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Musset L, Bouchaud O, Matheron S, Massias L, Le Bras J. Clinical atovaquone–proguanil resistance of Plasmodium falciparum associated with cytochrome b codon 268 mutations. Microbes Infect. 2006;8:2599–604.

    Article  CAS  PubMed  Google Scholar 

  33. 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 

  34. 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 

  35. 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 assay: retrospective and prospective investigations. BMC Med. 2015;13:305.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Conrad MD, Bigira V, Kapisi J, Muhindo M, Kamya MR, Havlir DV, et al. Polymorphisms in K13 and falcipain-2 associated with artemisinin resistance are not prevalent in Plasmodium falciparum isolated from Ugandan children. PLoS ONE. 2014;9:105690.

    Article  Google Scholar 

  37. Mwai L, Kiara SM, Abdirahman A, Pole L, Rippert A, Diriye A, et al. In vitro activities of piperaquine, lumefantrine, and dihydroartemisinin in Kenyan Plasmodium falciparum isolates and polymorphisms in pfcrt and pfmdr1. Antimicrob Agents Chemother. 2009;53:5069–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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 

  39. Dahlström S, Aubouy A, Maïga-Ascofaré O, Faucher J-F, Wakpo A, Ezinmègnon S, et al. Plasmodium falciparum polymorphisms associated with ex vivo drug susceptibility and clinical effectiveness of artemisinin-based combination therapies in Benin. Antimicrob Agents Chemother. 2014;58:1–10.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Sisowath C, Stromberg J, Martensson A, Msellem M, Obondo C, Björkman A, et al. In vivo selection of Plasmodium falciparum·pfmdr1 86 N coding alleles by artemether-lumefantrine (Coartem). J Infect Dis. 2005;191:1014–7.

    Article  CAS  PubMed  Google Scholar 

  41. Dokomajilar C, Nsobya SL, Greenhouse B, Rosenthal PJ, Dorsey G. Selection of Plasmodium falciparum pfmdr1 alleles following therapy with artemether–lumefantrine in an area of Uganda where malaria is highly endemic. Antimicrob Agents Chemother. 2006;50:1893–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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  Google Scholar 

Download references

Authors’ contributions

JBR, SV and NP carried out the diagnostic tests, monitored the patient, collected clinical and epidemiological data. BP and MM carried out the molecular genetic study. JBR, BP, SV, FS and EJ wrote the first manuscript. All authors have read and approved the final manuscript.

Acknowledgements

The authors thank Nicolas Benoit from the Equipe Résidente de Recherche en Infectiologie Tropicale, Institut de Recherche Biomédical des Armées for his technical support.

Competing interests

The authors have declared that they have no competing interests.

Ethics approval and consent to participate

According to the French legislation, bio-banking and secondary use for scientific purposes of human clinical samples used for malaria diagnostics are possible as long as the corresponding patients are informed and have not indicated any objections. This requirement was fulfilled here because oral information was given to the patient, and no immediate or delayed patient opposition was reported by the hospital clinicians. Moreover, samples received at the French Malaria Reference Center were registered and declared for research purposes as a bio-bank for the French National Institute of Health Survey. No institutional review board approval is required according to French legislation (article L. 1111–7 du Code de la Santé Publique, article L. 1211–2 du Code de Santé Publique, articles 39 et suivants de la loi 78–17 du 6 janvier 1978 modifiée en 2004 relative à l’informatique, aux fichiers, et aux libertés).

Funding

This study was supported by the Institut national de veille sanitaire (grant CNR Paludisme).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Bruno Pradines or Emilie Javelle.

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

Roseau, J.B., Pradines, B., Paleiron, N. et al. Failure of dihydroartemisinin plus piperaquine treatment of falciparum malaria by under-dosing in an overweight patient. Malar J 15, 479 (2016). https://doi.org/10.1186/s12936-016-1535-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12936-016-1535-8

Keywords