ArticlePDF Available

Population Pharmacokinetics of Intramuscular Artesunate in African Children With Severe Malaria: Implications for a Practical Dosing Regimen

Authors:

Abstract and Figures

Parenteral artesunate (ARS) is the drug of choice for the treatment of severe malaria. Pharmacokinetics data on intramuscular ARS are limited with respect to the main treatment group that carries the highest mortality, namely, critically ill children with severe malaria. A population pharmacokinetic study of ARS and dihydroartemisinin (DHA) was conducted from sparse sampling in 70 Tanzanian children of ages 6 months to 11 years. All the children had been admitted with severe falciparum malaria and were treated with intramuscular ARS (2.4 mg/kg at 0, 12, and 24 h). Venous plasma concentration-time profiles were characterized using nonlinear mixed-effects modeling (NONMEM). A one-compartment disposition model accurately described first-dose population pharmacokinetics of ARS and DHA. Body weight significantly affected clearance and apparent volume of distribution (P < 0.001), resulting in lower ARS and DHA exposure levels in smaller children. An adapted dosing regimen including a practical dosing table per weight band is proposed for young children based on the pharmacokinetic model.Clinical Pharmacology & Therapeutics (2013); advance online publication 20 March 2013. doi:10.1038/clpt.2013.26.
Content may be subject to copyright.
Articles
nature publishing group
Severe falciparum malaria is one of the main causes of death
in African children, who account for >85% of the worldwide
malaria-related mortality.1 In children, the course of the disease
is more rapid than in adults, with most deaths occurring within
the rst 24 h aer admission despite parenteral antimalarial
treatment.2,3
Artesunate (ARS), administered parenterally, is currently
the drug of choice for the treatment of severe malaria in all
age groups and all malaria-endemic settings.4 A large trial
(SEAQUAMAT) carried out in Asia in a study population con-
sisting mostly of adult patients (n = 1,461) with severe falcipa-
rum malaria showed a 35% reduction in mortality with ARS
treatment as compared with quinine.
5
More recently, a larger
trial (AQUAMAT) performed in 5,425 African children with
severe malaria showed a 22.5% lower mortality in children
treated with parenteral ARS as compared with quinine.6
ARS has a broader stage-specicity and more potent para-
siticidal eect than quinine.
7,8
ARS is water-soluble and can be
administered either as an i.v. slow bolus or as an i.m. injection. e
latter route is more practical in the majority of African hospital
and clinic settings with limited facilities. e absorption of i.m.
ARS is rapid and reliable, with peak concentrations occurring
within 1 h.9,10 Aer being injected, ARS is rapidly and almost
completely converted into its active metabolite, dihydroarte-
misinin (DHA).
11
e elimination of ARS is very rapid, and anti-
malarial activity is determined mainly by DHA exposure levels.
DHA has a terminal elimination half-life of ~45 min
10,12,13
and is
~93% plasma protein–bound in patients with falciparum infec-
tion.
14
e current dosing recommendation for ARS in the treat-
ment of severe malaria is 2.4 mg/kg on admission, followed by
the same dose aer 12 h and then a daily dose until the patient is
able to take oral antimalarial therapy reliably. is recommenda-
tion, for the most part, was derived empirically from studies in
adults.15,16
In small children with severe disease, the pharmacokinetic
properties of antimalarials may dier from those reported in
Parenteral artesunate (ARS) is the drug of choice for the treatment of severe malaria. Pharmacokinetics data on
intramuscular ARS are limited with respect to the main treatment group that carries the highest mortality, namely,
critically ill children with severe malaria. A population pharmacokinetic study of ARS and dihydroartemisinin (DHA)
was conducted from sparse sampling in 70 Tanzanian children of ages 6 months to 11 years. All the children had been
admitted with severe falciparum malaria and were treated with intramuscular ARS (2.4 mg/kg at 0, 12, and 24 h). Venous
plasma concentration–time profiles were characterized using nonlinear mixed-effects modeling (NONMEM). A one-
compartment disposition model accurately described first-dose population pharmacokinetics of ARS and DHA. Body
weight significantly affected clearance and apparent volume of distribution (P < 0.001), resulting in lower ARS and DHA
exposure levels in smaller children. An adapted dosing regimen including a practical dosing table per weight band is
proposed for young children based on the pharmacokinetic model.
Received 8 August 2012; accepted 23 January 2013; advance online publication 20 March 2013. doi:10.1038/clpt.2013.26
Clinical Pharmacology & erapeutics
10.1038/clpt.2013.26
Articles
20March2013
93
5
8August2012
23January2013
Population Pharmacokinetics of Intramuscular
Artesunate in African Children With Severe
Malaria: Implications for a Practical Dosing
Regimen
ICE Hendriksen1,2, G Mtove3, A Kent4, S Gesase5, H Reyburn6, MM Lemnge5, N Lindegardh1,2,
NPJ Day1,2, L von Seidlein7, NJ White1,2, AM Dondorp1,2 and J Tarning1,2
1Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; 2Centre for Tropical Medicine, Churchill
Hospital, University of Oxford, Oxford, UK; 3National Institute for Medical Research, Amani Centre, Tanga, Tanzania; 4Joint Malaria Programme, Moshi, United Republic
of Tanzania; 5National Institute for Medical Research, Tanga Medical Research Centre, Tanga, Tanzania; 6London School of Hygiene and Tropical Medicine, London, UK;
7Menzies School of Health Research, Casuarina, Australia. Correspondence: J Tarning ( joel@tropmedres.ac)
Open
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 93 NUMBER 5 | MAY 2013 443
Articles
adults.17–19 Only limited data are available on the pharmacoki-
netic properties of parenteral, rectal, and oral ARS in children
with moderate to severe malaria.
9,13,20–22
e primary aim of
this study was to characterize the population pharmacokinetic
properties of ARS and its active metabolite DHA in the treat-
ment of severe malaria in African children and to determine a
practical dosing regimen.
RESULTS
Clinical details
Seventy patients of ages 7 months to 11 years were included, of
whom 59 (84%) were <5 years of age. During the study, nine of
the children died (case fatality 13%). Of these, one died within
15 min of admission, and two died within the rst 24 h aer
admission. One patient of age 2.5 years had severe neurologic
sequelae 28 days aer discharge, comprising spastic hemiparesis,
blindness, and hearing impairment. At admission this child had
a count of 880,205 parasites/µl and plasma Plasmodium falcipa-
rum histidine-rich protein-2 concentration of 1,875 ng/ml, both
indicating an extremely heavy parasite burden. At the 3-month
follow-up, the neurologic sequelae had resolved completely.
Demographic, clinical, and laboratory characteristics of the
study population are described in Tab l e 1. Severe prostration,
severe acidosis, convulsions, and severe anemia were the most
common severity criteria. Eleven patients (16%) presented
with decompensated or compensated shock. In addition,
three patients (4.3%) had blood culture–proven septicemia
(unspeciated Gram-negative rods, Klebsiella pneumoniae, or
Staphylococcus aureus); none of these presented with shock. HIV
coinfection was detected in 3/70 (4.3%) of the patients. None of
these patients were receiving antiretroviral treatment.
Pretreatment with an oral antimalarial was reported with
respect to 31 patients (6 with quinine, 3 with amodiaquine, 13
with sulfadoxine-pyrimethamine, 8 with artemether-lumefan-
trine, and 1 with amodiaquine followed by artemether-lumefan-
trine). In addition, 12 patients had received pretreatment with
i.m. quinine within the 24 h preceding admission, the median
(range) total dose being 16.1 mg/kg (10.1–53.7 mg/kg).
Each of the patients received an i.m. ARS injection of 2.4 mg/
kg shortly aer admission. Supportive treatments included blood
transfusions and uid resuscitation. Hypoglycemia was corrected
with a 10% dextrose bolus at the time of admission and in those
who developed hypoglycemia aer admission (eight patients).
Peripheral blood asexual parasite counts aer 24 h were nega-
tive in 12/66 (18%) patients (24-h slide was not available for four
patients, three of them as a result of death). e geometric mean
(95% condence interval (CI)) parasite count aer 24 h was 1,128
(537–2,368) parasites/µl in the rest of the study population (n =
54). e overall geometric mean (95% CI) fractional reduction in
parasite counts at 24 h was 96% (94–98%).
Population pharmacokinetic-pharmacodynamic analysis
A total of 274 ARS and DHA postdose samples, randomly distrib-
uted over the rst 12 h of the study, were included in the model. A
one-compartment disposition model for both ARS and DHA was
adequate to describe the observed plasma concentration–time
data (Table 2). All combinations of two-compartment disposi-
tion models displayed signicant model misspecication despite
signicantly lower objective function values. A similar nding
has been reported previously for DHA.
23
Zero-order distribution/
absorption from the injection site(s) to the central compartment
provided the best description of the data, but too few samples
were collected during the absorption phase and the absorption
rate was therefore xed for an accurate estimation. Interindividual
variability could be estimated reliably for ARS and DHA clearance
and ARS volume of distribution, showing correlation between
ARS clearance and volume.
Table 1 Demographic, clinical, and laboratory characteristics of
children admitted with severe malaria
Variable Value
Total number of patients 70
Age (y) (median, range) 2.5 (0.6 to 11)
Weight (kg) 10.8 (9 to 13.5)
Weight-for-age Z-scorea−1.2 (1.0)
Coma (based on GCS/BCS) 19 (27%)
Prostration 46 (66%)
Convulsions 26 (37%)
Shockb11 (16%)
Respiratory distress 1 (1%)
Acidosis (base excess <−8 mmol/l) 28 (43%)
Hypoglycemia (glucose <3 mmol/l) 11 (16%)
Anemia (hemoglobin <5 g/dl) 21 (30%)
Hemoglobinuria 1 (1%)
Axillary temperature (°C) 38.3 (1.0)
Heart rate (beats/min) 158 (141 to 176)
Respiratory rate (breaths/min) 49 (40 to 58)
Glucose (mg/dl) 102 (88 to 127)
Blood urea nitrogen (mg/dl)c12 (8 to 16)
Hemoglobin (g/dl) 7.1 (5.1 to 9.2)
pHc7.39 (7.33 to 7.43)
HCO3 (mmol/l)c17.8 (13.2 to 21.5)
Base excess (mmol/l)c−7 (−13 to −3)
Aspartate transaminase (U/l)d71 (49 to 116)
Alanine aminotransferase (U/l)d25 (14 to 42)
Total bilirubin (mol/l)d31 (24 to 49)
HIV-positive 3/70 (4.3%)
Parasitemia (parasites/µl); (geometric
mean, 95% CI)
88,391 (53,547 to 145,909)
Plasma PfHRP2 (ng/ml);e (geometric
mean, 95% CI)
1,893 (1,387 to 2,584)
Data are median (interquartile range), mean (SD), or n (%), unless otherwise stated.
BCS, Blantyre Coma Score; CI, confidence interval; GCS, Glasgow Coma Score; PfHRP2,
Plasmodium falciparum histidine-rich protein-2.
aWeight-for-age Z-scores for children ≤10 years43; data missing for n = 1 child of
age >10 years. bChildren with compensated shock (n = 4) and decompensated
shock (n = 7) combined. cMissing data for n = 5 children because of missing i-STAT
measurements. dMissing data for n = 5, n = 6, and n = 4 children with respect to
aspartate transaminase, alanine aminotransferase, and total bilirubin, respectively.
eMissing data because of missing samples in n = 3 children with undetectable
PfHRP2 concentrations.
444 VOLUME 93 NU MBER 5 | MAY 2013 | www.nature.com/cpt
Articles
When body weight was used as a xed allometric function on
all elimination clearance (power of 0.75) and apparent volume of
distribution (power of 1) parameters, a signicantly better model
t was observed (Δobjective function value = −14.6). DHA clear-
ance increased 10.2% per unit (g/dl) of decrease of hemoglobin.
Interindividual variability in clearance decreased from 63.6%
coecient of variation to 55.3% coecient of variation when this
covariate was included, suggesting that it accounts for a limited
but signicant part of the observed variability.
e nal model described the observed data well, with ade-
quate goodness-of-t diagnostics (Figure 1; Eta-shrinkage:
elimination clearance for ARS 10.6%, central volume of dis-
tribution for ARS 12.2%, elimination clearance for DHA 6.81;
Epsilon-shrinkage: 49.4 and 22.5% for ARS and DHA, respec-
tively). A prediction-corrected visual predictive check of the
nal model resulted in no model misspecication and good
simulation properties (Figure 2). e numerical predictive
check (n = 2,000) for ARS resulted in 4.35% (95% CI 1.45–9.42)
and 5.07% (95% CI 1.45–9.42) of observations above and below
the 90% prediction interval, respectively; for DHA, these values
were 2.97% (95% CI 1.79–9.52) and 5.95% (95% CI 1.79–8.92),
respectively.
ARS and DHA exposures aer the standard 2.4 mg/kg dose
were simulated at each body weight level (1,000 simulations
each at 1-kg intervals from 6 to 25 kg) using a uniform distri-
bution of hemoglobin concentrations within the observed range
(2.72–13.6 g/dl) to account for the observed covariate relationship
(Figure 3a,b). Children with body weights between 6 and 10 kg
showed a mean reduction of 20.4% (P < 0.0001) in DHA exposure
as compared with children with body weights between 21 and
25 kg (median (25th to 75th percentile) exposure: 3,380 (2,130–
5,470) ng × h/ml in the 6–10 kg patients, 3,780 (2,430–6,060) ng
Figure 1 Goodness-of-fit diagnostics of the final population pharmacokinetic model of (a,b,c) artesunate and (d,e,f) dihydroartemisinin in children with severe
malaria. The broken line represents a locally weighted least-squares regression; the solid line is the line of identity. The observed concentrations, population
predictions, and individual predictions were transformed into their logarithms (base 10).
10,000
1,000
100
Observations (nmol/l)
10
1
110 100
Population predictions (nmol/l)
1,000 10,000
10,000
1,000
100
Observations (nmol/l)
10
1
10
4
2
0
Conditional weighted residuals
2
4
123
Time (hours)
456
10 100
Individual predictions (nmol/l)
1,000 10,000
10,000
1,000
100
Observations (nmol/l)
10
1
110 100
Population predictions (nmol/l)
1,000 10,000
10,000
1,000
100
Observations (nmol/l)
10
1
10
4
2
0
Conditional weighted residuals
2
4
2
Time (hours)
46810
10 100
Individual predictions (nmol/l)
1,000 10,000
ab c
de f
Figure 2 Simulation-based diagnostics of the final model describing the
population pharmacokinetics of (a,c) artesunate and (b,d) dihydroartemisinin
in children with severe malaria. Graphs in a and b display a prediction-corrected
visual predictive check with venous plasma concentrations transformed into
their logarithms (base 10). Open circles represent observed data points; solid
lines represent the 5th, 50th, and 95th percentiles of the observed data; the
shaded area represents the 95% confidence interval (CI) of simulated (n = 2,000)
5th, 50th, and 95th percentiles. Graphs in c and d display the observed fraction
of data points below the limit of quantification (solid lines) and the 95% CI of the
simulated (n = 2,000) fraction below the limit of quantification (shaded area).
10,000
1.0
0.8
0.6
0.4
Fraction censored
0.2
0
0120
Time (hours)
12
1,000
100
Concentration (nmol/l)
10
1
1234 1
Time (hours)
234
ab
cd
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 93 NUMBER 5 | MAY 2013 445
Articles
× h/ml in the 11–15 kg patients, 4,100 (2,570–6,590) ng × h/ml in
the 16–20 kg patients, and 4,240 (2,700–6,840) ng × h/ml in the
21–25 kg patients).
is suggests that smaller children need higher doses of ARS
to attain ARS exposures similar to those in children with higher
body weights. Using the nal model, we evaluated various dos-
ing regimens based on body weight bands. e proposed regi-
men (Tabl e 3 ) resulted in similar exposures in all weight bands
aer the rst dose of i.m. ARS (Figure 3c,d). e same simula-
tions were performed assuming a uniform distribution of body
weights at dierent levels of hemoglobin; these resulted in lower
DHA exposures with decreasing hemoglobin levels (Figure 4).
ere was no statistical dierence between survivors and non-
survivors with respect to total exposure (ARS P = 0.8060, DHA
P = 0.4828) or maximum concentration (ARS P = 0.7655, DHA
P = 0.6865) aer the rst dose. Similarly, an exposure–response
relationship could not be established using nonlinear mixed-
eects modeling (NONMEM) in a time-to-event approach.
However, this might be a consequence of the relatively low num-
ber of deaths and the high proportion of pretreatments with
dierent antimalarial drugs, doses, and administration routes.
DISCUSSION
Parenteral ARS is currently the drug of choice for the treatment of
severe malaria. Optimal treatment strategies depend on detailed
knowledge of the pharmacokinetic properties of drugs in the tar-
get population in which the drug is used. Age, disease status, and
severity are all factors that may aect drug absorption, distribu-
tion, metabolism, and elimination.
17
e importance of pharma-
cokinetics in determining the therapeutic response is illustrated
by the study of artemether, the first parenteral artemisinin
derivative that was compared with quinine for eectiveness in
the treatment of severe malaria in large clinical trials.24,25 In a
meta-analysis of randomized trials in severe malaria, artemether
signicantly reduced mortality in Southeast Asian adults but did
not do so in African children.26 Subsequent pharmacokinetic
studies showed that the oil-based artemether was released slowly
and erratically from the i.m. injection site, and that this likely
counterbalanced its pharmacodynamic advantages relative to qui-
nine.
10,27
Despite having pharmacodynamic properties similar to
those of artemether, ARS is superior to artemether as a treatment
because of its more favorable pharmacokinetic properties.28
Dosing regimens for children are oen derived from studies
in adults, and this practice has led to substantial underdosing
of several antimalarials in children. A pharmacokinetic study of
sulfadoxine- pyrimethamine in African children with uncom-
plicated falciparum malaria showed that, with the usual dose of
25/1.25 mg/kg, the area under the concentration–time curve in
children 2–5 years of age was half that in adults. is may have
caused failure of antimalarial treatment in small children, thereby
contributing to the spread of resistance. is information came
decades aer the introduction of sulfadoxine-pyrimethamine.
18
It has also been shown that piperaquine exposure levels are lower
in small children if a standard body weight-based dose regimen
is followed.19
Only limited data are available on the pharmacokinetics of
parenteral, rectal, and oral ARS in children. is is the rst
population pharmacokinetic study of i.m. ARS in African chil-
dren with severe malaria. A one-compartment model accurately
described the distribution of ARS and DHA. ARS was converted
rapidly into DHA, the ARS elimination half-life being ~26 min.
is is in agreement with ARS half-life values reported in other
Figure 3 Simulated total first-dose exposure levels (AUC0–12h) of (a) ARS and (b) DHA after the standard 2.4 mg/kg dosing in children at different body weights.
Simulated total first-dose exposure levels (AUC0–12h) of (c) ARS and (d) DHA after the suggested adjusted dose regimen (Table 3). Open circles represent median
values, and bars indicate the 25th to 75th percentiles of simulations (1,000 simulations at each body weight). The broken line represents the median exposure for
the largest weight group (i.e., 700 h × ng/ml and 1,230 h × ng/ml for ARS and DHA, respectively). ARS, artesunate; AUC0–12h, area under the concentration–time
curve from time point 0 to 12 h; DHA, dihydroartemisinin.
6
0
500
1,000
ARS AUC012 h (h × ng/ml)
1,500
810121416
Body weight (kg)
18 20 22 24 6810 12 14 16
Body weight (kg)
18 20 22 24
a
0
500
1,000
DHA AUC012 h (h × ng/ml)
2,500
2,000
1,500
b
6
0
500
1,000
ARS AUC012 h (h × ng/ml)
1,500
810121416
Body weight (kg)
18 20 22 24 6810 12 14 16
Body weight (kg)
18 20 22 24
c
0
500
1,000
DHA AUC012 h (h × ng/ml)
2,500
2,000
1,500
d
446 VOLUME 93 NU MBER 5 | MAY 2013 | www.nature.com/cpt
Articles
pharmacokinetic studies of i.m. ARS
9,10,29
but is considerably
longer than that associated with the i.v. route.
20,30
is is because
the elimination rate of ARS is limited by the rate of absorption
from the i.m. injection site (i.e., “ip-op” pharmacokinetics).9
e volume of distribution values, maximum concentration
values, and area under the concentration–time curves of ARS
and DHA are also comparable with the ndings from a previ-
ous small and densely sampled pharmacokinetic study of i.m.
ARS in children.
9
However, the area under the concentration–
time curve of DHA in this study was considerably lower than
that reported in adult patients and healthy volunteers aer i.v.
administration of ARS.30,31 Despite the lower predicted maxi-
mum concentration value aer i.m. injection as compared with
i.v. administration, this is still far greater than the in vitro
dened DHA concentration value of 2.28 ng/ml that is required
for 99% inhibition (IC
99
). e excellent bioavailability aer i.m.
injection (~90%),
9,29
fast absorption, and comparable estimates
for ARS and DHA exposure support the use of i.m. ARS as a
suitable alternative to i.v. ARS.30
Our study had only a relatively small number of patients, thereby
limiting its power to detect covariate relationships. A parsimonious
Table 3 Proposed body weight–adjusted dosing regimen for i.m.
artesunate
Weight (kg) Dose i.m. (mg)
Prepared
solution (ml)aDose i.m. (mg/kg)
6–7 20 2 2.86–3.33
8–9 25 2.5b2.78–3.13
10–11 30 3b2.73–3.00
12–13 35 3.5b2.69–2.92
14–16 40 2 2.50–2.86
17–20 50 2.5 2.50–2.94
21–25 60 3 2.40–2.86
i.m., intramuscular.
aFor children of body weights <14 kg, dilute to 10 mg/ml; for children with body
weights ≥14 kg, dilute to 20 mg/ml. bDivide the dose equally and administer into both
thighs.
Figure 4 (a) Simulated total first-dose exposure levels (AUC0–12h) of DHA
after the standard 2.4 mg/kg dosing in children at different hemoglobin
levels. Open circles represent median values, and bars indicate the 25th
to 75th percentiles of simulations (1,000 simulations at each hemoglobin
level). (b) Simulated total first-dose exposure (AUC0–12h) of DHA in children at
different body weights at very low (open circles: 3 g/dl), low (open squares:
8 g/dl), and normal (open triangles: 13 g/dl) hemoglobin levels. The broken
line represents the median exposure for the largest weight group (i.e.,
1,230 h × ng/ml). AUC0–12h, area under the concentration–time curve from
time point 0 to 12 h; DHA, dihydroartemisinin.
3456789
Hemoglobin (g/dl)
10 11 12 13
6
0
1,000
2,000
3,000
4,000
0
1,000
2,000
3,000
4,000
810121416
Body weight (kg)
18 20 22 24
a
b
DHA AUC
012 h
(h × ng/ml)
DHA AUC012 h (h × ng/ml)
Table 2 Parameter estimates of the final model describing
the population pharmacokinetics of artesunate and
dihydroartemisinin in children (n = 70) with severe malaria
Variable
Population
estimatea (% RSEb)95% CIb
Fixed effects
CL/FARS (l/h) 45.8 (8.10) 38.8–53.7
V/FARS (l) 28.2 (11.4) 22.7–35.2
CL/FDHA (l/h) 22.4 (8.40) 19.2–26.5
V/FDHA (l) 13.5 (9.69) 11.2–16.3
DUR (min) 1.00 (fixed)
Covariate effect
Negative effect of hemoglobin
on CL/F DHA (%)
10.2 (14.9) 6.84–12.8
Random effects
ηCL/F ARS 0.415 (45.3) 0.0890–0.755
ηV/F ARS 0.680 (54.6) 0.111–1.373
ηCL/F ARS ~ ηV/F ARS 0.497 (52.3) 0.0732–0.969
ηCL/F DHA 0.306 (37.9) 0.136–0.546
σ
ARS 0.0942 (29.2) 0.0266–0.249
σ
DHA 0.211 (12.5) 0.122–0.320
Post hoc estimatesc
CL/FARS (l/h/kg) 4.27 1.18–11.0
V/FARS (l/kg) 2.58 0.479–8.06
t1/2 ARS (h) 0.425 0.238–0.727
Cmax ARS (ng/ml) 943 329–5,090
AUC0–12h ARS (h × n/ml) 570 281–2,170
CL/FDHA (l/h/kg) 2.01 0.736–5.95
V/FDHA (l/kg) 1.24
t1/2 DHA (h) 0.427 0.145–1.18
Tmax DHA (h) 0.608 0.321–1.04
Cmax DHA (ng/ml) 547 284–890
AUC0–12h DHA (h × ng/ml) 890 297–2,510
ARS, artesunate; AUC0–12h, area under the concentration–time curve from time point
0 to 12 h; CL/F, elimination clearance; Cmax, predicted maximum concentration;
DHA, dihydroartemisinin; DUR, duration of zero-order absorption; F, intramuscular
bioavailability; t1/2, terminal elimination half-life; Tmax, time to maximum
concentration; V/F, central volume of distribution; η, interindividual variability;
ηCL/F ~ ηV/F , correlation of random effects on CL/F and V/F; σ, additive residual variance.
aComputed population mean values from nonlinear mixed-effects modeling are
calculated for a typical patient with a body weight of 10.9 kg and a hemoglobin
value of 7.1 g/dl. bAssessed by nonparametric bootstrap method (n = 974 successful
iterations out of 1,000) of the final pharmacokinetic model. Relative standard error
(% RSE) is calculated as 100 × (SD/mean value). 95% Confidence interval (95% CI) is
displayed as the 2.5–97.5 percentiles of bootstrap estimates. cPost hoc estimates are
displayed as median values with 2.5–97.5 percentiles of empirical Bayes estimates.
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 93 NUMBER 5 | MAY 2013 447
Articles
approach (P < 0.001 for a covariate to be retained in the model) was
applied in order to avoid artifactual covariate relationships. e
most signicant covariate identied in the present study was body
weight. is has also been reported in other population pharma-
cokinetic studies of oral and rectal ARS in pediatric32,33 and mixed
adult–pediatric
13
populations. In general, physiological processes
do not scale linearly with body weight; consequently children
with lower body weights will have higher body weight–normal-
ized elimination clearance values. is has also been reported
previously for other antimalarials.
18,19
Dosing simulations using
an exact dose of 2.4 mg/kg resulted in lower ARS and DHA expo-
sures in small children as compared with those with body weights
of 25 kg, suggesting the need for higher dosing in small children. In
addition, hemoglobin concentration was also a signicant covari-
ate, resulting in lower DHA exposure in more anemic children.
Severe anemia is a common presenting feature of severe malaria
in young children. In severe malarial anemia there is usually sub-
stantial intravascular hemolysis, and the released heme may cause
iron-mediated degradation of the artemisinin peroxide bridge.
is is also the cause of the degradation of ARS in hemolyzed
plasma samples ex vivo.
34
Hemoglobin concentration was found
to be associated with reduced exposure levels of ARS and DHA,
independent of body weight. is supports the idea that young
children with severe malaria, who are generally more anemic than
older children, should receive adjusted higher doses.35,36
Underdosing in young children with severe malaria may have
immediate adverse consequences with respect to outcomes.
Parenteral and oral ARS are extremely well tolerated. e only
dose-dependent toxicity identied to date is neutropenia. A
recent study carried out in Cambodia has shown that oral ARS
at a dose of 6 mg/kg/day for 7 days resulted in a reduction in
neutrophil counts (to <1.0 × 10
3
/μl) and short-term neutropenia
in 19% of the patients.
37
Because oral ARS has a bioavailability of
~80%,
38,39
this corresponds to a total parenteral dose of 33.6 mg/
kg, which would be reached only if parenteral treatment were
continued for 14 doses. In the AQUAMAT trial, the median
(interquartile range) number of doses of parenteral treatment
in surviving children was 3 (2–4) doses.6
ARS is currently the rst-choice treatment for severe malaria,
and a product produced according to Good Manufacturing
Practices, which is WHO prequalied and available. To facilitate
implementation of an optimized dosing regimen in the treatment
of severe malaria in African children, we dened a simplied
weight-band–based dosing regimen based on the current popu-
lation pharmacokinetics model, taking into account accuracy and
practicality issues. We considered 0.5 ml to be the minimum vol-
ume of prepared ARS solution that can be measured accurately
and administered with commonly available types of syringes.
Because the weight bands are smaller for children with body
weights <14 kg, we propose an incremental ARS dose increase of
5 instead of 10 mg and a dilution of 10 mg/ml for i.m. administra-
tion in this group, similar to the dosage regimen in the current
study. In children with body weights of <14 kg, doses should be
split and administered in both thighs if injection volumes exceed
2 ml. In children of body weights ≥14 kg, larger injection volumes
can be avoided by using a dilution of 20 mg/ml. Binning of weight
bands was also chosen in accordance with the currently available
vial, which contains a dose of 60 mg, demarcating the upper limits
of weight bands at 25 kg.
The dosing recommendations we propose do not extend
beyond the weight ranges of the children included in this study.
No children with body weight <6.5 kg were included; therefore
more studies of ARS population pharmacokinetics are needed to
evaluate dosing in these very small children and to support the
current dosing recommendations. More extensive sampling in
the rst 15 min and during the rst 12 h aer the dose could give
more information than the current study provided.
In conclusion, ARS and DHA exposures were lower in small
children aer i.m. administration of ARS in severe malaria, war-
ranting dose adaptation in this group. Independently, hemolytic
anemia may aggravate the lower exposures in young children. We
propose a body weight–adjusted and convenient dosing regimen
for i.m. ARS in children with body weights between 6 and 25 kg.
METHODS
Study design. This pharmacokinetic assessment of ARS was part of
the AQUAMAT trial (registration number ISRCTN 50258054), a large
multinational trial for which the results have been published else-
where.6 This substudy was conducted at Teule Hospital in Muheza,
Tanzania, from May 2009 to July 2010. Except for the additional blood
sampling, the procedures for this substudy were part of the AQUAMAT
study protocol.6 Ethical approval was obtained from the Tanzania
Medical Research Coordinating Committee and the Oxford Tropical
Research Ethics Committee. A total of 18 patients were co-enrolled in
the “FEAST” trial evaluating fluid bolus therapy in children with com-
pensated shock.40
Children ≤14 years with a clinical diagnosis of severe malaria con-
rmed by Plasmodium lactate dehydrogenase (pLDH)-based rapid diag-
nostic test (OptiMAL, Diamed, Cressier, Switzerland) were recruited,
and written informed consent was obtained from the respective parents
or caregivers. Severe malaria was dened as the presence of at least one
of the following: coma (Glasgow Coma Score ≤10 or Blantyre Coma
Score ≤2 in preverbal children), convulsions (duration >30 min or ≥2
episodes in the 24 h preceding admission), respiratory distress (nasal alar
aring, costal indrawing/recession or use of accessory muscles, severe
tachypnea) or acidotic breathing (“deep” breathing), shock (capillary rell
time ≥3 sec and/or temperature gradient and/or systolic blood pressure
<70 mm Hg), severe symptomatic anemia (<5 g/dl with respiratory dis-
tress), hypoglycemia (<3 mmol/l), hemoglobinuria, severe jaundice, or,
in older children, a convincing history of anuria or oliguria. Patients
who had received full treatment with parenteral quinine or a parenteral
artemisinin derivative >24 h before admission were excluded.
Physical examination was carried out at admission, and a venous blood
sample was taken for peripheral blood parasite count, quantitative assess-
ment of plasma Plasmodium falciparum histidine-rich protein-2 (a meas-
ure of total body parasite burden),41 HIV serology (SD Bio-Line HIV
1/2 3.0; Standard Diagnostics, Kyonggi-do, Korea/Determine HIV-1/2,
Abbott Laboratories, IL), blood culture, liver function tests (aspartate ami-
notransferase, alanine transaminase, γ-glutamyltransferase, total bilirubin,
creatinine, and urea, by Reotron, Roche Diagnostics, Basel, Switzerland),
hematocrit, biochemistry, and acid–base parameters (EC8+ cartridge for
i-STAT handheld blood analyzer, Abbott Laboratories, Abbott Park, IL).
Hematocrit was reported from the i-STAT reading or, when not available,
measured by HemoCue (HemoCue AB, Ängelholm, Sweden) (n = 5). A
neurologic examination was conducted at discharge, and repeated at day
28 in children who had not made a full neurologic recovery at discharge.
Antimalarial treatment. ARS (Guilin Pharmaceutical Factory,
Guangxi, China) was administered as an i.m. injection (2.4 mg/kg)
shortly after admission, again at 12 h and 24 h, and daily thereafter. The
448 VOLUME 93 NU MBER 5 | MAY 2013 | www.nature.com/cpt
Articles
contents of each 60 mg vial of ARS were dissolved in 1 ml 5% sodium
bicarbonate (provided with the drug) and further diluted with 5 ml
5% dextrose (final concentration of 10 mg/ml) before administration
as a deep i.m. injection into the anterolateral thigh. Dosing was based
on the measured body weight of the patient, and injection volumes of
>2–3 ml were split and divided into two injections, one in each thigh.
When the patient was well enough to take oral medication, but after
a minimum of 24 h (two doses of i.m. ARS), a full 3-day course of oral
artemether-lumefantrine (Co-artem; Novartis, Basel, Switzerland) was
given to complete the treatment.
Patient management. Vital signs and glucose were monitored at least
every 6 h and also at any sign of deterioration in clinical condition. A
majority of the patients (i.e., other than those who were able to be orally
fed) received an infusion with dextrose 5%. Hypoglycemia (defined
here as blood glucose <3 mmol/l) was treated with an i.v. bolus of 5 ml/
kg of 10% dextrose. Blood transfusion (20 ml/kg) was given to children
with hemoglobin concentrations of <5 g/dl. Fluid bolus was given to
children with signs of shock.40 All the children were treated empiri-
cally with i.v. antibiotics. Convulsions were treated with i.v. diazepam
or phenobarbitone if persisting. Peripheral blood smears were repeated
after 24 h.
Blood sampling. Blood samples (1.5 ml) were drawn from an indwell-
ing catheter into prechilled fluoride oxalate tubes42 for ARS and DHA
quantification before the first dose (at baseline). Four subsequent sam-
ples were taken from each patient at preset random time points within
the following time windows: 0–1, 1–4, 4–12, and 12–24 h after the first
dose. Randomization of sampling times was done by computer-gener-
ated randomization (STATA version 12 (Stata, College Station, TX).
Immediately aer blood collection, the blood samples for drug mea-
surements were centrifuged at 4 °C at 2,000g for 7 min. Plasma samples
(0.5 ml) were stored at −80 °C and shipped on dry ice to the MORU
Department of Clinical Pharmacology, Bangkok, ailand, for drug
quantication. ARS drug content and quality were checked in vials taken
randomly from the purchase lots (see Supplementary Data online).6
Drug analysis. ARS and DHA plasma concentrations were measured
using liquid chromatography–tandem mass-spectrometry.43 Quality
control samples at low, middle, and high concentrations were analyzed
in triplicate within each analytical batch to ensure accuracy and preci-
sion during the analysis. The total coefficients of variation were <8%
for all quality control samples. The lower limit of quantification was set
at 1.2 ng/ml for ARS and 2.0 ng/ml for DHA.
Pharmacokinetic modeling. Venous plasma concentrations were trans-
formed into molar units and modeled as natural logarithms, using
NONMEM v.7 (ICON Development Solutions, Ellicott City, MD).
ARS and DHA were modeled simultaneously, using a drug–metabolite
model with complete in vivo conversion of ARS into DHA (for details,
see Supplementary Data online). The first-order conditional esti-
mation method with interaction was used throughout the modeling.
Model selection was based on the objective function values computed
by NONMEM, goodness-of-fit graphical analysis, and physiologi-
cal plausibility. Potential covariates were investigated using a stepwise
forward addition and backward elimination approach. A P value of
0.05 was used in the forward step and a P value of 0.001 in the back-
ward step to compensate for the relatively small population studied.
Simulation-based diagnostics (visual and numerical predictive checks)
and bootstrap diagnostics were used to evaluate the performance of the
final model.44
Monte Carlo simulations using the nal model with the observed vari-
ability were performed for dierent body weights to obtain representative
population estimates of the exposure levels during the rst day of dosing
(area under the concentration–time curve from time point 0 to 12 h) aer
prospective dose regimens. No drug exposure target is dened for paren-
teral ARS; therefore, for the purpose of arriving at a practical parenteral
dosing regimen, dierent body weight “bins” were evaluated to ensure
similar target exposures in all weight bands in agreement with the expo-
sure in children of body weight 25 kg. e same simulations were used
to evaluate the eect of other signicant covariates on drug exposure.
Pharmacodynamics. Peripheral blood smears were taken at admis-
sion and after 24 h. Reduction in parasite load over 24 h, survival, and
severe neurologic sequelae were evaluated as part of the pharmacody-
namic analysis. The effects of ARS and DHA exposures on outcomes
were investigated using a time-to-event analysis in NONMEM, with
predicted drug concentrations being used to modulate the hazard
function in a traditional maximum effect (Emax) relationship. Group
comparisons were performed using the nonparametric Mann–
Whitney U-test in STATA.
SUPPLEMENTARY MATERIAL is linked to the online version of the paper at
http://www.nature.com/cpt
ACKNOWLEDGMENTS
We are grateful to the patients and their caregivers. We thank Ben Amos from
Teule Hospital in Muheza for microbiology and laboratory management.
Permission to publish this work was given by the director general, National
Institute for Medical Research, Tanzania. This work was supported by
The Wellcome Trust of Great Britain (grants 076908 and 082541) and was
coordinated as part of the Mahidol-Oxford Tropical Medicine Research
Programme funded by the Wellcome Trust of Great Britain.
AUTHOR CONTRIBUTIONS
J.T., I.C.E.H., and A.M.D. wrote the manuscript. J.T., I.C.E.H., N.L., N.P.J.D., L.v.S.,
N.J.W., and A.M.D. designed research. I.C.E.H., G.M., and A.K. performed
research. J.T., I.C.E.H., and N.L. analyzed data. S.G., H.R., and M.M.L.
contributed new reagents/analytical tools.
CONFLICT OF INTEREST
The authors declared no conflict of interest.
Study Highlights
WHAT IS THE CURRENT KNOWLEDGE ON THIS TOPIC?
3 Parenteral ARS is currently the drug of choice for the
treatment of severe malaria in all age groups. e cur-
rent recommended dosing regimen was, for the most
part, derived empirically from the results of studies
in adults. Most deaths from severe malaria occur in
children, but pharmacokinetic data on i.m. ARS in this
age group are scarce.
WHAT QUESTION DID THIS STUDY ADDRESS?
3 We studied the pharmacokinetic properties of i.m.
ARS and its active metabolite, DHA, in the treatment
of severe malaria in African children. From our nd-
ings, we derived a practical dosing regimen that avoids
underdosing in smaller children.
WHAT THIS STUDY ADDS TO OUR KNOWLEDGE
3 Body weight signicantly aected elimination clearance
and apparent volume of distribution, resulting in lower
exposure levels of ARS and DHA in smaller children.
HOW THIS MIGHT CHANGE CLINICAL PHARMACOLOGY
AND THERAPEUTICS
3 We propose an adapted ARS dosing regimen for small
children, including a practical dosing table per weight
band.
CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 93 NUMBER 5 | MAY 2013 449
Articles
© 2013 American Society for Clinical Pharmacology and Therapeutics
1. World Health Organization. World Malaria Report 2008. <http://www.who.int/
malaria/publications/atoz/9789241563697/en/index.html> (2008). Accessed
10 October 2011.
2. Marsh, K. et al. Indicators of life-threatening malaria in African children. N. Engl.
J. Med. 332, 1399–1404 (1995).
3. Waller, D. et al. Clinical features and outcome of severe malaria in Gambian
children. Clin. Infect. Dis. 21, 577–587 (1995).
4. World Health Organization. Guidelines for the treatment of malaria
2010, second edition-Rev.1. <http://www.who.int/malaria/publications/
atoz/9789241547925/en/index.html> (2011). Accessed 4 May 2011.
5. Dondorp, A., Nosten, F., Stepniewska, K., Day, N. & White, N. Artesunate versus
quinine for treatment of severe falciparum malaria: a randomised trial. Lancet
366, 717–725 (2005).
6. Dondorp, A.M. et al. Artesunate versus quinine in the treatment of
severe falciparum malaria in African children (AQUAMAT): an open-label,
randomised trial. Lancet 376, 1647–1657 (2010).
7. ter Kuile, F., White, N.J., Holloway, P., Pasvol, G. & Krishna, S. Plasmodium
falciparum: in vitro studies of the pharmacodynamic properties of drugs used
for the treatment of severe malaria. Exp. Parasitol. 76, 85–95 (1993).
8. White, N.J. Assessment of the pharmacodynamic properties of antimalarial
drugs in vivo. Antimicrob. Agents Chemother. 41, 1413–1422 (1997).
9. Nealon, C. et al. Intramuscular bioavailability and clinical ecacy of artesunate
in gabonese children with severe malaria. Antimicrob. Agents Chemother. 46,
3933–3939 (2002).
10. Hien, T.T. et al. Comparative pharmacokinetics of intramuscular artesunate
and artemether in patients with severe falciparum malaria. Antimicrob. Agents
Chemother. 48, 4234–4239 (2004).
11. Navaratnam, V., Mansor, S.M., Sit, N.W., Grace, J., Li, Q. & Olliaro, P.
Pharmacokinetics of artemisinin-type compounds. Clin. Pharmacokinet. 39,
255–270 (2000).
12. Davis, T.M. et al. Pharmacokinetics and pharmacodynamics of intravenous
artesunate in severe falciparum malaria. Antimicrob. Agents Chemother. 45,
181–186 (2001).
13. Simpson, J.A. et al. Population pharmacokinetics of artesunate and
dihydroartemisinin following intra-rectal dosing of artesunate in malaria
patients. PLoS Med. 3, e444 (2006).
14. Batty, K.T., Ilett, K.F. & Davis, T.M. Protein binding and alpha : beta anomer ratio
of dihydroartemisinin in vivo. Br. J. Clin. Pharmacol. 57, 529–533 (2004).
15. Newton, P.N. et al. The pharmacokinetics of intravenous artesunate in adults
with severe falciparum malaria. Eur. J. Clin. Pharmacol. 62, 1003–1009 (2006).
16. Li, G.Q., Guo, X.B., Jin, R., Wang, Z.C., Jian, H.X. & Li, Z.Y. Clinical studies on
treatment of cerebral malaria with qinghaosu and its derivatives. J. Tradit.
Chin. Med. 2, 125–130 (1982).
17. Bartelink, I.H., Rademaker, C.M., Schobben, A.F. & van den Anker, J.N.
Guidelines on paediatric dosing on the basis of developmental physiology and
pharmacokinetic considerations. Clin. Pharmacokinet. 45, 1077–1097 (2006).
18. Barnes, K.I., Little, F., Smith, P.J., Evans, A., Watkins, W.M. & White, N.J.
Sulfadoxine-pyrimethamine pharmacokinetics in malaria: pediatric dosing
implications. Clin. Pharmacol. Ther. 80, 582–596 (2006).
19. Tarning, J. et al. Population pharmacokinetics and pharmacodynamics
of piperaquine in children with uncomplicated falciparum malaria. Clin.
Pharmacol. Ther. 91, 497–505 (2012).
20. Kremsner, P.G. et al. A simplied intravenous artesunate regimen for severe
malaria. J. Infect. Dis. 205, 312–319 (2012).
21. Karunajeewa, H.A. et al. Artesunate suppositories versus intramuscular
artemether for treatment of severe malaria in children in Papua New Guinea.
Antimicrob. Agents Chemother. 50, 968–974 (2006).
22. Bethell, D.B. et al. Pharmacokinetics of oral artesunate in children with
moderately severe Plasmodium falciparum malaria. Trans. R. Soc. Trop. Med.
Hyg. 91, 195–198 (1997).
23. Tarning, J. et al. Population pharmacokinetics of dihydroartemisinin and
piperaquine in pregnant and non-pregnant women with uncomplicated
malaria. Antimicrob Agents Chemother 56:1997–2007 (2012).
24. Tran, T.H. et al. A controlled trial of artemether or quinine in V
ietnamese adults with severe falciparum malaria. N. Engl. J. Med. 335, 76–83
(1996).
25. van Hensbroek, M.B. et al. A trial of artemether or quinine in children with
cerebral malaria. N. Engl. J. Med. 335, 69–75 (1996).
26. Artemether-Quinine Meta-analysis Study Group. A meta-analysis using
individual patient data of trials comparing artemether with quinine in the
treatment of severe falciparum malaria. Trans R Soc Trop Med Hyg 95, 637–650
(2001).
27. Murphy, S.A. et al. The disposition of intramuscular artemether in children
with cerebral malaria; a preliminary study. Trans. R. Soc. Trop. Med. Hyg. 91,
331–334 (1997).
28. Phu, N.H. et al. Randomized controlled trial of artesunate or
artemetherinVietnamese adults with severe falciparum malaria. Malar. J. 9,
97 (2010).
29. Ilett, K.F. et al. The pharmacokinetic properties of intramuscular artesunate
and rectal dihydroartemisinin in uncomplicated falciparum malaria. Br. J. Clin.
Pharmacol. 53, 23–30 (2002).
30. Morris, C.A., Duparc, S., Borghini-Fuhrer, I., Jung, D., Shin, C.S.
& Fleckenstein, L. Review of the clinical pharmacokinetics of
artesunateandits active metabolite dihydroartemisinin following
intravenous, intramuscular, oral or rectal administration. Malar. J. 10, 263
(2011).
31. Byak ika-Kibwika, P. et al. Pharmacokinetics and pharmacodynamics of
intravenous artesunate during severe malaria treatment in Ugandan adults.
Malar. J. 11, 132 (2012).
32. Karunajeewa, H.A. et al. Disposition of ar tesunate and dihydroartemisinin
after administration of artesunate suppositories in children from Papua
New Guinea with uncomplicated malaria. Antimicrob. Agents Chemother. 48,
2966–2972 (2004).
33. Stepniewska, K . et al. Population pharmacokinetics of artesunate and
amodiaquine in African children. Malar. J. 8, 200 (2009).
34. Lindegardh, N. et al. Quantication of dihydroartemisinin, artesunate and
artemisinin in human blood: overcoming the technical
challenge of protecting the peroxide bridge. Bioanalysis 3, 1613–1624
(2011).
35. Snow, R.W. et al. Relation between severe malaria morbidity in children and
level of Plasmodium falciparum transmission in Africa. Lancet 349, 1650–1654
(1997).
36. Billig, E.M., O’Meara, W.P., Riley, E.M. & McKenzie, F.E. Developmental allometry
and paediatric malaria. Malar J 11, 64 (2012).
37. Bethell, D. et al. Dose-dependent risk of neutropenia after 7-day
courses ofartesunate monotherapy in Cambodian patients with
acutePlasmodium falciparum malaria. Clin. Infect. Dis. 51, e105–e114
(2010).
38. Batty, K.T. et al. A pharmacokinetic and pharmacodynamic study of
intravenous vs oral artesunate in uncomplicated falciparum malaria. Br. J. Clin.
Pharmacol. 45, 123–129 (1998).
39. McGready, R. et al. Artesunate/dihydroartemisinin pharmacokinetics
in acute falciparum malaria in pregnancy: absorption,
bioavailability,disposition and disease eects. Br. J. Clin. Pharmacol. 73,
467–477 (2012).
40. Maitland, K. et al. Mor tality after uid bolus in African children with severe
infection. N. Engl. J. Med. 364, 2483–2495 (2011).
41. Hendriksen, I.C. et al. Diagnosing severe falciparum malaria in parasitaemic
African children: a prospective evaluation of plasma PfHRP2 measurement.
PLoS Med. 9, e1001297 (2012).
42. Lindegardh, N. et al. Major pitfalls in the measurement of artemisinin
derivatives in plasma in clinical studies. J. Chromatogr. B Analyt. Technol.
Biomed. Life Sci. 876, 54–60 (2008).
43. Hanpithakpong, W. et al. A liquid chromatographic-tandem mass
spectrometric method for determination of artesunate and its metabolite
dihydroartemisinin in human plasma. J. Chromatogr. B Analyt. Technol.
Biomed. Life Sci. 876, 61–68 (2008).
44. Bergstrand, M., Hooker, A.C., Wallin, J.E. & Karlsson, M.O. Prediction-corrected
visual predictive checks for diagnosing nonlinear mixed-eects models.
AAPSJ. 13, 143–151 (2011).
This work is licensed under a Creative
Commons Attribution-NonCommercial-No
Derivative Works 3.0 License. To view a copy of this license,
visit http://creativecommons.org/licenses/by-nc-nd/3.0/
450 VOLUME 93 NU MBER 5 | MAY 2013 | www.nature.com/cpt

Supplementary resource (1)

... However, given the findings in other age groups and favorable PK and PD properties of reliable absorption, high bioavailability, and rapid parasitological response, it can be assumed that artesunate is the most effective treatment in infants as well [23,26,31]. Population PK studies on IV and IM artesunate including infants (average age around three years and both including infants older than six months) inferred, that children with lower body weights had a larger apparent volume of distribution, as well as higher body weight-normalized elimination clearance values resulting in lower plasma concentrations of artesunate and its active metabolite dihydroartemisinin [32,33]. This caused the WHO to increase the recommended dose of artesunate in children <20 kg to 3 mg/kg/dose [7]. ...
... There are no obvious reasons to reject implementation of simplified intramuscular regimens for management of severe malaria in African children as the higher single daily dose suggested obviates the need for modeling and inferential studies [35,36]. WHO's choice to recommend higher doses for infants and small children also takes into account that artesunate treatment is generally safe and well tolerated and aims to minimize risks of suboptimal drug exposure in patients with severe malaria [31,33]. Posttreatment hemolysis occurred in 7-9% of African children treated with artesunate, with 1% requiring blood transfusion on day 14 [37,38]. ...
... The beneficial clinical properties of artesunate observed in older populations are likely to apply to neonates as well. Whether the WHO recommendation of a higher dose of IV artesunate (3 mg/kg) should apply to neonates seems questionable, as this recommendation is based on data from older children [32,33]. Plasma protein-binding capacity in neonates is generally lower compared to infants, increasing the free fraction of protein-bound drugs such as artesunate and renal excretion is generally lower due to immaturity of the renal system [20][21][22]. ...
Article
Introduction: Malaria in infants is common in high-transmission settings, especially in infants >6 months. Infants undergo physiological changes impacting pharmacokinetics and pharmacodynamics of anti-malarial drugs and, consequently, the safety and efficacy of malaria treatment. Yet, treatment guidelines and evidence on pharmacological interventions for malaria often fail to address this vulnerable age-group. This review aims to summarise the available data on anti-malarial treatment in infants. Areas covered: The standard recommended treatments for severe and uncomplicated malaria are generally safe and effective in infants. However, infants have an increased risk of drug-related vomiting and have distinct pharmacokinetic parameters of antimalarials compared with older patients. These include larger volumes of distribution, higher clearance rates and immature enzyme systems. Consequently, infants with malaria may be at increased risk of treatment failure and drug toxicity. Expert opinion: Knowledge expansion to optimize treatment can be achieved by including more infants in antimalarial drug trials and by reporting separately on treatment outcomes in infants. Additional evidence on the efficacy, safety, tolerability, acceptability and effectiveness of ACTs in infants is needed, as well as population pharmacokinetics studies on antimalarials in the infant population.
... The WHO's current treatment guidelines for severe malaria recommend 3 mg/kg IV AS in children weighing less than 20 kg compared with larger children and adults (2.4 mg/ kg/dose) to ensure comparable pharmacokinetic (PK) exposure to the active metabolite of AS, dihydroartemisinin (DHA), in terms of the DHA area under the curve from time 0 to 12 h after drug administration (AUC 0-12 ) [6]. Two published population PK analyses of intramuscular and IV AS compared the predicted DHA AUC across varying adult and pediatric patient populations [7,8]. A published DHA population PK meta-analysis by Zaloumis et al [8] reported that pediatric patients with severe malaria weighing 6 to 10 kg had lower DHA AUC 0-12 compared with those patients weighing 21 to 25 kg, when administered 2.4 mg/kg IV AS (geometric mean decrease of 13.7%; 95% confidence interval, 11.7-15.6%; ...
... Dosing recommendations for IV AS in pediatric patients with severe malaria based on modelbased PK simulations depend upon whether age-dependent maturation of UGT enzymes is considered. Hendriksen et al [7] did not take into account the age-dependent maturation effect and, as a result, recommended increasing the IV AS dose by 25% in children weighing 6 to 20 kg. Our simulations using the Zaloumis et al [8] DHA population PK model indicate that if age-dependent maturation is considered, an IV AS dose of 2.4 mg/kg results in relatively comparable DHA AUC 0-12 in children weighing 10 to 22 kg and a somewhat higher exposure in children weighing 2 to 9 kg. ...
... Second, the time course of glomerular filtration rate-maturation and time course of clearancematuration of drugs metabolized by UGT enzymes are similar [15]. Third, incorporation of this maturation function consistently showed improvement in model fit over the allometric body-weight function alone in population PK analyses by Hendriksen et al and Zaloumis et al, although the improvement was not considered statistically significant by Hendriksen et al [7,8]. ...
Article
For treatment of severe malaria, the WHO recommends 3 mg/kg intravenous artesunate in pediatric patients weighing less than 20 kg. Here we describe FDA's rationale for selecting 2.4 mg/kg in pediatric patients weighing less than 20 kg based on literature review and independent analyses.
... pharmacokinetic modelling of data from over 300 children and adults with severe malaria (3,4). The USFDA has recently challenged this recommendation (5). ...
... Younger children with severe malaria are significantly more anaemic than older children and adults. In both our population pharmacokinetic modelling studies lower hemoglobins were associated with reduced drug exposures (3,4). The USFDA concluded from their simulations that the doses of parenteral artesunate for children should be the same as in adults (2.4mg/kg) (5). ...
Article
Full-text available
To the Editor— Malaria still kills over 1000 children each day, mainly in sub-Saharan Africa. Artesunate is the treatment of choice for severe Plasmodium falciparum malaria as it substantially reduces mortality compared with quinine [1], the previously recommended antimalarial treatment. Since 2015, the World Health Organization (WHO) has recommended parenteral artesunate treatment doses of 3 mg/kg for patients weighing <20 kg (ie. over 90% of African children with severe malaria) [2]. This was based on evidence from population pharmacokinetic modelling of data from over 300 children and adults with severe malaria [3, 4]. The US Food and Drug Administration (US FDA) has recently challenged this recommendation [5]. Using a population pharmacokinetic model that we developed [4], the US FDA simulated a virtual pediatric population with severe malaria from US Centers for Disease Control and Prevention (CDC) growth charts, added a maturation effect for uridine diphosphate (UDP) glucuronosyltransferase (an important contributor to dihydroartemisinin clearance), and predicted slower clearance and thus increased drug exposure (area under the curve [AUC]0 –12 hours) in children < 10 kg. Of note, their simulations assumed incorrectly that weight (and age) are independent of hemoglobin. Younger children with severe malaria are significantly more anemic than older children and adults. In both our population pharmacokinetic modeling studies, lower hemoglobins were associated with reduced drug exposures [3, 4]. The US FDA concluded from their simulations that the doses of parenteral artesunate for children should be the same as in adults (2.4 mg/kg) [5]. In other words, without analyzing any measured drug concentrations, without adjusting for other relevant covariates, and without considering at all the pharmacometrics of the life-saving benefit conferred by artesunate, the US FDA has effectively made a recommendation for dose reduction for one of the major lethal infections of childhood, and it has done this based entirely on an unvalidated model-based simulation.
... Artesunate is currently widely used in fixed combination therapies with other malarial drugs for oral treatment of non-severe malaria and alone as a dual pack formulation for treatment of severe/cerebral malaria via intravenous or intramuscular injection [3,4]. In the latter case, solid artesunate is mixed with 5% aqueous sodium bicarbonate and diluted with aqueous saline or dextrose immediately prior to injection, which must be carried out rapidly to avoid hydrolysis to DHA [12][13][14]. Artesunate administered intravenously has a half-life of less than 15 min; in comparison, DHA generated from artesunate in situ has a half-life of 30-60 min [15,16]. As artesunate is a hemi-ester of DHA, it is intrinsically unstable to hydrolysis via alkyl-oxygen cleavage under acidic conditions, or via acyl-oxygen cleavage under basic conditions [17]. ...
Article
For the purpose of establishing the optimum processing parameters and storage conditions associated with nanolipid formulations of the artemisinin derivative artesunate, it was necessary to evaluate the thermal stability and solubility profiles of artesunate in aqueous solutions at various temperatures and pH. The effect of increased temperature and humidity on artesunate was determined by storing samples of the raw material in a climate chamber for 3 months and analyzing these by an established HPLC method. Artesunate remained relatively stable during storage up to 40°C ± 0.5°C and 75% relative humidity for 3 months, wherein it undergoes approximately 9% decomposition. At higher temperatures, substantially greater decomposition supervenes, with formation of dihydroartemisinin (DHA) and other products. In solution, artesunate is relatively stable at 15°C with less than 10% degradation over 24 h. The aqueous solubility of artesunate at different pH values after 60 min are pH 1.2 (0.1 M HCl) 0.26 mg/mL, pH 4.5 (acetate buffer) 0.92 mg/mL, distilled water 1.40 mg/mL, and pH 6.8 (phosphate buffer) 6.59 mg/mL, thus relating to the amount of ionized drug present. Overall, for optimal preparation and storage of the designated formulations of artesunate, relatively low temperatures will have to be maintained throughout.
... In 2012, the simplified regimen was examined in a randomized controlled trial (RCT) as an alternative to the standard regimen in resource-poor settings in Africa. In 2015, the standard (14,15). The dose for smaller children was increased in the revised regimen to provide comparable drug exposure to adults and larger children. ...
Article
Full-text available
Most deaths from severe falciparum malaria occur within 24 hours of presentation to hospital. Intravenous (i.v.) artesunate is the first-line treatment for severe falciparum malaria, but its efficacy may be compromised by delayed parasitological responses. In patients with severe malaria the life-saving benefit of the artemisinin derivatives is their ability to clear circulating parasites rapidly, before they can sequester and obstruct the microcirculation. To evaluate the dosing of i.v. artesunate for the treatment of artemisinin-sensitive and reduced ring stage sensitivity to artemisinin severe falciparum malaria infections Bayesian pharmacokinetic-pharmacodynamic modelling of data from 94 patients with severe malaria (80 children from Africa and 14 adults from Southeast Asia) was performed. Assuming delayed parasite clearance reflects a loss of ring stage sensitivity to artemisinin derivatives, the median (95% credible interval) percentage of patients clearing ≥99% parasites within 24 hours (PC24≥99%) for standard (2.4 mg/kg i.v. artesunate at 0 and 12 hours) and simplified (4 mg/kg i.v. artesunate at 0 hours) regimens were 65% (52.5%-74.5%) versus 44% (25%-61.5%) for adults, 62% (51.5%-74.5%) versus 39% (20.5%-58.5%) for larger children (≥20 kg) and 60% (48.5%-70%) versus 36% (20%-53.5%) for smaller children (<20 kg). The upper limit of the credible intervals for all regimens was below a PC24≥99% of 80%, a threshold achieved on average in clinical studies of severe falciparum malaria infections. Rapid clearance of parasites, where there is loss of ring stage sensitivity to artemisinin, in patients with severe falciparum malaria is compromised with the currently recommended and proposed simplified i.v. artesunate dosing regimens.
Article
The majority of deaths from malaria are in young African children. Parenteral artesunate is the first‐line treatment for severe falciparum malaria. Since 2015 the World Health Organization has recommended individual doses of 3 mg/kg for children weighing less than 20 kg. Recently, the US Food and Drug Administration (FDA) has challenged this recommendation, based on a simulated pediatric population, and argued for a lower dose in younger children (2.4 mg/kg). In this study, we performed population pharmacokinetic modeling of plasma concentration data from 80 children with severe falciparum malaria in the Democratic Republic of Congo who were given 2.4 mg/kg of artesunate intravenously. Bayesian hierarchical modeling and a two‐compartment parent drug‐metabolite pharmacokinetic model for artesunate were used to describe the population pharmacokinetics of artesunate and its main biologically active metabolite dihydroartemisinin. We then generated a virtual population representative of the target population in which the drug is used and simulated the total first‐dose exposures. Our study shows that the majority of younger children given the lower 2.4 mg/kg dose of intravenous artesunate do not reach the same drug exposures as older children above 20 kg. This finding supports withdrawal of the FDA's recent lower artesunate dose recommendation as parenteral artesunate is an extremely safe and well‐tolerated drug and there is potential for harm from underdosing in this rapidly lethal infection.
Article
Full-text available
The combination antimalarial therapy of artemisinin-naphthoquine (ART-NQ) was developed as a single-dose therapy, aiming to improve adherence relative to the multiday schedules of other artemisinin combination therapies. The pharmacokinetics of ART-NQ has not been well characterized, especially in children. A pharmacokinetic study was conducted in adults and children over 5 years of age (6 to 10, 11 to 17, and ≥18 years of age) with uncomplicated malaria in Tanzania. The median weights for the three age groups were 20, 37.5, and 55 kg, respectively. Twenty-nine patients received single doses of 20 mg/kg of body weight for artemisinin and 8 mg/kg for naphthoquine, and plasma drug concentrations were assessed at 13 time points over 42 days from treatment. We used nonlinear mixed-effects modeling to interpret the data, and allometric scaling was employed to adjust for the effect of body size. The pharmacokinetics of artemisinin was best described by one-compartment model and that of naphthoquine by a two-compartment disposition model. Clearance values for a typical patient (55-kg body weight and 44.3-kg fat-free mass) were estimated as 66.7 L/h (95% confidence interval [CI], 57.3 to 78.5 L/h) for artemisinin and 44.2 L/h (95% CI, 37.9 to 50.6 L/h) for naphthoquine. Nevertheless, we show via simulation that patients weighing ≥70 kg achieve on average a 30% lower day 7 concentration compared to a 48-kg reference patient at the doses tested, suggesting dose increases may be warranted to ensure adequate exposure. (This study has been registered at ClinicalTrials.gov under identifier NCT01930331.).
Article
Malaria is a leading cause of death in children less than 5 years of age globally, and a common cause of fever in the returning North American traveler. New tools in the fight against malaria have been developed over the past decades: potent artemisinin derivatives; rapid diagnostic tests; long-lasting insecticidal bed nets; and a new vaccine, RTS,S/AS01. Thwarting these advances, parasite and Anopheles vector resistance are emerging. In the meantime, clinicians will continue to see malaria among febrile travelers from the tropics. Early recognition, diagnosis, and treatment can be lifesaving, but rely on the vigilance of frontline clinicians.
Article
Artesunate (ARS) is the only artemisinin-based intravenous drug approved for treatment of malaria in the clinic. ARS is rapidly metabolized in vivo to short lived (∼30-45 min) but fast acting, dihydroartemisinin (DHA). The short half-life of DHA necessitates multiple dose administration to circumvent the risk of recrudescence and development of artemisinin resistance. In this work, we report a stable, safe and potent alternative artemisinin-based injectable nanocomplex consisting of dimeric artesunate-choline conjugate (dACC) micelles coated with hyaluronic acid (HA). Firstly, dACC was synthesized by one-step esterification of two artesunate molecules with 3-(dimethylamino)-1,2-propanediol followed by quaternization. After that, dACC was self-assembled into cationic nanomicelles and further coated with anionic small molecular weight HA. The HA-coated dACC nanocomplex (dACC/HA nanocomplex) has a narrow size distribution of about 30 nm. Hemolytic toxicity and cytotoxicity studies revealed a favorable bio-safety profile. Finally, in vitro and in vivo studies showed the dACC/HA nanocomplex possess superior safety and antimalarial efficacy compared to ARS. Taken together, the dACC/HA nanocomplex is a promising injectable alternative to the traditional clinically used artesunate.
Article
Full-text available
In African children, distinguishing severe falciparum malaria from other severe febrile illnesses with coincidental Plasmodium falciparum parasitaemia is a major challenge. P. falciparum histidine-rich protein 2 (PfHRP2) is released by mature sequestered parasites and can be used to estimate the total parasite burden. We investigated the prognostic significance of plasma PfHRP2 and used it to estimate the malaria-attributable fraction in African children diagnosed with severe malaria.Admission plasma PfHRP2 was measured prospectively in African children (from Mozambique, The Gambia, Kenya, Tanzania, Uganda, Rwanda, and the Democratic Republic of the Congo) aged 1 month to 15 years with severe febrile illness and a positive P. falciparum lactate dehydrogenase (pLDH)-based rapid test in a clinical trial comparing parenteral artesunate versus quinine (the AQUAMAT trial, ISRCTN 50258054). In 3,826 severely ill children, Plasmadium falciparum PfHRP2 was higher in patients with coma (p = 0.0209), acidosis (p
Article
Full-text available
Background Severe malaria is a medical emergency with high mortality. Prompt achievement of therapeutic concentrations of highly effective anti-malarial drugs reduces the risk of death. The aim of this study was to assess the pharmacokinetics and pharmacodynamics of intravenous artesunate in Ugandan adults with severe malaria. Methods Fourteen adults with severe falciparum malaria requiring parenteral therapy were treated with 2.4 mg/kg intravenous artesunate. Blood samples were collected after the initial dose and plasma concentrations of artesunate and dihydroartemisinin measured by solid-phase extraction and liquid chromatography-tandem mass spectrometry. The study was approved by the Makerere University Faculty of Medicine Research and Ethics Committee (Ref2010-015) and Uganda National Council of Science and Technology (HS605) and registered with ClinicalTrials.gov (NCT01122134). Results All study participants achieved prompt resolution of symptoms and complete parasite clearance with median (range) parasite clearance time of 17 (8–24) hours. Median (range) maximal artesunate concentration (Cmax) was 3260 (1020–164000) ng/mL, terminal elimination half-life (T1/2) was 0.25 (0.1-1.8) hours and total artesunate exposure (AUC) was 727 (290–111256) ng·h/mL. Median (range) dihydroartemisinin Cmax was 3140 (1670–9530) ng/mL, with Tmax of 0.14 (0.6 – 6.07) hours and T1/2 of 1.31 (0.8–2.8) hours. Dihydroartemisinin AUC was 3492 (2183–6338) ng·h/mL. None of the participants reported adverse events. Conclusions Plasma concentrations of artesunate and dihydroartemisinin were achieved rapidly with rapid and complete symptom resolution and parasite clearance with no adverse events.
Article
Full-text available
Dihydroartemisinin-piperaquine is being increasingly used as a first-line artemisinin combination treatment for malaria. The aim of this study was to describe the pharmacokinetic and pharmacodynamic properties of piperaquine in 236 children with uncomplicated falciparum malaria in Burkina Faso. They received a standard body weight-based oral 3-day fixed-dose dihydroartemisinin-piperaquine regimen. Capillary plasma concentration-time profiles were characterized using nonlinear mixed-effects modeling. The population pharmacokinetics of piperaquine were described accurately by a two-transit-compartment absorption model and a three-compartment distribution model. Body weight was a significant covariate affecting clearance and volume parameters. The individually predicted day 7 capillary plasma concentration of piperaquine was an important predictor (P < 0.0001) of recurrent malaria infection after treatment. Young children (2-5 years of age) received a significantly higher body weight-normalized dose than older children (P = 0.025) but had significantly lower day 7 piperaquine concentrations (P = 0.024) and total piperaquine exposures (P = 0.021), suggesting that an increased dose regimen for young children should be evaluated.
Article
Full-text available
Pregnant women are particularly vulnerable to malaria. The pharmacokinetic properties of antimalarial drugs are often affected by pregnancy, resulting in lower drug concentrations and a consequently higher risk of treatment failure. The objective of this study was to evaluate the population pharmacokinetic properties of piperaquine and dihydroartemisinin in pregnant and nonpregnant women with uncomplicated malaria. Twenty-four pregnant and 24 matched nonpregnant women on the Thai-Myanmar boarder were treated with a standard fixed oral 3-day treatment, and venous plasma concentrations of both drugs were measured frequently for pharmacokinetic evaluation. Population pharmacokinetics were evaluated with nonlinear mixed-effects modeling. The main pharmacokinetic finding was an unaltered total exposure to piperaquine but reduced exposure to dihydroartemisinin in pregnant compared to nonpregnant women with uncomplicated malaria. Piperaquine was best described by a three-compartment disposition model with a 45% higher elimination clearance and a 47% increase in relative bioavailability in pregnant women compared with nonpregnant women. The resulting net effect of pregnancy was an unaltered total exposure to piperaquine but a shorter terminal elimination half-life. Dihydroartemisinin was best described by a one-compartment disposition model with a 38% lower relative bioavailability in pregnant women than nonpregnant women. The resulting net effect of pregnancy was a decreased total exposure to dihydroartemisinin. The shorter terminal elimination half-life of piperaquine and lower exposure to dihydroartemisinin will shorten the posttreatment prophylactic effect and might affect cure rates. The clinical impact of these pharmacokinetic findings in pregnant women with uncomplicated malaria needs to be evaluated in larger series.
Article
We conducted a meta-analysis using individual patient data from randomized controlled trials comparing artemether and quinine in severe falciparum malaria. Eleven trials were identified, of which 8 were clearly randomized. Original individual patient data on 1919 patients were obtained from 7 trials, representing 85% of the patients in the original 11 studies. Overall there were 136 deaths among the 961 patients treated with artemether, compared with 164 in the 958 treated with quinine [14% vs 17%, odds ratio (95% confidence interval) 0·8 (0·62 to 1·02), P = 0·08]. There were no differences between the 2 treatment groups in coma recovery or fever clearance times, or the development of neurological sequelae. However, the combined ‘adverse outcome’ of either death or neurological sequelae was significantly less common in the artemether group [odds ratio (95% CI) 0·77 (0·62 to 0·96), P = 0·02], and treatment with artemether was associated with significantly faster parasite clearance [hazard ratio (95% CI) 0·62 (0·56 to 0·69), P < 0·001]. In subgroup analyses artemether was associated with a significantly lower mortality than quinine in adults with multisystem failure. In the treatment of severe falciparum malaria artemether is at least as effective as quinine in terms of mortality and superior to quinine in terms of overall serious adverse events. There was no evidence of clinical neurotoxicity or any other major side-effects associated with its use.
Article
Aims To obtain comprehensive pharmacokinetic and pharmacodynamic data for artesunate (ARTS) and its active metabolite dihydroartemisinin (DHA) following i.v. and oral administration of ARTS to patients with acute, uncomplicated falciparum malaria. Methods Twenty-six Vietnamese patients with falciparum malaria were randomized to receive either i.v. ARTS (120 mg; group 1) or oral ARTS (100 mg; group 2), with the alternative preparation given 8 h later in an open crossover design. Mefloquine (750 mg) was administered at 24 h. Plasma concentrations of ARTS and DHA were determined by h.p.l.c. assay. Pharmacokinetic parameters were calculated by non-compartmental methods. The time to 50% parasite clearance (PCT50 ) was calculated by linear interpolation of parasite density determinations. Linear least squares and multiple linear regression analyses were used to evaluate pharmacokinetic-pharmacodynamic relationships. Results Following i.v. bolus, ARTS had a peak concentration of 29.5 μm (11 mg l−1 ), elimination t1/2=2.7 min, CL=2.33 l h−1 kg−1 and V=0.14 l kg−1. The Cmax for DHA was 9.3 μm (2.64 mg l−1 ), t1/2=40 min, CL=0.75 l h−1 kg−1 and V=0.76 l kg−1. Following oral ARTS, relative bioavailability of DHA was 82%, Cmax was 2.6 μm (0.74 mg l−1 ), t1/2=39 min, and MAT=67 min. Overall, the PCT50 and fever clearance time (FCT) were 6.5 h and 24 h, respectively. There was no correlation between PCT50 or FCT and AUC, Cmax or MRT for DHA. Conclusions Despite rapid clearance of ARTS and DHA in patients with uncomplicated falciparum malaria, prompt parasite and fever clearance were achieved. High relative bioavailability of DHA following oral ARTS administration, and clinical outcomes comparable with those after i.v. ARTS, support the use of the oral formulation in the primary care setting.
Article
Aims To obtain pharmacokinetic data for artesunate (ARTS) and its active metabolite dihydroartemisinin (DHA) following i.m. ARTS and rectal DHA administration. Methods Twelve Vietnamese patients with uncomplicated falciparum malaria were randomized to receive either i.v. or i.m. ARTS (120 mg), with the alternative preparation given 8 h later in an open crossover design. A further 12 patients were given i.v. ARTS (120 mg) at 0 h and rectal DHA (160 mg) 8 h later. Results Following i.v. bolus, ARTS had a peak concentration of 42 µm (16 mg l−1), elimination t1/2 = 3.2 min, CL = 2.8 l h−1 kg−1 and V = 0.22 l kg−1. The Cmax for DHA was 9.7 µm (2.7 mg l−1), t1/2 = 59 min, CL = 0.64 l h−1 kg−1 and V = 0.8 l kg−1. Following i.m. ARTS, Cmax was 2.3 µm (3.7 mg l−1), the apparent t1/2 = 41 min, CL = 2.9 l h−1 kg−1 and V = 2.6 l kg−1. The relative bioavailability of DHA was 88%, Cmax was 4.1 µm (1.16 mg l−1) and t1/2 = 64 min. In the rectal DHA study, relative bioavailability of DHA was 16%. Conclusions For patients with uncomplicated falciparum malaria i.m. ARTS is a suitable alternative to i.v. ARTS, at equal doses. To achieve plasma DHA concentrations equivalent to parenteral administration of ARTS, rectal DHA should be given at approximately four-fold higher milligram doses. Further studies are needed to determine whether these recommendations can be applied to patients with severe malaria.
Article
BACKGROUND: In the treatment of severe malaria, intravenous artesunate is more rapidly acting than intravenous quinine in terms of parasite clearance, is safer, and is simpler to administer, but whether it can reduce mortality is uncertain. METHODS: We did an open-label randomised controlled trial in patients admitted to hospital with severe falciparum malaria in Bangladesh, India, Indonesia, and Myanmar. We assigned individuals intravenous artesunate 2.4 mg/kg bodyweight given as a bolus (n=730) at 0, 12, and 24 h, and then daily, or intravenous quinine (20 mg salt per kg loading dose infused over 4 h then 10 mg/kg infused over 2-8 h three times a day; n=731). Oral medication was substituted when possible to complete treatment. Our primary endpoint was death from severe malaria, and analysis was by intention to treat. FINDINGS: We assessed all patients randomised for the primary endpoint. Mortality in artesunate recipients was 15% (107 of 730) compared with 22% (164 of 731) in quinine recipients; an absolute reduction of 34.7% (95% CI 18.5-47.6%; p=0.0002). Treatment with artesunate was well tolerated, whereas quinine was associated with hypoglycaemia (relative risk 3.2, 1.3-7.8; p=0.009). INTERPRETATION: Artesunate should become the treatment of choice for severe falciparum malaria in adults.