+ All Categories
Home > Documents > Interspecies Allometric Scaling of Antimalarial Drugs and Potential Application to Pediatric Dosing

Interspecies Allometric Scaling of Antimalarial Drugs and Potential Application to Pediatric Dosing

Date post: 02-Dec-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
12
Published Ahead of Print 4 August 2014. 10.1128/AAC.02538-14. 2014, 58(10):6068. DOI: Antimicrob. Agents Chemother. S. M. D. K. Ganga Senarathna and Kevin T. Batty Application to Pediatric Dosing Antimalarial Drugs and Potential Interspecies Allometric Scaling of http://aac.asm.org/content/58/10/6068 Updated information and services can be found at: These include: REFERENCES http://aac.asm.org/content/58/10/6068#ref-list-1 at: This article cites 151 articles, 38 of which can be accessed free CONTENT ALERTS more» articles cite this article), Receive: RSS Feeds, eTOCs, free email alerts (when new http://journals.asm.org/site/misc/reprints.xhtml Information about commercial reprint orders: http://journals.asm.org/site/subscriptions/ To subscribe to to another ASM Journal go to: on September 15, 2014 by Curtin University Library http://aac.asm.org/ Downloaded from on September 15, 2014 by Curtin University Library http://aac.asm.org/ Downloaded from
Transcript

  Published Ahead of Print 4 August 2014. 10.1128/AAC.02538-14.

2014, 58(10):6068. DOI:Antimicrob. Agents Chemother. S. M. D. K. Ganga Senarathna and Kevin T. Batty Application to Pediatric DosingAntimalarial Drugs and Potential Interspecies Allometric Scaling of

http://aac.asm.org/content/58/10/6068Updated information and services can be found at:

These include:

REFERENCEShttp://aac.asm.org/content/58/10/6068#ref-list-1at:

This article cites 151 articles, 38 of which can be accessed free

CONTENT ALERTS more»articles cite this article),

Receive: RSS Feeds, eTOCs, free email alerts (when new

http://journals.asm.org/site/misc/reprints.xhtmlInformation about commercial reprint orders: http://journals.asm.org/site/subscriptions/To subscribe to to another ASM Journal go to:

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

on S

eptember 15, 2014 by C

urtin University Library

http://aac.asm.org/

Dow

nloaded from

Interspecies Allometric Scaling of Antimalarial Drugs and PotentialApplication to Pediatric Dosing

S. M. D. K. Ganga Senarathna,a,b Kevin T. Battya,b

School of Pharmacy, Faculty of Health Sciences, Curtin University, Bentley, Western Australia, Australiaa; Curtin Health Innovation Research Institute, Curtin University,Bentley, Western Australia, Australiab

Pharmacopeial recommendations for administration of antimalarial drugs are the same weight-based (mg/kg of body weight)doses for children and adults. However, linear calculations are known to underestimate pediatric doses; therefore, interspeciesallometric scaling data may have a role in predicting doses in children. We investigated the allometric scaling relationships ofantimalarial drugs using data from pharmacokinetic studies in mammalian species. Simple allometry (Y � a � Wb) was utilizedand compared to maximum life span potential (MLP) correction. All drugs showed a strong correlation with clearance (CL) inhealthy controls. Insufficient data from malaria-infected species other than humans were available for allometric scaling. Theallometric exponents (b) for CL of artesunate, dihydroartemisinin (from intravenous artesunate), artemether, artemisinin, clin-damycin, piperaquine, mefloquine, and quinine were 0.71, 0.85, 0.66, 0.83, 0.62, 0.96, 0.52, and 0.40, respectively. Clearance wassignificantly lower in malaria infection than in healthy (adult) humans for quinine (0.07 versus 0.17 liter/h/kg; P � 0.0002) anddihydroartemisinin (0.81 versus 1.11 liters/h/kg; P � 0.04; power � 0.6). Interpolation of simple allometry provided better esti-mates of CL for children than MLP correction, which generally underestimated CL values. Pediatric dose calculations based onsimple allometric exponents were 10 to 70% higher than pharmacopeial (mg/kg) recommendations. Interpolation of interspeciesallometric scaling could provide better estimates than linear scaling of adult to pediatric doses of antimalarial drugs; however,the use of a fixed exponent for CL was not supported in the present study. The variability in allometric exponents for antimalar-ial drugs also has implications for scaling of fixed-dose combinations.

The World Health Organization and standard pharmacopeialsources recommend that antimalarial drugs be administered

at the same weight-based (mg/kg of body weight) or linear dosefor children and adults (1). However, this linear method of dosagecalculation is known to underestimate the optimum pediatricdose of many drugs (2–5). Recent clinical reports indicate thathigher doses of sulfadoxine-pyrimethamine (6, 7), chloroquine(6, 8, 9), quinine (10), piperaquine (11–14), and artesunate (15,16) are required for effective antimalarial therapy in children.

Allometric scaling is a well-established technique for relatingphysiological or pharmacokinetic parameters to body weight (17–22), and allometric relationships have been demonstrated for awide range of drugs, including antimicrobial agents (23–27). Theconventional applications of interspecies allometric scaling in-clude prediction of human doses by extrapolation from preclini-cal animal studies and dose estimates for other mammalian spe-cies in veterinary medicine (18, 23, 26, 28). Increasing interest hasbeen shown in the application of interspecies allometric scalingdata to predict pharmacokinetic parameters or drug doses in chil-dren (27, 29, 30). Despite a paucity of reports on antimalarialdrugs (31, 32), interpolation of allometric scaling of chloroquinesupports recommendations from clinical studies for higher chlo-roquine doses in children (8, 9, 33).

The rationale for using scaling techniques to estimate pharma-cokinetic parameters in children is that relatively few pediatricpharmacokinetic investigations are conducted, compared to adultstudies, and sparse sampling is normally required because thereare practical and ethical limitations to obtaining rich pharmaco-kinetic data sets from pediatric subjects (34). One alternative toconventional interspecies scaling is fixed-exponent scaling fromhuman adult pharmacokinetic data (2–5, 34–37). The principle ofscaling with a fixed exponent (e.g., 2/3 or 3/4) may provide some

consistency and practical advantages; however, the validity of a uni-versal exponent in pharmacokinetics is subject to conflicting evi-dence and debate (3, 5, 27, 34–40). Regardless of the scaling methodthat is used, caution is required for recommendations for very youngchildren, due to immature clearance mechanisms, and for diseasestates for which there are limited data available or evidence that clear-ance may be altered in compromised patients (30, 35, 36).

We sought to investigate the allometric scaling relationships ofantimalarial drugs using data from pharmacokinetic studies ofhealthy and malaria-infected mammalian species. Our hypothesiswas that linear scaling (i.e., exponent of 1.0) would not apply andthat interpolation of interspecies allometric scaling would be suit-able for estimating pediatric doses of antimalarial drugs.

MATERIALS AND METHODSData collection. A comprehensive literature search was conducted viaPubMed, OvidSP, Google Scholar, and citation records, using relevant keywords, including the specific antimalarial drugs and mammalian species.The initial target list of antimalarial drugs was determined from WHOtreatment guidelines (1): artemisinin, artesunate, artemether, dihydroar-temisinin, mefloquine, amodiaquine, piperaquine, chloroquine, quinine,primaquine, lumefantrine, sulfadoxine-pyrimethamine, atovaquone,proguanil, tetracycline, doxycycline, and clindamycin. Exclusions com-prised fixed-dose combinations where data for individual drugs were not

Received 12 February 2014 Returned for modification 6 May 2014Accepted 25 July 2014

Published ahead of print 4 August 2014

Address correspondence to Kevin Batty, [email protected].

Copyright © 2014, American Society for Microbiology. All Rights Reserved.

doi:10.1128/AAC.02538-14

6068 aac.asm.org Antimicrobial Agents and Chemotherapy p. 6068 – 6078 October 2014 Volume 58 Number 10

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

readily available (sulfadoxine-pyrimethamine, artemether-lumefantrine,atovaquone-proguanil), drugs for which pharmacokinetic data wereavailable from fewer than three mammalian species (amodiaquine andlumefantrine), and drugs that are contraindicated in children (tetracy-cline and doxycycline). Chloroquine (33) and doxycycline (41) had beenstudied previously and were excluded.

The pharmacokinetic studies were screened for suitability of the data,with a focus on uniformity of biological matrix (especially for quinolineswith high blood-plasma ratio), period over which blood and plasma sam-ples were collected for the pharmacokinetic study (preferably �3 half-lives), route of administration, and use of validated analytical methods.Although pharmacokinetic parameters from studies with intravenous (orparenteral) drug administration are preferred for allometric scaling, otherroutes of administration were more appropriate for some antimalarials, suchas those which are available only as oral formulations. The final list of drugs(and routes of administration) was as follows: artemisinin (intravenous [IV],intraperitoneal, and oral), artesunate (IV) and the active artemisinin metab-olite dihydroartemisinin (from IV artesunate), artemether (IV and intramus-cular), mefloquine (oral), piperaquine (oral), quinine (IV), and clindamycin(IV).

Pharmacokinetic parameters. Clearance (CL), volume of distribu-tion during terminal phase (Vz), and half-life (t1/2) data were collated ordetermined from the available data using model-independent equations(CL � k � V; Vz � CL/k). Comprehensive pharmacokinetic data (e.g.,mean residence time [MRT] and steady-state volume of distribution[Vss]) were reported in a limited range of studies and therefore were notincluded in the present analysis. In studies where different doses wereadministered to the same subjects and there was no evidence of dose-dependent variability, the mean value of the pharmacokinetic parameterwas used. However, if separate groups were given different doses within astudy, the data were treated independently except when the results werereported as group data. In human studies where body weight was notreported, 60 kg was used for Asian and African subjects, whereas 70 kg wasused for Caucasian subjects, based on standard references and other re-ports used in the present study. The body weight of animals was notprovided in some nonhuman studies; hence, published standard bodyweights of the respective species were used for the allometric scaling (17,25, 28, 42).

Allometric scaling. Simple allometry was the principal method of in-terspecies scaling, using the equation

Y � a � Wb (1)

where Y is the pharmacokinetic parameter (e.g., CL or Vz), W is the bodyweight of the species, a is the coefficient, and b is the allometric exponent(19, 24, 38).

The pharmacokinetic parameter data were plotted against bodyweight on a log-log scale (SigmaPlot version 12.5; Systat Software, Inc.,Chicago, IL) to determine the allometric coefficient and exponent by re-gression analysis.

Data from healthy control subjects were used for the allometric scal-ing, as there were insufficient pharmacokinetic studies in malaria-infectednonhuman species for each antimalarial drug that was investigated. How-ever, pharmacokinetic parameters from human (adults) studies of ma-laria infection were compared to the healthy-control data, as a guide to theapplication of allometric interpolation to decisions on pediatric doses.

Maximum life span potential (MLP) correction. Several alternativemethods of scaling and use of correction factors have been proposed andreviewed (17–19, 26, 43, 44), although the physiological relevance andapplication of some methods were not applicable in the present study. Themost relevant and best-studied method for our consideration was MLPcorrection, which is an integral feature of the “rule of exponents” ap-proach (26, 44). According to the rule of exponents, simple allometry isthe best predictor for exponents in the range from 0.50 to 0.70, whereasCL � MLP provides the best prediction method when the range is 0.71 to0.99 (44). As the CL exponent from simple allometry was �1 for all anti-malarial drugs, we compared MLP correction to simple allometry.

The maximum life span potential (MLP) was calculated from theequation (17)

MLP � 10.839 � BW0.636 � B�0.225 (2)

where BW is brain weight in grams and B is body weight in grams (thecoefficient 10.839 is replaced with 185.5 if the brain and body weights arein kg [44]). Brain and body weight data for mammalian species are wellestablished (17, 42, 45).

The CL � MLP data were plotted against body weight on a log-logscale (SigmaPlot) to determine the coefficient and exponent by regressionanalysis. Clearance exponents from simple allometry and MLP correctionwere used to determine interpolated CL for children and compared toavailable clinical study data.

Pediatric doses. A standard allometric model for pediatric dosing is touse one of the following equations:

dosechild � doseadult � �weightchild ⁄ weightadult�b (3)

CLchild � CLadult � �weightchild ⁄ weightadult�b (4)

where dosechild is the total dose for a child at the specified weight(weightchild), doseadult is the standard total dose for an adult at the speci-fied weight (weightadult), and CLchild and CLadult are the total CL of thedrug.

The exponent derived by allometric scaling (equation 1) was appliedin the present study (equation 3) to compare calculated doses to pharma-copeial or reference doses for arbitrary weights of 15 kg and 25 kg (chil-dren approximately 4 and 8 years of age, respectively). Dose estimates forchildren less than 2 years were not considered, due to the known physio-logical and pharmacokinetic differences between very young infants andadults (35, 36).

Statistical analyses. Data analysis and representation were performedwith SigmaPlot version 12.5 (Systat Software, Inc., Chicago, IL). Data aremeans � standard deviations (SD) unless otherwise indicated. The 95%confidence interval (CI) was determined for the allometric exponent[95% CI � mean � (1.96 � standard error)]. The Student t test was usedfor two-sample comparison as appropriate, with significance at a P valueof �0.05.

RESULTSSimple allometry. Simple allometric scaling data for CL and Vz

are shown in Tables 1 and 2 respectively. A strong correlation wasfound for both CL and Vz for all antimalarials except Vz for theprodrug artesunate (Tables 1 and 2). Artemether (Fig. 1) is nor-mally administered by intramuscular injection, but the inclusionof IV data from a rodent and rabbit study did not significantly alterthe allometric parameters. Artemisinin was given parenterally inthe studies of mice and rats, but as oral administration was used inhuman studies, CL data were corrected for oral bioavailability tofacilitate scaling (Table 1). Due to the mixed sources of data, ar-temisinin was excluded from subsequent analyses.

The 95% CI for CL of piperaquine, mefloquine, and quininedid not encompass 2/3 or 3/4, hence the application of these fixedexponents could not be supported by the present data (Table 1,Fig. 1). Linear dosing of piperaquine could be supported, based onthe 95% CI for CL in the present analysis (Table 1).

The 95% CI for Vz encompassed unity for three of the sevendrugs: artemether, piperaquine, and quinine (Table 2). Volume ofdistribution is normally used in calculations of loading dose andin pharmacokinetic modeling; therefore, no further analysis of Vz

was undertaken for the purposes of the present study.Control versus malaria. In the absence of pharmacokinetic

data from malaria-infected nonhuman species for each antimalar-ial drug, a direct comparison of pharmacokinetic parameters fromhuman (adult) healthy controls and malaria-infected patients was

Allometric Scaling of Antimalarial Drugs

October 2014 Volume 58 Number 10 aac.asm.org 6069

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

performed (Table 3). Artesunate could be excluded on the basisthat dose predictions according to the active metabolite dihydro-artemisinin may be more appropriate. Apart from quinine anddihydroartemisinin, which showed significantly lower CL in ma-laria infection than in healthy controls, the P values were �0.1 andthe power of the analyses were �30% for the other antimalarialdrugs. Despite limited data, it was also apparent that CL of quininein malaria infected children is likely to be significantly lower thanCL in healthy controls (Table 3). The power of the analysis fordihydroartemisinin CL was �80%; hence, a type I error cannot beexcluded in the present study.

MLP correction. The MLPs for mice, rats, rabbits, dogs, pigs,human adults, 15-kg children, and 25-kg children were 2.7, 4.8,8.7, 20, 16, 93, 109, and 106 years, respectively. The effect of MLPcorrection is therefore to multiply CL 3- to 5-fold in rodents, 16-to 20-fold in dogs, pigs, sheep, and cows (17), 35-fold in horses,93-fold in adult humans, and �100-fold in human children. Theresult was that scaling extended over four orders of magnitude(Fig. 2). Compared to simple allometry (Table 1), the r2 for MLP-corrected scaling of CL for dihydroartemisinin (0.99), mefloquine(0.96), and quinine (0.98) improved, but the r2 for artemether(0.98), clindamycin (0.96), and piperaquine (0.99) was similar orslightly lower.

The effect of MLP correction on interpolated prediction of CLin children, compared to simple allometry, is shown in Table 4.Results for interpolation of simple allometry for artemether andquinine are illustrated in Fig. 1, with interpolation at 10 kg and 15

kg, respectively, to facilitate comparison with clinical study data(Table 4). Dihydroartemisinin scaling by both simple allometryand MLP correction is provided in Fig. 2. Predictions based onsimple allometry were close to the results from clinical studies ofdihydroartemisinin and artemether, albeit in children with ma-laria infection (Table 4). In contrast, simple allometry showed anoverestimation of CL for mefloquine and quinine. It was observedthat MLP correction routinely underestimated CL (Table 4).

Pediatric doses. Pediatric dose calculations were based onpharmacopoeial (mg/kg) recommendations (1) and were 10 to70% higher, according to allometric predictions (Table 5). Meflo-quine and quinine had the lowest exponent, which led to predic-tions 1.6- to 2.5-fold higher than reference doses.

Although quinine CL is significantly lower in malaria infectionthan in healthy controls and data presented in Table 3 suggest thatquinine CL is lower in malaria-infected children than in adults(contrary to allometric principles), the allometric exponent forquinine CL (Table 1) was used in the present analysis. Cautiousinterpretation of the result for quinine is therefore required. How-ever, increased doses for dihydroartemisinin (as IV artesunate),clindamycin, and mefloquine appear to be warranted (a piper-aquine dose increase would likely not be clinically relevant orpractical).

DISCUSSION

The present study has demonstrated that interpolation of inter-species allometric scaling could be a useful strategy to include in

TABLE 1 Simple allometric scaling data for clearance (CL) of antimalarial drugs in healthy mammals

DrugRoute ofadministrationa No. of speciesb r2

Allometric exponent(95% CI)

Allometriccoefficient References

Artesunate IV 4 (r, d, p, h) 0.92 0.71 (0.53–0.88) 7.0 55, 58–62Dihydroartemisinin (from

IV artesunate)IV 4 (r, d, p, h) 0.94 0.85 (0.68–1.03) 3.3 55, 58–63

Artemether IM 3 (r, d, h) 0.98 0.66 (0.38–0.93) 3.3 59, 64–66Artemether IM or IV 4 (r, rb, d, h) 0.99 0.66 (0.56–0.75) 3.3 59, 64–67Artemisinin IV, IP, oral�Fc 3 (m, r, h) 0.92 0.83 (0.69–0.96) 4.2 68–79Clindamycin IV 3 (d, r, h) 0.98 0.62 (0.55–0.69) 1.4 80–87Piperaquine Oral 3 (m, r, h) 0.99 0.96 (0.86–1.05) 1.6 88–92Mefloquine Oral 3 (m, r, h) 0.90 0.52 (0.43–0.61) 0.2 93–110Quinine IV 3 (r, d, h) 0.89 0.40 (0.33–0.47) 1.9 111–123a IV, intravenous; IM, intramuscular; IP, intraperitoneal.b Species: m, mouse; r, rat; d, dog; p, pig; rb, rabbit; h, human.c Artemisinin dose was corrected for relative oral bioavailability (oral:intramuscular); F � 0.32 (78).

TABLE 2 Simple allometric scaling data for volume of distribution (Vz) of antimalarial drugs in healthy mammals

DrugRoute ofadministrationa No. of speciesb r2

Allometric exponent(95% CI)

Allometriccoefficient References

Artesunate IV 4 (r, d, p, h) 0.69 0.54 (0.22–0.85) 3.2 55, 58–62Dihydroartemisinin (from

IV artesunate)IV 4 (r, d, p, h) 0.99 0.86 (0.78–0.94) 4.2 55, 58–63

Artemether IM 3 (r, d, h) 0.99 0.93 (0.82–1.05) 12.4 59, 64–66Artemether IM or IV 4 (r, rb, d, h) 0.96 1.06 (0.75–1.38) 6.7 59, 64–67Clindamycin IV 3 (d, r, h) 0.98 0.81 (0.72–0.91) 2.5 80–86Piperaquine Oral 3 (m, r, h) 0.94 1.16 (0.87–1.45) 280 88–92Mefloquine Oral 3 (m, r, h) 0.91 0.78 (0.66–0.90) 39.8 93–110Quinine IV 4 (r, rb, d, h) 0.92 0.88 (0.74–1.01) 4.61 111–124a IV, intravenous; IM, intramuscular.b Species: m, mouse; r, rat; d, dog; p, pig; rb, rabbit; h, human.

Senarathna and Batty

6070 aac.asm.org Antimicrobial Agents and Chemotherapy

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

development and refinement of pediatric doses for antimalarialdrugs. We found strong allometric relationships for seven drugs(dihydroartemisinin, artemether, artemisinin, clindamycin, pip-eraquine, mefloquine, and quinine) and showed that linear scal-ing of clearance is not applicable, although the 95% confidenceintervals spanned unity for dihydroartemisinin and piperaquine(Table 1). In contrast, linear scaling of the volume of distributionappeared to be valid for some drugs, notably artemether and pip-eraquine (Table 2).

Allometric scaling is founded on sound theoretical principlesand evidence of a power law relationship between biological pa-

rameters and body weight; however, there are conflicting reportson the application of fixed exponents and correction factors (20,35, 36, 38–40, 46, 47). Historical research on basal metabolic rateestablished a 2/3 power scaling of body mass and is supported bythe concept of scaling surface area to volume of 3-dimensionalobjects (20, 39). The “1/4 power law” is based on seminal studiesthat scaled the metabolic rate of animals using an exponent of 3/4and is reported to be consistent with structure and function ob-servations in biology (20, 21, 38, 39). This power law has beentranslated to the use of an exponent of 3/4 for drug clearance, 1/4for elimination half-life, and 1 for volume of distribution (21, 22,35, 36, 38, 39). It has recently been argued that the small numericaldifference between 2/3 and 3/4 powers is of little clinical relevancein pharmacokinetics (36).

The physiological origins of interspecies relationships providesome context for the limitations of allometric scaling in pharma-cokinetics, which have been the subject of detailed review (39,48–50). One limitation of scaling pharmacokinetic parametersthat we encountered was a small range of mammalian species andlow number of subjects in each study (typically 6 to 16 in humanstudies and 6 to 8 in other species). We were able to obtain datafrom only two or three nonhuman species, a finding that is con-sistent with similar previous investigations (24, 31, 33). However,some pharmacokinetic studies, especially veterinary reports, werecompiled with data from at least 8 species (17, 19, 23, 51), andinterspecies scaling of physiological parameters, such as basalmetabolic rate, comprises data from over 20 species (21). A largerrange of animal studies would therefore enhance the quality ofallometric scaling data in future studies.

Further limitations may occur if there are species differences inthe pharmacokinetic properties of drugs that lead to unpredict-able and weak allometric scaling outcomes (39, 49, 50). Highlyprotein-bound drugs (�98% in humans), such as mefloquine(52) and piperaquine (53), may have inconsistent scaling if bind-ing is substantially lower in other species. Bioavailability and renalor hepatic clearance mechanisms also may have species differenceswhich could be relevant for antimalarial drugs that are subject tohepatic metabolism and commonly administered as oral formu-lations. However, there is generally a paucity of relevant data toaddress these potential limitations in the scaling of pharmacoki-netic parameters (49, 50).

Notwithstanding the limitations of interspecies scaling and thepower law relationship debate, allometric scaling data indicatethat fixed exponents of 2/3 or 3/4 may not be universally applica-ble in pharmacokinetics (24, 28, 29, 31, 33, 38, 44, 47). Our anal-yses showed that the 95% CI encompassed both 2/3 and 3/4 for CLof artemether and artemisinin, whereas the upper limit of the 95%CIs for mefloquine, quinine, and chloroquine (0.63, based on re-analysis of a previous study [33]) were all �0.65 (Table 1). Incontrast, the allometric exponents for dihydroartemisinin andpiperaquine CL were 0.85 and 0.96, respectively, and the 95% CIof the exponent spanned unity. Hence, the use of a fixed exponentfor CL of antimalarial drugs is not supported by the present study.

The influence of allometric exponents on interpolated dosepredictions can be substantial and varies according to bodyweight. For example, if the recommended adult dose of a drug is10 mg/kg (for a 70-kg adult), arbitrary exponents of 0.6 and 0.75would lead to predicted doses of 18.5 mg/kg and 14.7 mg/kg, re-spectively, for a 15-kg child and of 15.1 mg/kg and 12.9 mg/kg,respectively, for a 25-kg child. The differential between adult and

FIG 1 Simple allometric scaling relationship with 95% confidence interval(---) for CL of artemether and quinine. Artemether scaling (top) compriseddata from 5 studies (Œ). The scaled estimate of CL for a 10-kg child (o) was 1.5liters/h/kg. By comparison, the mean adult CL is 0.88 liter/h/kg (Table 3), theestimated CL based on a fixed exponent of 3/4 is 1.4 liters/h/kg, and the CLfrom one clinical study in children (9.5 kg) was 1.5 liters/h/kg (150). Quininescaling (bottom) comprised data from 12 studies (�). The scaled estimate ofCL for a 15-kg child (Œ) was 0.37 liter/h/kg. By comparison, the mean adult CLis 0.17 liter/h/kg (Table 3), the estimated CL based on a fixed exponent of 3/4is 0.25 liter/h/kg, and the CL from one clinical study in children (15 kg) was0.24 liter/h/kg (118).

Allometric Scaling of Antimalarial Drugs

October 2014 Volume 58 Number 10 aac.asm.org 6071

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

child doses will decrease for children with higher body weights andwill also decrease as the exponent approaches unity. The potentialeffect of these inverse relationships on dose predictions demon-strates the importance of high-quality data for allometric scaling.

It was evident from our detailed literature search that an exten-sive range of interspecies scaling for antimalarial drugs was pre-cluded by several factors. In particular, the target list of antimalar-ials was limited by a paucity of data for individual drugs withinsome common, fixed-dose combinations, and we could not ob-tain pharmacokinetic data from a minimum of three mammalianspecies for amodiaquine and lumefantrine. Studies with intrave-nous (or parenteral) drug administration would have been opti-mal, but inclusion of other routes of administration was necessaryfor artemisinin, artemether, mefloquine, and piperaquine. An im-portant issue was the small body of pharmacokinetic research inmalaria-infected nonhuman species for the antimalarial drugs,and this limitation was addressed by comparing drug clearancefrom studies in malaria-infected human adults with that reportedfor healthy controls (Table 3). Consistent with previous reports,only quinine was shown conclusively to have significantly differ-ent clearance in malaria infection (54). Our data indicate thatdihydroartemisinin clearance was higher in healthy controls thanpatients with malaria infection (Table 3); however, the power ofthe analysis was inconclusive. The apparent clearance (CL/F) ofpiperaquine and mefloquine could be higher in malaria infectionthan in healthy controls (Table 3) and may be a function of alteredbioavailability, as has been reported for dihydroartemisinin (55).

Our investigation of the relevance of correction factors for al-lometric scaling was to ascertain if the more sophisticated ap-proaches to interpolation of interspecies scaling would be benefi-cial for antimalarial drugs. Some of the techniques require rawconcentration-time data (17, 18, 43), which were not available inmost reports, and we therefore confined our analysis to maximumlife span potential (MLP). This correction method has been incor-porated in the rule of exponents, whereby simple allometry is usedfor exponents in the range from 0.50 to 0.77 and scaling CL �MLP against body weight is recommended when the exponentfrom simple allometry is in the range from 0.71 to 0.99 (44).

Our study showed that MLP correction led to substantiallylower interpolated CL estimates, compared to simple allometry

TABLE 3 Clearance of antimalarial drugs in malaria-infected patients compared to healthy adults

DrugRoute ofadministrationa

CL (liters/h/kg) (no.of groupsb) Healthy vs. malaria

ReferencesbHealthy Malaria P value Power

Artesunate IV 1.9 � 0.6 (3) 2.9 � 0.8 (8) 0.09 0.4 55, 60, 62, 125–130Dihydroartemisinin (from

IV artesunate)IV 1.11 � 0.16 (3) 0.81 � 0.2 (8) 0.04 0.56 55, 60, 62, 125–131

Artemether IM 0.88 � 0.35 (2) 1.8 � 0.9 (2) 0.32 0.13 64, 65, 132, 133Piperaquinec Oral 0.86 � 0.65 (6) 1.55 � 0.73 (4) 0.15 0.28 56, 88, 90, 91, 134–

137, 154Mefloquine Oral 0.030 � 0.014 (17) 0.039 � 0.014 (5) 0.24 0.21 94–109, 138, 139Quinine IV 0.17 � 0.05 (6) 0.07 � 0.03 (9) 0.0002 0.99 112, 114–117, 122,

140–145Quinine (children) IV 0.24 (1) 0.064 � 0.014 (5) NAd NA 118, 146, 147a IV, intravenous; IM, intramuscular.b Data are means � SD. Some studies comprised several groups.c Piperaquine studies included piperaquine alone and piperaquine-dihydroartemisinin data for healthy volunteers, but only piperaquine-dihydroartemisinin data for patients withmalaria. Our data and a previous report (148) indicate that there is no significant difference in piperaquine clearance when administered alone or in combination withdihydroartemisinin.d NA, not applicable.

FIG 2 Allometric scaling relationship for clearance of dihydroartemisinin.(Top) Simple allometric scaling of data from 7 studies (Œ). The scaled estimateof CL for a 15-kg child (o) was 2.2 liters/h/kg. By comparison, the mean adultCL is 1.1 liters/h/kg (Table 3), the estimated CL based on a fixed exponent of3/4 is 1.6 liters/h/kg, and the CL from one clinical study in children (13 kg) was2.2 liters/h/kg (149). (Bottom) CL � MLP correction (y axis) for the same 7studies (�). The scaled estimate of CL for a 15-kg child (Œ) is 0.53 liter/h/kg.

Senarathna and Batty

6072 aac.asm.org Antimicrobial Agents and Chemotherapy

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

and data from clinical reports, except for one group of childrentreated with mefloquine (Table 4). We conclude that the applica-tion of MLP correction for antimalarial drugs is not supported bythe present study. By comparison, simple allometry overestimatedthe CL/F of mefloquine in children with uncomplicated falcipa-rum malaria, which could lead to excessive dose predictions, andtherefore, this requires more detailed, contemporary clinical in-vestigations. Conversely, interpolation from simple allometryprovided good estimates of CL for dihydroartemisinin and arte-mether (Table 4). The piperaquine data appear to be conflicting;however, there were several clinical differences between the twopopulations and no conclusive explanation for the findings (56,57). Two recent reports of piperaquine pharmacokinetics showedlower CL/F values, suggesting that the rule of exponents may be

applicable to piperaquine, but as noted in Table 4, these data couldnot be included in the present study due to the use of pooledresults from different partner drugs (12) and use of capillary bloodanalysis to determine the pharmacokinetic parameters (13).

Quinine predictions are problematic, due to the difference inCL (and Vz) between healthy subjects and patients with malaria,and the reported lower CL in children than adults (Table 3).Therefore, cautious interpretation of allometric scaling results arerequired for quinine, due to the complex and multifactorial effectsof malaria infection on the pharmacokinetic properties of thisdrug.

The final component of our study was to translate the allomet-ric scaling results to dose estimates (Table 5). The recommenda-tions for dihydroartemisinin (as IV artesunate) and clindamycin

TABLE 4 Antimalarial drug clearance from clinical studies in children compared to interpolated CL from simple allometry and MLP correction

Drug

Allometric exponenta

Interpolated CL(liters/h/kg)a Clinical studyb

Simpleallometry

MLPcorrection

Simpleallometry

MLPcorrection CL (liters/h/kg) Wt (kg) Reference

Dihydroartemisinin 0.85 1.31 2.21 0.48 2.16 13 149Artemether 0.66 1.18 1.52 0.35 1.5 9.5 150Piperaquine 0.96 1.46 1.40 0.50 1.85c 16 56Piperaquine 0.96 1.46 1.39 0.57 0.85c 19.1 57Mefloquine 0.52 1.0 0.068 0.023 0.046d 9.5e 151Mefloquine 0.52 1.0 0.068 0.023 0.048d 9.5e 152Mefloquine 0.52 1.0 0.045 0.024 0.026d 23 153Quininef 0.40 0.93 0.37 0.13 0.24 15.4 118a Allometry was conducted in healthy controls; CL/F was determined for piperaquine and mefloquine.b Data from studies in malaria-infected children; CL/F was determined for piperaquine and mefloquine.c Piperaquine CL/F was determined from studies of piperaquine-dihydroartemisinin in malaria-infected children. There is no significant difference in piperaquine clearance whenpiperaquine is administered alone or in combination with dihydroartemisinin (148). One recent study reported a CL/F of 0.57 liter/h/kg; however, the data were pooled from aprevious study of piperaquine-dihydroartemisinin (57) and an investigation of piperaquine-artemisinin (12). A report of a piperaquine CL/F of 0.42 liter/h/kg from capillary bloodsamples could not be compared directly to investigations using venous plasma samples for determination of piperaquine pharmacokinetic parameters (13).d Mefloquine CL/F was determined from studies of mefloquine-sulfadoxine-pyrimethamine in malaria-infected children. Mefloquine clearance may be lower when mefloquine isadministered in combination with sulfadoxine and pyrimethamine (101).e Malaria-infected children were all �2 years of age in these studies; the mean age was 1.6 years.f All quinine data are from studies in healthy controls.

TABLE 5 Allometric interpolation of antimalarial doses in comparison to current pharmacopeial recommendations

DrugRoute ofadministration Standard regimena

Dose (mg) for:

15-kg child 25-kg child

Referenceb Allometryc Referenceb Allometryc

Dihydroartemisinin(as IVartesunate)

IV Dose at 0, 12, 24 h, then once/day(2.4 mg/kg/dose)

35 45 60 70

Artemetherd Oral Twice/day for 3 days 40 30 60 40Clindamycin Oral Twice/day for 7 days (10 mg/kg/dose) 150 260 250 360Piperaquine Oral Once/day for 3 days (18 mg/kg/day) 270 290 450 470Mefloquine Oral 2/3 total dose day 1; 1/3 total dose

day 2 (25-mg/kg total dose)375 790 625 1,025

Quinine IV Three times per day for 5–7 days (10mg/kg/dose)

150 370 250 460

a As reported by the World Health Organization (1) and confirmed by reference to the British National Formulary.b Based on mg/kg doses and practical recommendations.c Based on reference doses and equation 3, where adult dose was according to a 70-kg body weight and the allometric exponent was from Table 1. Doses were mostly rounded to a10-mg increment rather than the nearest practical dose.d Current artemether dose recommendation is for body weight range (1, 2, 3, and 4 20-mg tablets for patients weighing 5 to 14 kg, 15 to 24 kg, 25 to 34 kg, and �34 kg,respectively). The adult (70 kg) dose is therefore the same as that for a child of 34 kg (80 mg), which equates to 1.1 mg/kg for adults and 2.3 mg/kg for the 34-kg child. Hence,current practical dose recommendations are higher (mg/kg) for children and vary within the weight ranges.

Allometric Scaling of Antimalarial Drugs

October 2014 Volume 58 Number 10 aac.asm.org 6073

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

would be modest increases that can be achieved in the clinicalsetting, whereas the small predicted increase in piperaquine dosewould not be practical. It is notable that recent clinical studieshave recommended higher doses for piperaquine (11, 13, 14) andartesunate in children (15). Indeed, the doses proposed in thelatter study closely match the allometric predictions of 3 mg/kg(15-kg child) and 2.8 mg/kg (25-kg child), based on scaling fordihydroartemisinin in the present study.

The recommended mefloquine doses were 1.6- to 2-foldhigher than reference doses for adults (mg/kg), which could beviewed as excessive and to the best of our knowledge have not beenproposed in any clinical studies. However, recent allometric scal-ing of chloroquine indicated that pediatric doses 1.6-fold higherthan those for adults (mg/kg) were potentially appropriate, andthese doses were exceeded by clinical studies where 2-fold-higherdoses were used in children (8, 9, 33). Complementary data areavailable for quinine, notwithstanding our cautious interpreta-tion of the allometric scaling. Indeed, our results from simpleallometry of quinine suggest that 18 to 24 mg/kg could be moreappropriate than 10 mg/kg (Table 5), and a recent study supportsearlier recommendations in adults that 20 mg/kg quinine shouldbe used as a loading dose in children (10).

An important consideration from our allometric exponentdata is that disproportionate dose increases appear to apply toantimalarial drugs, and the implications for rational pediatric dos-ing of combination regimens are therefore significant. For exam-ple, an adult dose of a dihydroartemisinin-piperaquine (120/960mg; approximately 2/16 mg/kg for a 60-kg patient) oral fixed-dosecombination is reduced by linear calculations to 30/240 mg for a15-kg child. However, based on our allometric interpolation, thiswould result in an appropriate scaled dose of piperaquine and adihydroartemisinin dose that is 20% lower than optimum. Allo-metric scaling of fixed-dose combinations will be valid only if thesame exponent applies to both drugs (or a default-to-fixed-expo-nent scaling is used); however, our data indicate this is unlikely tobe appropriate for artemisinin-based combination therapies, dueto mismatch of the allometric exponents (Table 1).

We conclude that interpolation of interspecies allometric scal-ing is plausible for estimation of pediatric doses of antimalarialdrugs and would be valuable in designing clinical pharmacoki-netic or efficacy studies. The limitations of allometric scaling arewell documented; however, the ongoing use of linear scaling ofadult to pediatric doses (mg/kg) is recognized as flawed (36) andshould be the subject of further investigation in antimalarial che-motherapy.

ACKNOWLEDGMENTS

We gratefully acknowledge the statistical advice of Richard Parsons, Fac-ulty of Health Sciences, and the supervisory support of Andrew Crowe,School of Pharmacy, Curtin University. S.M.D.K.G.S. was the recipient ofa Curtin University Strategic International Research Scholarship.

REFERENCES1. World Health Organization. 2010. Guidelines for the treatment of ma-

laria. World Health Organization, Geneva, Switzerland.2. Johnson TN. 2008. The problems in scaling adult drug doses to children.

Arch. Dis. Child. 93:207–211. http://dx.doi.org/10.1136/adc.2006.114835.3. Mahmood I. 2006. Prediction of drug clearance in children from adults:

a comparison of several allometric methods. Br. J. Clin. Pharmacol. 61:545–557. http://dx.doi.org/10.1111/j.1365-2125.2006.02622.x.

4. Holford NH. 1996. A size standard for pharmacokinetics. Clin. Pharmaco-kinet. 30:329–332. http://dx.doi.org/10.2165/00003088-199630050-00001.

5. Cella M, Knibbe C, Danhof M, Della PO. 2010. What is the right dosefor children? Br. J. Clin. Pharmacol. 70:597– 603. http://dx.doi.org/10.1111/j.1365-2125.2009.03591.x.

6. Obua C, Hellgren U, Ntale M, Gustafsson LL, Ogwal-Okeng JW,Gordi T, Jerling M. 2008. Population pharmacokinetics of chloroquineand sulfadoxine and treatment response in children with malaria: sug-gestions for an improved dose regimen. Br. J. Clin. Pharmacol. 65:493–501. http://dx.doi.org/10.1111/j.1365-2125.2007.03050.x.

7. Barnes KI, Little F, Smith PJ, Evans A, Watkins WM, White NJ. 2006.Sulfadoxine-pyrimethamine pharmacokinetics in malaria: pediatricdosing implications. Clin. Pharmacol. Ther. 80:582–596. http://dx.doi.org/10.1016/j.clpt.2006.08.016.

8. Ursing J, Kofoed PE, Rodrigues A, Blessborn D, Thoft-Nielsen R,Bjorkman A, Rombo L. 2011. Similar efficacy and tolerability of double-dose chloroquine and artemether-lumefantrine for treatment of Plasmo-dium falciparum infection in Guinea-Bissau: a randomized trial. J. In-fect. Dis. 203:109 –116. http://dx.doi.org/10.1093/infdis/jiq001.

9. Ursing J, Kofoed PE, Rodrigues A, Bergqvist Y, Rombo L. 2009.Chloroquine is grossly overdosed and overused but well tolerated inGuinea-Bissau. Antimicrob. Agents Chemother. 53:180 –185. http://dx.doi.org/10.1128/AAC.01111-08.

10. Hendriksen IC, Maiga D, Lemnge MM, Mtove G, Gesase S, ReyburnH, Lindegardh N, Day NP, von Seidlein L, Dondorp AM, Tarning J,White NJ. 2013. Population pharmacokinetic and pharmacodynamicproperties of intramuscular quinine in Tanzanian children with severefalciparum malaria. Antimicrob. Agents Chemother. 57:775–783. http://dx.doi.org/10.1128/AAC.01349-12.

11. Price RN, Hasugian AR, Ratcliff A, Siswantoro H, Purba HL, Kenan-galem E, Lindegardh N, Penttinen P, Laihad F, Ebsworth EP, AnsteyNM, Tjitra E. 2007. Clinical and pharmacological determinants of thetherapeutic response to dihydroartemisinin-piperaquine for drug-resistant malaria. Antimicrob. Agents Chemother. 51:4090 – 4097. http://dx.doi.org/10.1128/AAC.00486-07.

12. Salman S, Page-Sharp M, Batty KT, Kose K, Griffin S, Siba PM, IlettKF, Mueller I, Davis TM. 2012. Pharmacokinetic comparison of twopiperaquine-containing artemisinin combination therapies in PapuaNew Guinean children with uncomplicated malaria. Antimicrob. AgentsChemother. 56:3288 –3297. http://dx.doi.org/10.1128/AAC.06232-11.

13. Tarning J, Zongo I, Some FA, Rouamba N, Parikh S, Rosenthal PJ,Hanpithakpong W, Jongrak N, Day NP, White NJ, Nosten F, Oue-draogo JB, Lindegardh N. 2012. Population pharmacokinetics andpharmacodynamics of piperaquine in children with uncomplicated fal-ciparum malaria. Clin. Pharmacol. Ther. 91:497–505. http://dx.doi.org/10.1038/clpt.2011.254.

14. Creek DJ, Bigira V, McCormack S, Arinaitwe E, Wanzira H, KakuruA, Tappero JW, Sandison TG, Lindegardh N, Nosten F, Aweeka FT,Parikh S. 2013. Pharmacokinetic predictors for recurrent malaria afterdihydroartemisinin-piperaquine treatment of uncomplicated malaria inUgandan infants. J. Infect. Dis. 207:1646 –1654. http://dx.doi.org/10.1093/infdis/jit078.

15. Hendriksen IC, Mtove G, Kent A, Gesase S, Reyburn H, Lemnge MM,Lindegardh N, Day NP, von Seidlein L, White NJ, Dondorp AM,Tarning J. 2013. Population pharmacokinetics of intramuscular artesu-nate in African children with severe malaria: implications for a practicaldosing regimen. Clin. Pharmacol. Ther. 93:443– 450. http://dx.doi.org/10.1038/clpt.2013.26.

16. Morris CA, Tan B, Duparc S, Borghini-Fuhrer I, Jung D, Shin CS,Fleckenstein L. 2013. Effects of body size and gender on the populationpharmacokinetics of artesunate and its active metabolite dihydroarte-misinin in pediatric malaria patients. Antimicrob. Agents Chemother.57:5889 –5900. http://dx.doi.org/10.1128/AAC.00635-13.

17. Boxenbaum H. 1982. Interspecies scaling, allometry, physiological time,and the ground plan of pharmacokinetics. J. Pharmacokinet. Biopharm.10:201–227. http://dx.doi.org/10.1007/BF01062336.

18. Mordenti J. 1986. Man versus beast: pharmacokinetic scaling inmammals. J. Pharm. Sci. 75:1028 –1040. http://dx.doi.org/10.1002/jps.2600751104.

19. Ings RM. 1990. Interspecies scaling and comparisons in drug develop-ment and toxicokinetics. Xenobiotica 20:1201–1231. http://dx.doi.org/10.3109/00498259009046839.

20. White CR, Seymour RS. 2005. Allometric scaling of mammalianmetabolism. J. Exp. Biol. 208:1611–1619. http://dx.doi.org/10.1242/jeb.01501.

Senarathna and Batty

6074 aac.asm.org Antimicrobial Agents and Chemotherapy

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

21. West GB. 2012. The importance of quantitative systemic thinkingin medicine. Lancet 379:1551–1559. http://dx.doi.org/10.1016/S0140-6736(12)60281-5.

22. Rowland M, Tozer TN. 2011. Clinical pharmacokinetics and pharma-codynamcs: concepts and applications. Lippincott Williams & Wilkins,Philadelphia, PA.

23. Riviere JE, Martin-Jimenez T, Sundlof SF, Craigmill AL. 1997. Inter-species allometric analysis of the comparative pharmacokinetics of 44drugs across veterinary and laboratory animal species. J. Vet. Pharmacol.Ther. 20:453– 463. http://dx.doi.org/10.1046/j.1365-2885.1997.00095.x.

24. Hu TM, Hayton WL. 2001. Allometric scaling of xenobiotic clearance:uncertainty versus universality. AAPS PharmSci 3:E29. http://dx.doi.org/10.1208/ps030429.

25. Cox SK. 2007. Allometric scaling of marbofloxacin, moxifloxacin, dano-floxacin and difloxacin pharmacokinetics: a retrospective analysis. J.Vet. Pharmacol. Ther. 30:381–386. http://dx.doi.org/10.1111/j.1365-2885.2007.00886.x.

26. Mahmood I. 2007. Application of allometric principles for the predic-tion of pharmacokinetics in human and veterinary drug development.Adv. Drug Deliv. Rev. 59:1177–1192. http://dx.doi.org/10.1016/j.addr.2007.05.015.

27. Mahmood I. 2010. Interspecies scaling for the prediction of drug clear-ance in children: application of maximum lifespan potential and an em-pirical correction factor. Clin. Pharmacokinet. 49:479 – 492. http://dx.doi.org/10.2165/11531830-000000000-00000.

28. Mahmood I, Martinez M, Hunter RP. 2006. Interspecies allometricscaling. Part I: prediction of clearance in large animals. J. Vet. Pharmacol.Ther. 29:415– 423. http://dx.doi.org/10.1111/j.1365-2885.2006.00786.x.

29. Knibbe CA, Zuideveld KP, Aarts LP, Kuks PF, Danhof M. 2005.Allometric relationships between the pharmacokinetics of propofol inrats, children and adults. Br. J. Clin. Pharmacol. 59:705–711. http://dx.doi.org/10.1111/j.1365-2125.2005.02239.x.

30. Peeters MY, Allegaert K, Blusse van Oud-Alblas HJ, Cella M, TibboelD, Danhof M, Knibbe CA. 2010. Prediction of propofol clearance inchildren from an allometric model developed in rats, children and adultsversus a 0.75 fixed-exponent allometric model. Clin. Pharmacokinet.49:269 –275. http://dx.doi.org/10.2165/11319350-000000000-00000.

31. Brocks DR, Toni JW. 1999. Pharmacokinetics of halofantrine in the rat:stereoselectivity and interspecies comparisons. Biopharm. Drug Dispos.20:165–169. http://dx.doi.org/10.1002/(SICI)1099-081X(199904)20:3�165::AID-BDD170�3.0.CO;2-Z.

32. Davis CB, Bambal R, Moorthy GS, Hugger E, Xiang H, Park BK,Shone AE, O’Neill PM, Ward SA. 2009. Comparative preclinical drugmetabolism and pharmacokinetic evaluation of novel 4-aminoquinolineanti-malarials. J. Pharm. Sci. 98:362–377. http://dx.doi.org/10.1002/jps.21469.

33. Moore BR, Page-Sharp M, Stoney JR, Ilett KF, Jago JD, Batty KT.2011. Pharmacokinetics, pharmacodynamics, and allometric scaling ofchloroquine in a murine malaria model. Antimicrob. Agents Che-mother. 55:3899 –3907. http://dx.doi.org/10.1128/AAC.00067-11.

34. Tod M, Jullien V, Pons G. 2008. Facilitation of drug evaluation inchildren by population methods and modelling. Clin. Pharmacokinet.47:231–243. http://dx.doi.org/10.2165/00003088-200847040-00002.

35. Anderson BJ, Holford NH. 2008. Mechanism-based concepts of size andmaturity in pharmacokinetics. Annu. Rev. Pharmacol. Toxicol. 48:303–332.http://dx.doi.org/10.1146/annurev.pharmtox.48.113006.094708.

36. Holford N, Heo YA, Anderson B. 2013. A pharmacokinetic standard forbabies and adults. J. Pharm. Sci. 102:2941–2952. http://dx.doi.org/10.1002/jps.23574.

37. Mahmood I. 2007. Prediction of drug clearance in children: impact ofallometric exponents, body weight, and age. Ther. Drug Monit. 29:271–278. http://dx.doi.org/10.1097/FTD.0b013e318042d3c4.

38. Mahmood I. 2010. Theoretical versus empirical allometry: facts behindtheories and application to pharmacokinetics. J. Pharm. Sci. 99:2927–2933. http://dx.doi.org/10.1002/jps.22073.

39. Tang H, Mayersohn M. 2011. Controversies in allometric scaling forpredicting human drug clearance: an historical problem and reflectionson what works and what does not. Curr. Top. Med. Chem. 11:340 –350.http://dx.doi.org/10.2174/156802611794480945.

40. McLeay SC, Morrish GA, Kirkpatrick CM, Green B. 2012. Therelationship between drug clearance and body size: systematic reviewand meta-analysis of the literature published from 2000 to 2007. Clin.

Pharmacokinet. 51:319 –330. http://dx.doi.org/10.2165/11598930-000000000-00000.

41. Riond JL, Riviere JE. 1990. Allometric analysis of doxycycline pharma-cokinetic parameters. J. Vet. Pharmacol. Ther. 13:404 – 407. http://dx.doi.org/10.1111/j.1365-2885.1990.tb00795.x.

42. Davies B, Morris T. 1993. Physiological parameters in laboratory ani-mals and humans. Pharm. Res. 10:1093–1095. http://dx.doi.org/10.1023/A:1018943613122.

43. Ritschel WA, Vachharajani NN, Johnson RD, Hussain AS. 1992. Theallometric approach for interspecies scaling of pharmacokinetic param-eters. Comp. Biochem. Physiol. C 103:249 –253. http://dx.doi.org/10.1016/0742-8413(92)90003-P.

44. Mahmood I, Balian JD. 1996. Interspecies scaling: predicting clear-ance of drugs in humans. Three different approaches. Xenobiotica26:887– 895.

45. Haddad S, Restieri C, Krishnan K. 2001. Characterization of age-relatedchanges in body weight and organ weights from birth to adolescence inhumans. J. Toxicol. Environ. Health A 64:453– 464. http://dx.doi.org/10.1080/152873901753215911.

46. West GB, Brown JH, Enquist BJ. 1997. A general model for the origin ofallometric scaling laws in biology. Science 276:122–126. http://dx.doi.org/10.1126/science.276.5309.122.

47. Tang H, Hussain A, Leal M, Fluhler E, Mayersohn M. 2011. Contro-versy in the allometric application of fixed- versus varying-exponentmodels: a statistical and mathematical perspective. J. Pharm. Sci. 100:402– 410. http://dx.doi.org/10.1002/jps.22316.

48. Bonate PL, Howard D. 2000. Prospective allometric scaling: does theemperor have clothes? J. Clin. Pharmacol. 40:335–340. http://dx.doi.org/10.1177/00912700022009017.

49. Lin JH. 1995. Species similarities and differences in pharmacokinetics.Drug Metab. Dispos. 23:1008 –1021.

50. Lin JH. 1998. Applications and limitations of interspecies scaling and invitro extrapolation in pharmacokinetics. Drug Metab. Dispos. 26:1202–1212.

51. Dinev TG. 2008. Comparison of the pharmacokinetics of five aminogly-coside and aminocyclitol antibiotics using allometric analysis in mam-mal and bird species. Res. Vet. Sci. 84:107–118. http://dx.doi.org/10.1016/j.rvsc.2007.04.008.

52. Davis TM. 2005. Mefloquine, p 1091–1104. In Yu VL, Edwards G, McK-innon PS, Peloquin C, Morse GD (ed), Antimicrobial therapy and vac-cines, vol. II. Antimicrobial agents. Williams & Wilkins, Baltimore, MD.

53. Davis TM, Hung TY, Sim IK, Karunajeewa HA, Ilett KF. 2005.Piperaquine: a resurgent antimalarial drug. Drugs 65:75– 87. http://dx.doi.org/10.2165/00003495-200565010-00004.

54. Krishna S, White NJ. 1996. Pharmacokinetics of quinine, chloroquineand amodiaquine: clinical implications. Clin. Pharmacokinet. 30:263–299. http://dx.doi.org/10.2165/00003088-199630040-00002.

55. Binh TQ, Ilett KF, Batty KT, Davis TM, Hung NC, Powell SM, ThuLT, Thien HV, Phuong HL, Phuong VDB. 2001. Oral bioavailability ofdihydroartemisinin in Vietnamese volunteers and in patients with falcip-arum malaria. Br. J. Clin. Pharmacol. 51:541–546. http://dx.doi.org/10.1046/j.1365-2125.2001.01395.x.

56. Hung TY, Davis TM, Ilett KF, Karunajeewa HA, Hewitt S, Denis MB,Lim C, Socheat D. 2004. Population pharmacokinetics of piperaquine inadults and children with uncomplicated falciparum or vivax malaria. Br.J. Clin. Pharmacol. 57:253–262. http://dx.doi.org/10.1046/j.1365-2125.2003.02004.x.

57. Karunajeewa HA, Ilett KF, Mueller I, Siba P, Law I, Page-Sharp M, LinE, Lammey J, Batty KT, Davis TM. 2008. Pharmacokinetics and efficacyof piperaquine and chloroquine in Melanesian children with uncompli-cated malaria. Antimicrob. Agents Chemother. 52:237–243. http://dx.doi.org/10.1128/AAC.00555-07.

58. Bennett K, Si Y, Steinbach T, Zhang J, Li Q. 2008. Pharmacokinetic andpharmacodynamic evaluation of intramuscular artesunate in healthybeagle dogs. Am. J. Trop. Med. Hyg. 79:36 – 41.

59. Li QG, Peggins JO, Fleckenstein LL, Masonic K, Heiffer MH, BrewerTG. 1998. The pharmacokinetics and bioavailability of dihydroartemis-inin, arteether, artemether, artesunic acid and artelinic acid in rats. J.Pharm. Pharmacol. 50:173–182.

60. Li Q, Cantilena LR, Leary KJ, Saviolakis GA, Miller RS, Melendez V,Weina PJ. 2009. Pharmacokinetic profiles of artesunate after single in-travenous doses at 0.5, 1, 2, 4, and 8 mg/kg in healthy volunteers: a phase

Allometric Scaling of Antimalarial Drugs

October 2014 Volume 58 Number 10 aac.asm.org 6075

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

I study. Am. J. Trop. Med. Hyg. 81:615– 621. http://dx.doi.org/10.4269/ajtmh.2009.09-0150.

61. Sinou V, Taudon N, Mosnier J, Aglioni C, Bressolle FM, Parzy D.2008. Pharmacokinetics of artesunate in the domestic pig. J. Antimicrob.Chemother. 62:566 –574. http://dx.doi.org/10.1093/jac/dkn231.

62. Miller RS, Li Q, Cantilena LR, Leary KJ, Saviolakis GA, Melendez V,Smith B, Weina PJ. 2012. Pharmacokinetic profiles of artesunate fol-lowing multiple intravenous doses of 2, 4, and 8 mg/kg in healthy volun-teers: phase 1b study. Malar. J. 11:255. http://dx.doi.org/10.1186/1475-2875-11-255.

63. Xie LH, Li Q, Zhang J, Weina PJ. 2009. Pharmacokinetics, tissuedistribution and mass balance of radiolabeled dihydroartemisinin inmale rats. Malar. J. 8:112. http://dx.doi.org/10.1186/1475-2875-8-112.

64. Zhao KC, Chen ZX, Lin BL, Guo XB, Li GQ, Song ZY. 1988. Studieson the phase 1 clinical pharmacokinetics of artesunate and artemether.Chin. J. Clin. Pharmacol. 4:76 – 81.

65. Karbwang J, Na-Bangchang K, Congpuong K, Molunto P, ThanavibulA. 1997. Pharmacokinetics and bioavailability of oral and intramuscularartemether. Eur. J. Clin. Pharmacol. 52:307–310. http://dx.doi.org/10.1007/s002280050295.

66. Zhao KC, Chen QM, Song ZY. 1986. Studies on the pharmacokineticsof qinghaosu and two of its active derivatives in dogs. Acta Pharm. Sin.21:736 –739.

67. Zeng YL, Zhang YD, Xu GY, Wang CG, Jiang JR. 1984. The pharma-cokinetics and bioavailability of o-methyldihydroartemisinine in therabbit. Acta Pharm. Sin. 19:81– 84.

68. Ashton M, Gordi T, Trinh NH, Nguyen VH, Nguyen DS, Nguyen TN,Dinh XH, Johansson M, Le DC. 1998. Artemisinin pharmacokinetics inhealthy adults after 250, 500 and 1000 mg single oral doses. Biopharm. DrugDispos.19:245–250.http://dx.doi.org/10.1002/(SICI)1099-081X(199805)19:4�245::AID-BDD99�3.0.CO;2-Z.

69. Ashton M, Hai TN, Sy ND, Huong DX, Van Huong N, Nieu NT, CongLD. 1998. Artemisinin pharmacokinetics is time-dependent during re-peated oral administration in healthy male adults. Drug Metab. Dispos.26:25–27.

70. Ashton M, Johansson L, Thornqvist AS, Svensson US. 1999. Quantitativein vivo and in vitro sex differences in artemisinin metabolism in rat. Xeno-biotica 29:195–204. http://dx.doi.org/10.1080/004982599238740.

71. Benakis A, Paris M, Loutan L, Plessas CT, Plessas ST. 1997. Pharma-cokinetics of artemisinin and artesunate after oral administration inhealthy volunteers. Am. J. Trop. Med. Hyg. 56:17–23.

72. Duc DD, de Vries PJ, Nguyen XK, Le Nguyen B, Kager PA, van BoxtelCJ. 1994. The pharmacokinetics of a single dose of artemisinin in healthyVietnamese subjects. Am. J. Trop. Med. Hyg. 51:785–790.

73. Hien TT, Hanpithakpong W, Truong NT, Dung NT, Toi PV, Farrar J,Lindegardh N, Tarning J, Ashton M. 2011. Orally formulated artemis-inin in healthy fasting Vietnamese male subjects: a randomized, four-sequence, open-label, pharmacokinetic crossover study. Clin. Ther. 33:644 – 654. http://dx.doi.org/10.1016/j.clinthera.2011.04.017.

74. Isacchi B, Arrigucci S, la Marca G, Bergonzi MC, Vannucchi MG,Novelli A, Bilia AR. 2011. Conventional and long-circulating liposomesof artemisinin: preparation, characterization, and pharmacokinetic pro-file in mice. J. Liposome Res. 21:237–244. http://dx.doi.org/10.3109/08982104.2010.539185.

75. Rath K, Taxis K, Walz G, Gleiter CH, Li SM, Heide L. 2004. Pharma-cokinetic study of artemisinin after oral intake of a traditional prepara-tion of Artemisia annua L. (annual wormwood). Am. J. Trop. Med. Hyg.70:128 –132.

76. Li L, Pabbisetty D, Carvalho P, Avery MA, Williamson JS, Avery BA.2008. Ultra-performance liquid chromatography-tandem mass spectro-metric method for the determination of artemisinin in rat serum and itsapplication in pharmacokinetics. J. Chromatogr. B Analyt. Technol.Biomed. Life Sci. 867:131–137. http://dx.doi.org/10.1016/j.jchromb.2008.01.057.

77. Dien TK, de Vries PJ, Khanh NX, Koopmans R, Binh LN, Duc DD,Kager PA, van Boxtel CJ. 1997. Effect of food intake on pharmacoki-netics of oral artemisinin in healthy Vietnamese subjects. Antimicrob.Agents Chemother. 41:1069 –1072.

78. Titulaer HA, Zuidema J, Kager PA, Wetsteyn JC, Lugt CB, MerkusFW. 1990. The pharmacokinetics of artemisinin after oral, intramuscu-lar and rectal administration to volunteers. J. Pharm. Pharmacol. 42:810 – 813. http://dx.doi.org/10.1111/j.2042-7158.1990.tb07030.x.

79. Zhang S, Hai TN, Ilett KF, Huong DX, Davis TM, Ashton M. 2001.

Multiple dose study of interactions between artesunate and artemisininin healthy volunteers. Br. J. Clin. Pharmacol. 52:377–385. http://dx.doi.org/10.1046/j.0306-5251.2001.01461.x.

80. Batzias GC, Delis GA, Athanasiou LV. 2005. Clindamycin bioavailabil-ity and pharmacokinetics following oral administration of clindamycinhydrochloride capsules in dogs. Vet. J. 170:339 –345. http://dx.doi.org/10.1016/j.tvjl.2004.06.007.

81. Budsberg SC, Kemp DT, Wolski N. 1992. Pharmacokinetics of clinda-mycin phosphate in dogs after single intravenous and intramuscular ad-ministrations. Am. J. Vet. Res. 53:2333–2336.

82. Flaherty JF, Rodondi LC, Guglielmo BJ, Fleishaker JC, Townsend RJ,Gambertoglio JG. 1988. Comparative pharmacokinetics and serum in-hibitory activity of clindamycin in different dosing regimens. Antimi-crob. Agents Chemother. 32:1825–1829. http://dx.doi.org/10.1128/AAC.32.12.1825.

83. Gatti G, Flaherty J, Bubp J, White J, Borin M, Gambertoglio J. 1993.Comparative study of bioavailabilities and pharmacokinetics of clinda-mycin in healthy volunteers and patients with AIDS. Antimicrob. AgentsChemother. 37:1137–1143. http://dx.doi.org/10.1128/AAC.37.5.1137.

84. Lavy E, Ziv G, Shem-Tov M, Glickman A, Dey A. 1999. Pharmacoki-netics of clindamycin HCl administered intravenously, intramuscularlyand subcutaneously to dogs. J. Vet. Pharmacol. Ther. 22:261–265. http://dx.doi.org/10.1046/j.1365-2885.1999.00221.x.

85. Plaisance KI, Drusano GL, Forrest A, Townsend RJ, Standiford HC.1989. Pharmacokinetic evaluation of two dosage regimens of clindamy-cin phosphate. Antimicrob. Agents Chemother. 33:618 – 620. http://dx.doi.org/10.1128/AAC.33.5.618.

86. Yang SH, Lee MG. 2007. Dose-independent pharmacokinetics of clin-damycin after intravenous and oral administration to rats: contributionof gastric first-pass effect to low bioavailability. Int. J. Pharm. 332:17–23.http://dx.doi.org/10.1016/j.ijpharm.2006.11.019.

87. Townsend RJ, Baker RP. 1987. Pharmacokinetic comparison of threeclindamycin phosphate dosing schedules. Drug Intell. Clin. Pharm. 21:279 –281.

88. Ahmed T, Sharma P, Gautam A, Varshney B, Kothari M, Ganguly S,Moehrle JJ, Paliwal J, Saha N, Batra V. 2008. Safety, tolerability, andsingle and multiple-dose pharmacokinetics of piperaquine phosphate inhealthy subjects. J. Clin. Pharmacol. 48:166 –175. http://dx.doi.org/10.1177/0091270007310384.

89. Batty KT, Moore BR, Stirling V, Ilett KF, Page-Sharp M, Shilkin KB,Mueller I, Karunajeewa HA, Davis TM. 2008. Toxicology and pharma-cokinetics of piperaquine in mice. Toxicology 249:55– 61. http://dx.doi.org/10.1016/j.tox.2008.04.004.

90. Nguyen TC, Nguyen NQ, Nguyen XT, Bui D, Travers T, Edstein MD.2008. Pharmacokinetics of the antimalarial drug piperaquine in healthyVietnamese subjects. Am. J. Trop. Med. Hyg. 79:620 – 623.

91. Sim IK, Davis TM, Ilett KF. 2005. Effects of a high-fat meal on therelative oral bioavailability of piperaquine. Antimicrob. Agents Che-mother. 49:2407–2411. http://dx.doi.org/10.1128/AAC.49.6.2407-2411.2005.

92. Tarning J, Lindegardh N, Sandberg S, Day NP, White NJ, Ashton M.2008. Pharmacokinetics and metabolism of the antimalarial piperaquineafter intravenous and oral single doses to the rat. J. Pharm. Sci. 97:3400 –3410. http://dx.doi.org/10.1002/jps.21226.

93. Baudry S, Pham YT, Baune B, Vidrequin S, Crevoisier C, Gimenez F,Farinotti R. 1997. Stereoselective passage of mefloquine through theblood-brain barrier in the rat. J. Pharm. Pharmacol. 49:1086 –1090. http://dx.doi.org/10.1111/j.2042-7158.1997.tb06047.x.

94. Boudreau EF, Fleckenstein L, Pang LW, Childs GE, Schroeder AC,Ratnaratorn B, Phintuyothin P. 1990. Mefloquine kinetics in cured andrecrudescent patients with acute falciparum malaria and in healthy vol-unteers. Clin. Pharmacol. Ther. 48:399 – 409. http://dx.doi.org/10.1038/clpt.1990.168.

95. Charles BG, Blomgren A, Nasveld PE, Kitchener SJ, Jensen A, GregoryRM, Robertson B, Harris IE, Reid MP, Edstein MD. 2007. Populationpharmacokinetics of mefloquine in military personnel for prophylaxisagainst malaria infection during field deployment. Eur. J. Clin. Pharma-col. 63:271–278. http://dx.doi.org/10.1007/s00228-006-0247-3.

96. Crevoisier C, Handschin J, Barre J, Roumenov D, Kleinbloesem C.1997. Food increases the bioavailability of mefloquine. Eur. J. Clin. Phar-macol. 53:135–139. http://dx.doi.org/10.1007/s002280050351.

97. Franssen G, Rouveix B, Lebras J, Bauchet J, Verdier F, Michon C,Bricaire F. 1989. Divided-dose kinetics of mefloquine in man. Br. J.

Senarathna and Batty

6076 aac.asm.org Antimicrobial Agents and Chemotherapy

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

Clin. Pharmacol. 28:179 –184. http://dx.doi.org/10.1111/j.1365-2125.1989.tb05413.x.

98. Gimenez F, Pennie RA, Koren G, Crevoisier C, Wainer IW, FarinottiR. 1994. Stereoselective pharmacokinetics of mefloquine in healthy Cau-casians after multiple doses. J. Pharm. Sci. 83:824 – 827. http://dx.doi.org/10.1002/jps.2600830613.

99. Juma FD, Ogeto JO. 1989. Mefloquine disposition in normals and in pa-tients with severe Plasmodium falciparum malaria. Eur. J. Drug Metab.Pharmacokinet. 14:15–17. http://dx.doi.org/10.1007/BF03190836.

100. Karbwang J, Back DJ, Bunnag D, Breckenridge AM. 1988. A compar-ison of the pharmacokinetics of mefloquine in healthy Thai volunteersand in Thai patients with falciparum malaria. Eur. J. Clin. Pharmacol.35:677– 680. http://dx.doi.org/10.1007/BF00637607.

101. Karbwang J, Bunnag D, Breckenridge AM, Back DJ. 1987. The phar-macokinetics of mefloquine when given alone or in combination withsulphadoxine and pyrimethamine in Thai male and female subjects. Eur.J. Clin. Pharmacol. 32:173–177. http://dx.doi.org/10.1007/BF00542191.

102. Karbwang J, Looareesuwan S, Back DJ, Migasana S, Bunnag D, Breck-enridge AM. 1988. Effect of oral contraceptive steroids on the clinicalcourse of malaria infection and on the pharmacokinetics of mefloquinein Thai women. Bull. World Health Organ. 66:763–767.

103. Karbwang J, Looareesuwan S, Phillips RE, Wattanagoon Y, MolyneuxME, Nagachinta B, Back DJ, Warrell DA. 1987. Plasma and wholeblood mefloquine concentrations during treatment of chloroquine-resistant falciparum malaria with the combination mefloquine-sulphadoxine-pyrimethamine. Br. J. Clin. Pharmacol. 23:477– 481. http://dx.doi.org/10.1111/j.1365-2125.1987.tb03079.x.

104. Khaliq Y, Gallicano K, Tisdale C, Carignan G, Cooper C, McCarthy A.2001. Pharmacokinetic interaction between mefloquine and ritonavir inhealthy volunteers. Br. J. Clin. Pharmacol. 51:591– 600. http://dx.doi.org/10.1046/j.1365-2125.2001.01393.x.

105. Kolawole JA, Mustapha A, Abudu-Aguye I, Ochekpe N. 2000. Meflo-quine pharmacokinetics in healthy subjects and in peptic ulcer patientsafter cimetidine administration. Eur. J. Drug Metab. Pharmacokinet. 25:165–170. http://dx.doi.org/10.1007/BF03192309.

106. Looareesuwan S, White NJ, Warrell DA, Forgo I, Dubach UG, RanalderUB, Schwartz DE. 1987. Studies of mefloquine bioavailability and kineticsusing a stable isotope technique: a comparison of Thai patients with falcip-arum malaria and healthy Caucasian volunteers. Br. J. Clin. Pharmacol. 24:37–42. http://dx.doi.org/10.1111/j.1365-2125.1987.tb03133.x.

107. Pennie RA, Koren G, Crevoisier C. 1993. Steady state pharmacokineticsof mefloquine in long-term travellers. Trans. R. Soc. Trop. Med. Hyg.87:459 – 462. http://dx.doi.org/10.1016/0035-9203(93)90036-P.

108. Ridtitid W, Wongnawa M, Mahatthanatrakul W, Raungsri N, Sunb-hanich M. 2005. Ketoconazole increases plasma concentrations of anti-malarial mefloquine in healthy human volunteers. J. Clin. Pharm. Ther.30:285–290. http://dx.doi.org/10.1111/j.1365-2710.2005.00651.x.

109. Ridtitid W, Wongnawa M, Mahatthanatrakul W, Chaipol P, Sunb-hanich M. 2000. Effect of rifampin on plasma concentrations of meflo-quine in healthy volunteers. J. Pharm. Pharmacol. 52:1265–1269. http://dx.doi.org/10.1211/0022357001777243.

110. Rozman RS, Molek NA, Koby R. 1978. The absorption, distribution,and excretion in mice of the antimalarial mefloquine, erythro-2,8-bis(trifluoromethyl)-alpha-(2-piperidyl)-4-quinolinemethanol hydro-chloride. Drug Metab. Dispos. 6:654 – 658.

111. Brum L, Leal MG, Uchoa FT, Kaiser M, Guterres SS, Costa TD. 2011.Determination of quinine and doxycycline in rat plasma by LC–MS–MS:application to a pharmacokinetic study. Chromatographia 73:1081–1088. http://dx.doi.org/10.1007/s10337-011-1949-y.

112. Claessen FA, van Boxtel CJ, Perenboom RM, Tange RA, Wetsteijn JC,Kager PA. 1998. Quinine pharmacokinetics: ototoxic and cardiotoxiceffects in healthy Caucasian subjects and in patients with falciparummalaria. Trop. Med. Int. Health 3:482– 489. http://dx.doi.org/10.1046/j.1365-3156.1998.00252.x.

113. Clohisy DR, Gibson TP. 1982. Comparison of pharmacokinetic parametersof intravenous quinidine and quinine in dogs. J. Cardiovasc. Pharmacol.4:107–110. http://dx.doi.org/10.1097/00005344-198201000-00017.

114. Karbwang J, Thanavibul A, Molunto P, Na BK. 1993. The pharmaco-kinetics of quinine in patients with hepatitis. Br. J. Clin. Pharmacol.35:444 – 446. http://dx.doi.org/10.1111/j.1365-2125.1993.tb04165.x.

115. Karbwang J, Davis TM, Looareesuwan S, Molunto P, Bunnag D,White NJ. 1993. A comparison of the pharmacokinetic and pharmaco-

dynamic properties of quinine and quinidine in healthy Thai males. Br. J.Clin. Pharmacol. 35:265–271.

116. Paintaud G, Alvan G, Ericsson O. 1993. The reproducibility of quininebioavailability. Br. J. Clin. Pharmacol. 35:305–307. http://dx.doi.org/10.1111/j.1365-2125.1993.tb05698.x.

117. Salako LA, Sowunmi A. 1992. Disposition of quinine in plasma, redblood cells and saliva after oral and intravenous administration tohealthy adult Africans. Eur. J. Clin. Pharmacol. 42:171–174. http://dx.doi.org/10.1007/BF00278479.

118. Pussard E, Barennes H, Daouda H, Clavier F, Sani AM, Osse M,Granic G, Verdier F. 1999. Quinine disposition in globally malnour-ished children with cerebral malaria. Clin. Pharmacol. Ther. 65:500 –510.http://dx.doi.org/10.1016/S0009-9236(99)70069-X.

119. Wanwimolruk S, Wong SM, Coville PF, Viriyayudhakorn S, Thitiar-chakul S. 1993. Cigarette smoking enhances the elimination of quinine.Br. J. Clin. Pharmacol. 36:610 – 614. http://dx.doi.org/10.1111/j.1365-2125.1993.tb00424.x.

120. Zhang H, Wong CW, Coville PF, Wanwimolruk S. 2000. Effect of thegrapefruit flavonoid naringin on pharmacokinetics of quinine in rats.Drug Metab. Drug Interact. 17:351–363.

121. Watari N, Wakamatsu A, Kaneniwa N. 1989. Comparison of disposi-tion parameters of quinidine and quinine in the rat. J. Pharmacobiodyn.12:608 – 615. http://dx.doi.org/10.1248/bpb1978.12.608.

122. White NJ, Chanthavanich P, Krishna S, Bunch C, Silamut K. 1983.Quinine disposition kinetics. Br. J. Clin. Pharmacol. 16:399 – 403. http://dx.doi.org/10.1111/j.1365-2125.1983.tb02184.x.

123. Zhang H, Ramsay N, Coville PF, Wanwimolruk S. 1999. Effect oferythromycin, rifampicin and isoniazid on the pharmacokinetics of qui-nine in rats. Pharm. Pharmacol. Commun. 5:467– 472. http://dx.doi.org/10.1211/146080899128735180.

124. Hasan MM, Hassan MA, Rawashdeh NM. 1990. Effect of oral activatedcharcoal on the pharmacokinetics of quinidine and quinine adminis-tered intravenously to rabbits. Pharmacol. Toxicol. 67:73–76. http://dx.doi.org/10.1111/j.1600-0773.1990.tb00785.x.

125. Batty KT, Thu LT, Davis TM, Ilett KF, Mai TX, Hung NC, Tien NP,Powell SM, Thien HV, Binh TQ, Kim NV. 1998. A pharmacokineticand pharmacodynamic study of intravenous vs oral artesunate in un-complicated falciparum malaria. Br. J. Clin. Pharmacol. 45:123–129.

126. Batty KT, Thu LT, Ilett KF, Tien NP, Powell SM, Hung NC, Mai TX,Chon VV, Thien HV, Binh TQ, Kim NV, Davis TM. 1998. A pharma-cokinetic and pharmacodynamic study of artesunate for vivax malaria.Am. J. Trop. Med. Hyg. 59:823– 827.

127. Byakika-Kibwika P, Lamorde M, Mayito J, Nabukeera L, Mayanja-Kizza H, Katabira E, Hanpithakpong W, Obua C, Pakker N, Linde-gardh N, Tarning J, de Vries PJ, Merry C. 2012. Pharmacokinetics andpharmacodynamics of intravenous artesunate during severe malariatreatment in Ugandan adults. Malar. J. 11:132. http://dx.doi.org/10.1186/1475-2875-11-132.

128. Davis TM, Phuong HL, Ilett KF, Hung NC, Batty KT, Phuong VDB,Powell SM, Thien HV, Binh TQ. 2001. Pharmacokinetics and pharma-codynamics of intravenous artesunate in severe falciparum malaria. An-timicrob. Agents Chemother. 45:181–186. http://dx.doi.org/10.1128/AAC.45.1.181-186.2001.

129. Ilett KF, Batty KT, Powell SM, Binh TQ, Thu LT, Phuong HL, HungNC, Davis TM. 2002. The pharmacokinetic properties of intramuscularartesunate and rectal dihydroartemisinin in uncomplicated falciparummalaria. Br. J. Clin. Pharmacol. 53:23–30. http://dx.doi.org/10.1046/j.0306-5251.2001.01519.x.

130. Newton P, Suputtamongkol Y, Teja-Isavadharm P, PukrittayakameeS, Navaratnam V, Bates I, White N. 2000. Antimalarial bioavailabilityand disposition of artesunate in acute falciparum malaria. Antimicrob.Agents Chemother. 44:972–977. http://dx.doi.org/10.1128/AAC.44.4.972-977.2000.

131. Newton PN, Barnes KI, Smith PJ, Evans AC, Chierakul W, Ruangveer-ayuth R, White NJ. 2006. The pharmacokinetics of intravenous artesu-nate in adults with severe falciparum malaria. Eur. J. Clin. Pharmacol.62:1003–1009. http://dx.doi.org/10.1007/s00228-006-0203-2.

132. Silamut K, Newton PN, Teja-Isavadharm P, Suputtamongkol Y, Siri-yanonda D, Rasameesoraj M, Pukrittayakamee S, White NJ. 2003.Artemether bioavailability after oral or intramuscular administration inuncomplicated falciparum malaria. Antimicrob. Agents Chemother. 47:3795–3798. http://dx.doi.org/10.1128/AAC.47.12.3795-3798.2003.

133. Karbwang J, Na-Bangchang K, Tin T, Sukontason K, Rimchala W,

Allometric Scaling of Antimalarial Drugs

October 2014 Volume 58 Number 10 aac.asm.org 6077

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from

Harinasuta. 1998. Pharmacokinetics of intramuscular artemether in pa-tients with severe falciparum malaria with or without acute renal failure.Br. J. Clin. Pharmacol. 45:597– 600. http://dx.doi.org/10.1046/j.1365-2125.1998.00723.x.

134. Hai TN, Hietala SF, Van HN, Ashton M. 2008. The influence of food onthe pharmacokinetics of piperaquine in healthy Vietnamese volunteers.Acta Trop. 107:145–149. http://dx.doi.org/10.1016/j.actatropica.2008.05.013.

135. Roshammar D, Hai TN, Hietala SF, Huong NV, Ashton M. 2006.Pharmacokinetics of piperaquine after repeated oral administrationof the antimalarial combination CV8 in 12 healthy male subjects. Eur. J.Clin. Pharmacol. 62:335–341. http://dx.doi.org/10.1007/s00228-005-0084-9.

136. Staehli Hodel EM, Guidi M, Zanolari B, Mercier T, Duong S, Kabany-wanyi AM, Ariey F, Buclin T, Beck HP, Decosterd LA, Olliaro P,Genton B, Csajka C. 2013. Population pharmacokinetics of mefloquine,piperaquine and artemether-lumefantrine in Cambodian and Tanzanianmalaria patients. Malar. J. 12:235. http://dx.doi.org/10.1186/1475-2875-12-235.

137. Tarning J, Rijken MJ, McGready R, Phyo AP, Hanpithakpong W, DayNP, White NJ, Nosten F, Lindegardh N. 2012. Population pharmaco-kinetics of dihydroartemisinin and piperaquine in pregnant and non-pregnant women with uncomplicated malaria. Antimicrob. Agents Che-mother. 56:1997–2007. http://dx.doi.org/10.1128/AAC.05756-11.

138. Gbotosho GO, Happi CT, Lawal O, Sijuade A, Sowunmi A, Oduola A.2012. A high performance liquid chromatographic assay of mefloquinein saliva after a single oral dose in healthy adult Africans. Malar. J. 11:59.http://dx.doi.org/10.1186/1475-2875-11-59.

139. Karbwang J, Na BK, Back DJ, Bunnag D. 1991. Effect of ampicillin onmefloquine pharmacokinetics in Thai males. Eur. J. Clin. Pharmacol.40:631– 633.

140. Abdelrahim II, Adam I, Elghazali G, Gustafsson LL, Elbashir MI,Mirghani RA. 2007. Pharmacokinetics of quinine and its metabolites inpregnant Sudanese women with uncomplicated Plasmodium falciparummalaria. J. Clin. Pharm. Ther. 32:15–19. http://dx.doi.org/10.1111/j.1365-2710.2007.00788.x.

141. Couet W, Laroche R, Floch JJ, Istin B, Fourtillan JB, Sauniere JF. 1991.Pharmacokinetics of quinine and doxycycline in patients with acute fal-ciparum malaria: a study in Africa. Ther. Drug Monit. 13:496 –501. http://dx.doi.org/10.1097/00007691-199111000-00005.

142. Davis TM, Supanaranond W, Pukrittayakamee S, Karbwang J, MoluntoP, Mekthon S, White NJ. 1990. A safe and effective consecutive-infusionregimen for rapid quinine loading in severe falciparum malaria. J. Infect. Dis.161:1305–1308. http://dx.doi.org/10.1093/infdis/161.6.1305.

143. Davis TM, White NJ, Looareesuwan S, Silamut K, Warrell DA. 1988.Quinine pharmacokinetics in cerebral malaria: predicted plasma con-centrations after rapid intravenous loading using a two-compartmentmodel. Trans. R. Soc. Trop. Med. Hyg. 82:542–547. http://dx.doi.org/10.1016/0035-9203(88)90498-1.

144. Pukrittayakamee S, Looareesuwan S, Keeratithakul D, Davis TM,

Teja-Isavadharm P, Nagachinta B, Weber A, Smith AL, Kyle D, WhiteNJ. 1997. A study of the factors affecting the metabolic clearance ofquinine in malaria. Eur. J. Clin. Pharmacol. 52:487– 493. http://dx.doi.org/10.1007/s002280050323.

145. White NJ, Looareesuwan S, Warrell DA, Warrell MJ, Bunnag D,Harinasuta T. 1982. Qunine pharmacokinetics and toxicity in cerebraland uncomplicated falciparum malaria. Am. J. Med. 73:564 –572. http://dx.doi.org/10.1016/0002-9343(82)90337-0.

146. Sabchareon A, Chongsuphajaisiddhi T, Attanath P. 1982. Serum qui-nine concentrations following the initial dose in children with falcipa-rum malaria. Southeast Asian J. Trop. Med. Public Health 13:556 –562.

147. Shann F, Stace J, Edstein M. 1985. Pharmacokinetics of quinine inchildren. J. Pediatr. 106:506 –510. http://dx.doi.org/10.1016/S0022-3476(85)80692-2.

148. Liu C, Zhang R, Hong X, Huang T, Mi S, Wang N. 2007. Pharmaco-kinetics of piperaquine after single and multiple oral administrations inhealthy volunteers. Yakugaku Zasshi 127:1709 –1714. http://dx.doi.org/10.1248/yakushi.127.1709.

149. Nealon C, Dzeing A, Muller-Romer U, Planche T, Sinou V, KombilaM, Kremsner PG, Parzy D, Krishna S. 2002. Intramuscular bioavail-ability and clinical efficacy of artesunate in gabonese children with severemalaria. Antimicrob. Agents Chemother. 46:3933–3939. http://dx.doi.org/10.1128/AAC.46.12.3933-3939.2002.

150. Mithwani S, Aarons L, Kokwaro GO, Majid O, Muchohi S, Edwards G,Mohamed S, Marsh K, Watkins W. 2004. Population pharmacokineticsof artemether and dihydroartemisinin following single intramusculardosing of artemether in African children with severe falciparum malaria.Br. J. Clin. Pharmacol. 57:146 –152. http://dx.doi.org/10.1046/j.1365-2125.2003.01986.x.

151. Bourahla A, Martin C, Gimenez F, Singhasivanon V, Attanath P,Sabchearon A, Chongsuphajaisiddhi T, Farinotti R. 1996. Stereoselec-tive pharmacokinetics of mefloquine in young children. Eur. J. Clin.Pharmacol. 50:241–244. http://dx.doi.org/10.1007/s002280050100.

152. Singhasivanon V, Chongsuphajaisiddhi T, Sabcharoen A, Attanath P,Webster HK, Wernsdorfer WH, Sheth UK, Djaja L, I. 1992. Pharmaco-kinetics of mefloquine in children aged 6 to 24 months. Eur. J. Drug Metab.Pharmacokinet. 17:275–279. http://dx.doi.org/10.1007/BF03190160.

153. Singhasivanon V, Chongsuphajaisiddhi T, Sabchareon A, Attanath P,Webster HK, Edstein MD, Lika ID. 1994. Pharmacokinetic study ofmefloquine in Thai children aged 5–12 years suffering from uncompli-cated falciparum malaria treated with MSP or MSP plus primaquine.Eur. J. Drug Metab. Pharmacokinet. 19:27–32. http://dx.doi.org/10.1007/BF03188819.

154. Tarning J, Ashley EA, Lindegardh N, Stepniewska K, Phaiphun L, DayNP, McGready R, Ashton M, Nosten F, White NJ. 2008. Populationpharmacokinetics of piperaquine after two different treatment regimenswith dihydroartemisinin-piperaquine in patients with Plasmodium fal-ciparum malaria in Thailand. Antimicrob. Agents Chemother. 52:1052–1061. http://dx.doi.org/10.1128/AAC.00955-07.

Senarathna and Batty

6078 aac.asm.org Antimicrobial Agents and Chemotherapy

on Septem

ber 15, 2014 by Curtin U

niversity Libraryhttp://aac.asm

.org/D

ownloaded from


Recommended