Clinical Efficacy of Artemether-Lumefantrine Artesunate-Amodiaquine of Children from Three Chadian Provinces with Acute of Uncomplicated Falciparum malariae from 6 Months to 5 Years

Abstract

Introduction: Artemisinin-based combination therapies are the first-line antimalarial drugs used to treat uncomplicated Plasmodium falciparum malaria in many endemic countries worldwide. In Chad, since the adoption of artemisinin-based combination therapy (ACTs) in its first-line treatment policy for uncomplicated malaria in 2005, Artemether-Lumefantrine and Artesunate-Amodiaquine have been used in many hospitals and health centers. The main objective of this study was to provide the baseline data of Artemether-Lumefantrine and Artesunate-Amodiaquine efficacy in three regions where many people suffered from malaria disease. Material and Methods: The baseline efficacy of two combination therapies was evaluated between January and April 2020 in Mosoro, Mondou and Dourbali Provinces in Chad. A two-arm single cohort study was conducted to assess the clinical efficacy of artesunate-amodiaquine and artemether-lumefantrine for the treatment of 1113 children aged from 6 to 59 months with uncomplicated Falciparum malariae diagnosed by thick blood smear examination, using the World Health Organization validated protocol. Results: On day 3, all patients in both groups had cleared parasitemia, after treatment, the patients presented a higher hemoglobin level in both groups artemether-lumefantrine (10.97 ± 1.39) and artesunate-amodiaquine (11.87 ± 1.81), respectively. On day 28, all patients had adequate clinical and parasitological responses with 99.82% of artesunate-amodiaquine and 99.10% of artemether-lumefantrine. Overall, both drugs were well tolerated at the clinical and biological level, no late parasitological failures have been recorded in artemether-lumefantrine and artesunate-amodiaquine groups, also both forms of Artemisinin-based combination therapy were still effective and safe in the treatment of uncomplicated P. falciparum malaria in Chad.

Share and Cite:

Adam, I. , Jacques, N. , Alio, H. , Patrick, A. , Lawane, A. and Bolti, M. (2022) Clinical Efficacy of Artemether-Lumefantrine Artesunate-Amodiaquine of Children from Three Chadian Provinces with Acute of Uncomplicated Falciparum malariae from 6 Months to 5 Years. Advances in Infectious Diseases, 12, 788-798. doi: 10.4236/aid.2022.124056.

1. Introduction

Malaria is widespread in tropical areas worldwide and causes high morbidity. The emergence of parasite resistance to antimalarial medicines constitutes a challenge impeding countries’ abilities to advance towards elimination; these include lack of sustainable funding, risks posed by conflict in malaria endemic zones, and mosquito resistance to insecticides [1]. It is estimated that there were 229 million clinical cases of malaria and around 409,000 deaths in 2019, of which nearly 67% were children aged from 0 to 59 months [2]. Almost 85% of malaria deaths worldwide in 2018 were concentrated in 20 countries in Africa region and India. Nigeria alone accounted for nearly 24% of these deaths, followed by the Democratic Republic of Congo (11%), the United Republic of Tanzania (5%), as well as Angola, Mozambique and Niger (4%) each. It is estimated that nearly 24 million children in sub-Saharan Africa suffered from P. falciparum malaria infections in 2018. In Chad, malaria causes a serious health problem that represents 42% of first consultation cases in different health centers, 39% of hospitalization and 32% of death cases [3].

To reduce malaria-related morbidity and mortality, Artemisinin Combination Therapies (ACTs) have been integrated into the recent success of global malaria control, and protecting their efficacy for the treatment of P. falciparum malaria is a global health priority. In Chad, antimalarial treatment for uncomplicated falciparum malaria is based on ACTs in accordance with the World Health Organization recommendation for regions with resistance to antimalarials. The majority of the studies analyzing efficacy of this antimalarial regimen are conducted in high transmission regions, and selection of mutations that could possibly confer P. falciparum resistance to ACT has been reported both in vivo and ex-vivo by [4] [5]. The advice for government health programs is to closely monitor parasite genotypic, phenotypic and clinical dynamics of P. falciparum infections in response to ACTs, despite its continued efficacy.

Due to the high levels of clinical resistance to chloroquine, amodiaquine, and sulphadoxine-pyrimethamine, the Chadian Ministry of Public Health changed the national antimalarial drug policy in 2005. Two Artemisinin-based Combination Therapies (ACTs) were adopted: Artesunate-Amodiaquine and Artemether-Lumefantrine for the first-line and second-line treatment of uncomplicated malaria. The criteria used to select subjects for an antimalarial drug efficacy trial define the study population and can impact treatment outcomes and study results. For example, in endemic areas, increasing age is associated with greater antimalarial immunity and improved therapeutic response [4] [6] and a higher parasite burden at the time of treatment may be associated with less favourable outcomes [7]. The WHO recommended that patient selection criteria are restrictive, and do not cover the full range of patients with uncomplicated malaria. In practice, the criteria used to enroll patients into antimalarial drug efficacy studies often vary with respect to age, parasite density, and baseline [8].

2. Material and Method

2.1. Study Area

The present study took place from January to April 2020 in three provinces of Chad; the main objective of this study was to provide the baseline data of Artemether-Lumefantrine (AL) and Artesunate-Amodiaquine (ASAQ) efficacy in three regions where much people suffered from malaria disease. Febrile children aged between 6 months and 5 years were enrolled after a written informed consent was obtained from the parents or the legal guardian.

2.2. Study Procedures

Patients with malaria-like symptoms were received at selected hospitals. Meticulous clinical examinations of the patients and laboratory investigations were conducted immediately after inclusion. Patients who met baseline inclusion criteria were randomly assigned to one of the two treatment groups. Drug administration was supervised by a member of the research team and patients were kept under observation. After the initial dose, patients had a follow-up within 28 days. Each patient was scheduled for follow-up examinations on day 0, 1, 2, 3, 7, 14, 21, 28, any other time the participant felt unwell during the study period will be treated. During the visit day, the following information was collected: 1) clinical data, 2) biological data (diagnosis of malaria and determination of parasitemia), 3) any details about drug adverse events. In case of treatment failure, the placement therapy was offered according to the national treatment guidelines.

2.3. Procedures during Inclusion and Follow-Up

After obtaining the informed written consent, a complete medical history (symptoms, current medications and previous use of anti-malarial drugs), biographic and contact details were noted. A complete physical examination was performed and a case record form was filled in for each patient. Body weight was recorded at day 0; temperature was recorded using a thermometer with a precision of 0.1˚C at baseline and on follow-up days, and additionally measured as clinically indicated. On each follow-up, clinical signs and symptoms were registered, including symptoms such as fever, headache and abdominal pain; blood have been drowned from vain and conserved put it in EDTA tube for hemoglobin test, blood smears were also performed to detect malaria parasites. During the study period, all male and female patients aged 6 to 59 months inclusive, with fever (axillary temperature ≥ 37.5˚C) or history of fever in the last 24 h, visiting the peripheral health centers of Massakory, Dourbali or Mondo health districts. Children weighing less than 5 kg or more, with a confirmed P. falciparum (parasitaemia ≥ 4000/μL to 200,000/μL) mono-infection, hemoglobin level above 5.0 g/dl, and agreed to participate whenever applicable by giving their assent and if their parents or guardian provided written informed consent. Patients were not included if they were not willing to participate or had participated to any drug trial within the last 30 days, or had known hypersensitivity to the study drug, or were severely malnourished (defined as weight for height < 70% of the median NCHS/WHO reference), or had severe malaria.

2.4. Laboratory Analysis

Microscopic blood examination and quality control. Thick and thin blood films were prepared and stained with Giemsa for screening and subsequent species diagnosis and parasitemia calculation on days 0, 1, 2, 3, 7, 14, 21 and 28 or if reassessment was required. Parasite counts were done on thick films and the number of parasites per 200 white blood cells (WBCs) were counted by light microscopy. Parasite density, expressed as the number of asexual parasites per μL of blood, was calculated by dividing the number of asexual parasites by the number of WBCs counted, and then multiplying it by an assumed WBC density of typically 8000 per μL. A blood-slide sample was considered negative when examination of 200 fields containing at least ten WBCs per field revealed no asexual parasites. The presence of gametocytes on the day the patient was enrolled or on the day of follow up was also recorded.

2.5. Hemoglobin and Antimalarial Drug Blood Concentration

Hemoglobin was determined on days 0 and 28 in 10 μL of capillary blood sample with the HemoCue blood Hemoglobin system. Specimens were be labeled anonymously (study number, day of follow-up, date).

2.6. Follow-Up and Loss

Follow-up visits and procedures were scheduled per protocol on days 1, 2, 3, 7, 14, 21, and 28. Patients were allowed to return to hospital at any time if they had fever or any general danger signs as described under exclusion criteria. The study team made home visits as follow-ups for study participants that were late for their scheduled visits. Patients who failed to return on days 1 or 2 and missed one dose of the treatment or enrolled patients who could not attend scheduled visits were considered lost to follow-up and excluded from study.

2.7. Ethical Considerations

The study protocol was reviewed and approved by the National Malaria Control Program (NMCP) in Chad. Written consent was obtained from the children’s parents before entering the study. The study was conducted according to the WHO good clinical practices guidelines and according to the Chadian ministry of health regulations.

2.8. Statistical Analysis

All data were recorded using IBM SPSS Statistics, version 25, comparisons of different parameters in both arms were done using, Fisher’s exact test. The level of significance for statistical tests was set at 0.05.

3. Results

3.1. Baseline Characteristics of Participants

During the study period, 1113 patients were screened and treated with either AL or ASAQ for twenty eight days. About 50.31% of the participants treated with AL and 51.21% treated with ASAQ. The mean of parasitemia was 11720.24 with the interval of 2000.200000 and 17834.84 with the interval of 2000.200000 respectively in AL and ASAQ groups. Other characteristics were described in (Table 1).

3.2. Efficacy Results

On day 0, parasitemia was identified in 28 patients in AL groups, with a mean of parasite load of 11,720 parasites/μl and 27 patients in ASAQ groups with a mean of parasite load of 9534 parasites/μl; eleven patients were positive on day 1, with a mean of 3063.38 parasites in AL groups and 144 patients with a mean of 1782.02 in ASAQ groups (Table 2). On day 3, all patients in both two groups had cleared parasitemia. An adequate clinical and parasitological response, defined by a negative thick blood smear on day 28, was found in ASAQ and AL treatments were 99.82% and 99.10% on day 28 respectively, and no late parasitological failures were detected in two groups of treatments and no serious adverse events were registered, on other hand 2 (0.36) and 1(0.17) of patients had en earlier treatment failure in both groups AL and ASAQ (Table 2).

The study showed that Patients presented a lower hemoglobin level in AL groups (7.64 ± 1.89) and (8.11 ± 1.77) in ASAQ groups in day 0, (8.88 ± 1.91) and 9.38 ± 1.11 in day 7, but in day 28 after treatment the patients presented a higher hemoglobin level in both groups AL (10.97 ± 1.39) and ASAQ (11.87 ± 1.81) respectively (Table 3).

4. Discussion

Clinical response to antimalarial therapy involves a complex interaction between the parasites, drugs and the host response; In vivo tests are considered the gold standard method of measuring antimalarial drug resistance [9]. Host immunity

Table 1. Demographic and clinical characteristics of patients.

Legend: AL = Arthemether lumefantrine; ASAQ = Artesunate-Amodiaquine; n = Number of population examinated; (%) = Parcentage.

Table 2. Treatment outcomes at 28 days in 1113 patients.

Legend: ETF = Earlier treatment failure; ECF = Earlier clinical failure, LPF: Late parasitological failure; ACPR = Adequate clinical and parasitological responses; n = Number of population ex-aminated; (%) = Parcentage

Table 3. Patients distribution according to hemoglobin rate variations.

Legend: AL = Arthemether lumefantrine; ASAQ = Artesunate-Amodiaquine,; P = p value; D0 = day 0; D7 = seven days; D14 = fourteen days; D28 = twenty eight days.

plays a critical role in the clearance of malarial parasites. For example, the ability of monocytes to kill intracellular parasites is a function of antibody-dependent mechanisms [10]. The production of specific antibodies has been associated with protection against clinical attacks of malaria. Acquired antimalarial immunity develops following repeated exposure to infecting parasites [11]. The objective of this study was to provide the baseline data of Artemether-Lumefantrine and Artesunate-Amodiaquine efficacy in three regions where much people suffered from malaria disease. The AL and ASAQ appeared to be a better treatment option on the basis of non-PCR-corrected responses, based on the lower percentage of recurrent parasitaemia observed. The results showed a high cure rate for both regimens after a standard 28-day follow-up with an ACPR rate adjusted by 99.82% in ASAQ and 99.10% in AL. This is consistent with efficacy results reported from several studies of [12], in Cameroon and [13] in Ghana, the fact that there was more reinfection than recrudescence shows that malaria transmission is high in all three study sites, indeed in Chad, malaria is perennial with a peak during rainy seasons in three sites. Our study found that the cure treatment rate of ASAQ was superior to AL during 28 days of observation, same results reported by [14] in Bourkina faso who noted the higher rate of ACPR in ASAQ group. Also this result is different from the result of [15] [16] [17] who compared ASAQ with AL over a follow-up duration of 28 days and demonstrated a lower reinfection rate for AL, and found that AL was superior to ASAQ in preventing new infections. In the present study on day 3, all patients in two groups had cleared parasitemia with an adequate clinical and parasitological response defined by a negative thick blood smear on day 28, no late parasitological failures were detected in two groups of treatments and no serious adverse events were registered. This finding is consistent with the study of [18] [19] in which no serious adverse effects were reported, and that could be explained by the higher efficacy of combination treatment for decreasing the gametocyte rate during the treatment with ACTs and therefore leads to a significant reduction in the spread of resistance. The clinical tolerance was good with minor adverse events in both treatment groups confirming previous studies such as [12]. In this study, a progressive haematological improvement was observed from baseline to day 28 between the two treatment groups; however, patients from ASAQ group had relatively higher Hb at enrolment which was maintained throughout the follow-up. Higher mean Hb in ASAQ than AL might be attributed to differences in nutritional status and other conditions associated with anaemia such as concurrent infections [20]. It could also be due to differences in age, since patients enrolled in ASAQ had significantly higher mean age compared to AL (4.33 vs 3.77 years for ASAQ and AL, respectively). Improvements in Hb during follow-up could suggest that malaria might be a major contributing factor to the low haemoglobin levels and anaemia at recruitment as reported in other studies done in Tanzania by [21], and elsewhere in sub-Saharan Africa by [22] [23]. On the other hand, this result was confirmed by [24] in Ivory Coast who noted in his study that the patients of two groups of treatment had a higher average of hemoglobin level on day 28 after treatment, and that could be explained by the efficacy of the combination treatment in two groups. This study was limited by the lack of PCR correction to distinguish true recrudescence with new infection and the limited follow up period of 28 days for these ACTs; nonetheless, the results obtained are of great relevance since it described the real time behavior of the drugs in term of efficacy and tolerability and the late occurrence of the failures were in line with clinical trial previously reports; the time limit of follow up to 28 days may have contributed to underestimate the failures but has the advantage to reduce the risk of new infection. More generally, these ACTs (AL and ASAQ) retained a certain efficacy in the treatment of uncomplicated malaria; however, some studies such as [25] [26] reported an efficacy level of AL during 28 days follow up below the threshold set for the ACTs adopted; for all these studies, PCR corrected outcomes were above 90% success. More than scientific interest or intrinsic efficacy of the drugs, it is questionable to whether ACTs real life efficacy should be based on the PCR corrected results or the uncorrected one or a combination of the two approaches. It may be time to initiate the discussion on current first line drugs replacement and investigate to which extend retrieved drugs (non ACT drugs like chloroquine) could be given a new life for the treatment of malaria [27].

5. Conclusion

During this study, ASAQ and AL were effective and well-tolerated in the treatment of uncomplicated falciparum malaria. Therefore, this result supports the continued use of these ACTs in the management of malaria with added advantage provided in public health facilities in slowing the spread of malaria drug resistance and of global reduction or elimination of malaria in Chad.

Acknowledgements

We wish to thank all the children and their families for their excellent cooperation and we are very grateful to the local health authorities in Massakory, Mondo and Dourbali regions and the National Malaria Control Program (NMCP) in Chad. Thanks are to the team of laboratory of parasitology unit for their excellent cooperation and technical assistance.

Author’s Contribution

All authors contributed to the conduct of this work and to the drafting of the manuscript. All authors have read and approved the final version of the manuscript.

Conflicts of Interest

The authors declare that they have no competing interests.

References

[1] WHO (2018) 2017 World Malaria Report. WHO, Geneva.
[2] WHO (2019) 2019 World Malaria Report. WHO, Geneva.
[3] National Malaria Control Program Chad NMCP (2020).
[4] Lobo, E., de Sousa, B., Rosa, S., Figueiredo, P., Lobo, L., Pateira, S., Fernandes, N. and Nogueira, F. (2014) Prevalence of pfmdr1 Alleles Associated with Artemether-Lumefantrine Tolerance/Resistance in Maputo before and after the Implementation of Artemisinin-Based Combination Therapy. Malaria Journal, 13, Article No. 300.
https://doi.org/10.1186/1475-2875-13-300
[5] Dahlström, S., Aubouy, A., Maïga-Ascofaré, O., Faucher, J.F., Wakpo, A., Ezinmègnon, S., Massougbodji, A., Houzé, P., Kendjo, E., Deloron, P., Le Bras, J. and Houzé, S. (2014) Plasmodium falciparum Polymorphisms Associated with ex Vivo Drug Susceptibility and Clinical Effectiveness of Artemisinin-Based Combination Therapies in Benin. Antimicrobial Agents and Chemotherapy, 58, 1-10.
https://doi.org/10.1128/AAC.01790-12
[6] Kavishe, R.A., Paulo, P., Kaaya, R.D., Kalinga, A., van Zwetselaar, M., Chilongola, J. and Alifrangis, M. (2014) Surveillance of Artemether-Lumefantrine Associated Plasmodium falciparum Multidrug Resistance Protein-1 Gene Polymorphisms in Tanzania. Malaria Journal, 13, Article No. 264.
https://doi.org/10.1186/1475-2875-13-264
[7] Achieng, A.O., Muiruri, P., Ingasia, L.A., Opot, B.H., Juma, D.W., Yeda, R. and Kamau, E. (2015) Temporal Trends in Prevalence of Plasmodium falciparum Molecular Markers Selected for by Artemether-Lumefantrine Treatment in Pre-ACT and Post-ACT Parasites in Western Kenya. International Journal for Parasitology: Drugs and Drug Resistance, 5, 92-99.
https://doi.org/10.1016/j.ijpddr.2015.05.005
[8] Vreden, S.G., Jitan, J.K., Bansie, R.D. and Adhin, M.R. (2013) Evidence of an Increased Incidence of Day 3 Parasitaemia in Suriname: An Indicator of the Emerging Resistance of Plasmodium falciparum to Artemether. Memórias do Instituto Oswaldo Cruz, 108, 968-973.
https://doi.org/10.1590/0074-0276130167
[9] Egan, A.F., Morris, J., Barnish, G., Allen, S., Greenwood, B.M., Kaslow, D.C., Holder, A.A. and Riley, E.M. (1996) Clinical Immunity to Plasmodium falciparum Malaria Is Associated with Serum Antibodies to the 19-kDa C-Terminal Fragment of the Merozoite Surface Antigen, PfMSP-1. Journal of Infectious Diseases, 173, 765-768.
https://doi.org/10.1093/infdis/173.3.765
[10] Bouharoun-Tayoun, H., Oeuvray, C., Lunel, F. and Druilhe, P. (1995) Mechanisms Underlying the Monocyte-Mediated Antibody Dependent Killing of Plasmodium falciparum Asexual Blood Stages. The Journal of Experimental Medicine, 182, 409-418.
https://doi.org/10.1084/jem.182.2.409
[11] Tobón-Castaño, A., Garcés-Murillo, L., Ríos-Orrego, A., Montiel-Ramos, J., De Las Salas, B., Mesa-Echeverry, E. and Niebles-Bolivar, M. (2018) Artemeter-Lumefantrine Therapeutic Efficacy, Safety and Plasma Levels in Patients with Uncomplicated Falciparum Malaria from the Colombian Pacific Region. Infectio, 22, 199-205.
https://doi.org/10.22354/in.v22i4.738
[12] Faye, B., Kuété, T., Kiki-Barro, C.P., Tine, R.C., Nkoa, T., Ndiaye, J.L.A. and Moussa, K. (2012) Multicentre Study Evaluating the Non-Inferiority of the New Paediatric Formulation of Artesunate/Amodiaquine versus Artemether/Lumefantrine for the Management of Uncomplicated Plasmodium falciparum Malaria in Children in Cameroon, Ivory Coast and Senegal. Malaria Journal, 11, Article No. 433.
https://doi.org/10.1186/1475-2875-11-433
[13] Kobbe, R., Klein, P., Adjei, S., Amemasor, S., Thompson, W.N., Heidemann, H. and May, J. (2008) A Randomized Trial on Effectiveness of Artemether-Lumefantrine versus Artesunate plus Amodiaquine for Unsupervised Treatment of Uncomplicated Plasmodium falciparum Malaria in Ghanaian Children. Malaria Journal, 7, Article No. 261.
https://doi.org/10.1186/1475-2875-7-261
[14] Zongo, I., Compaoré, Y.D., Nikiéma, F., Zongo, M., Barry, N., Somé, F.A., Kaboré, N., & Ouédraogo, J.B. (2020) Efficacy of Artemether-Lumefantrine and Artesunate-Amodiaquine as First Line Therapy of Uncomplicated Malaria in Burkina Faso, 11 Years after Policy Change. Pan African Medical Journal, 35, Article No. 68.
https://doi.org/10.11604/pamj.2020.35.68.20849
[15] Bukirwa, H., Yeka, A., Kamya, M.R., Talisuna, A., Banek, K., Bakyaita, N. and Staedke, S.G. (2006) Artemisinin Combination Therapies for Treatment of Uncomplicated Malaria in Uganda. PLOS Clinical Trials, 1, e7.
https://doi.org/10.1371/journal.pctr.0010007
[16] Mårtensson, A., Strömberg, J., Sisowath, C., Msellem, M.I., Gil, J.P., Montgomery, S.M. and Björkman, A. (2005) Efficacy of Artesunate plus Amodiaquine versus That of Artemether-Lumefantrine for the Treatment of Uncomplicated Childhood Plasmodium falciparum Malaria in Zanzibar, Tanzania. Clinical Infectious Diseases, 41, 1079-1086.
https://doi.org/10.1086/444460
[17] Schramm, B., Valeh, P., Baudin, E., Mazinda, C.S., Smith, R., Pinoges, L. and Guérin, P.J. (2013) Efficacy of Artesunate-Amodiaquine and Artemether-Lumefantrine Fixed-Dose Combinations for the Treatment of Uncomplicated Plasmodium falciparum Malaria among Children Aged Six to 59 Months in Nimba County, Liberia: An Open-Label Randomized Non-Inferiority Trial. Malaria Journal, 12, Article No. 251.
https://doi.org/10.1186/1475-2875-12-251
[18] Carrasquilla, G., Barón, C., Monsell, E.M., Cousin, M., Walter, V., Lefèvre, G. and Fisher, L.M. (2012) Randomized, Prospective, Three-Arm Study to Confirm the Auditory Safety and Efficacy of Artemether-Lumefantrine in Colombian Patients with Uncomplicated Plasmodium falciparum Malaria. The American Journal of Tropical Medicine and Hygiene, 86, 75-83.
https://doi.org/10.4269/ajtmh.2012.11-0192
[19] Nambozi, M., Kabuya, J.B.B., Hachizovu, S., Mwakazanga, D., Mulenga, J., Kasongo, W. and D’Alessandro, U. (2017) Artemisinin-Based Combination Therapy in Pregnant Women in Zambia: Efficacy, Safety and Risk of Recurrent Malaria. Malaria Journal, 16, Article No. 199.
https://doi.org/10.1186/s12936-017-1851-7
[20] Shayo, A., Buza, J. and Ishengoma, D.S. (2015) Monitoring of Efficacy and Safety of Artemisinin-Based Anti-Malarials for Treatment of Uncomplicated Malaria: A Review of Evidence of Implementation of Anti-Malarial Therapeutic Efficacy Trials in Tanzania. Malaria Journal, 14, Article No. 135.
https://doi.org/10.1186/s12936-015-0649-8
[21] Bousema, T., Okell, L., Shekalaghe, S., Griffin, J.T., Omar, S., Sawa, P., et al. (2010) Revisiting the Circulation Time of Plasmodium falciparum Gametocytes: Molecular Detection Methods to Estimate the Duration of Gametocyte Carriage and the Effect of Gametocytocidal Drugs. Malaria Journal, 9, Article No. 136.
https://doi.org/10.1186/1475-2875-9-136
[22] Ndiaye, J.L., Faye, B., Gueye, A., Tine, R., Ndiaye, D. and Tchania, C. (2011) Repeated Treatment of Recurrent Uncomplicated Plasmodium falciparum Malaria in Senegal with Fixed-Dose Artesunate plus Amodiaquine versus Fixed-Dose Artemether plus Lumefantrine: A Randomized, Open-Label Trial. Malaria Journal, 10, Article No. 237.
https://doi.org/10.1186/1475-2875-10-237
[23] Ogouyemi-Hounto, A., Azandossessi, C., Lawani, S., Damien, G., de Tove, Y.S. and Remoue, F. (2016) Therapeutic Efficacy of Artemether-Lumefantrine for the Treatment of Uncomplicated Falciparum Malaria in Northwest Benin. Malaria Journal, 15, Article No. 37.
https://doi.org/10.1186/s12936-016-1091-2
[24] Yavo, W., Konaté, A., Kassi, F.K., Djohan, V., Angora, E.K., Kiki-Barro, P.C. and Menan, E.I.H. (2015) Efficacité et innocuité de l’artésunate-amodiaquine par rapport à l’artéméther-luméfantrine dans le traitement du paludisme simple à Plasmodium falciparum dans les sites sentinelles à travers la Côte d’Ivoire.
[25] Sondo, P., Derra, K., Diallo-Nakanabo, S., Tarnagda, Z., Zampa, O. and Kazienga, A. (2015) Effectiveness and Safety of Artemether-Lumefantrine versus Artesunate-Amodiaquine for Unsupervised Treatment of Uncomplicated Falciparum Malaria in Patients of All Age Groups in Nanoro, Burkina Faso: A Randomized Open Label Trial. Malaria Journal, 14, Article No. 325.
https://doi.org/10.1186/s12936-015-0843-8
[26] Dama, S., Niangaly, H., Djimde, M., Sagara, I., Guindo, C.O. and Zeguime, A. (2018) A Randomized Trial of Dihydroartemisinin-Piperaquine versus Artemether-Lumefantrine for Treatment of Uncomplicated Plasmodium falciparum Malaria in Mali. Malaria Journal, 17, Article No. 347.
https://doi.org/10.1186/s12936-018-2496-x
[27] Dagnogo, O., Ako, A.B., Ouattara, L., Dago, N.D., Coulibaly, D.N. and Touré, A.O. (2018) Towards a Re-Emergence of Chloroquine Sensitivity in Côte d’Ivoire. Malaria Journal, 17, Article No. 413.
https://doi.org/10.1186/s12936-018-2551-7

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.