DETERMINANTS OF RESPONSES TO ANTIMALARIAL DRUGS IN CHILDREN WITH UNCOMPLICATED PLASMODIUM FALCIPARUM MALARIA BY RY SIJUADE Abayomi Olusola RA BSc (Chemistry) Ilorin, MSc (Pharmacology & TherapIeButics) Ibadan Matric No. 108845 L AN A thesis in the DAeDpartment of PHARMACOLOFG YI B AND THERAPEUTICS O Submitted to the IFTacYulty of Basic Medical Sciences in partial fulfillment S of the requirements for the degree of ER IV DOCTOR OF PHILOSOPHY N OF THE U UNIVERSITY OF IBADAN March 2011 1 ABSTRACT Drug resistance is a challenge to malaria control efforts and several factors including parasite genetics, host factors and pharmacokinetics may contribute to the selection of drug resistant Plasmodium falciparum. Understanding the role of these factors in patient response to antimalarial drugs is therefore essential in the management of malaria. The aim of the study was to determine the factors contributing to delay in malaria Parasite Clearance (PC) in children and evaluating the effects of pharmRacoYkinetic variability on treatment outcome. A Children (n=2,752), aged 6 months -12 years, with falcBiparRum malaria, were enrolled over a period of eight years and treated with stan dLardI doses of Chloroquine (CQ), Sulphadoxine-Pyrimethamine (SP) or AmodiaNquine (AQ) given alone or in combination with artemisinin. Each patient was folloAwed up for at least 14 days. Age, axillary temperature, parasite density and AgamDetocytaemia were assessed for their potential association with delay in P CI Band treatment outcomes. Filter-paper blood samples were collected from some Fof the children (n=148) before treatment and on days 1-7, 14, 28 and 35 after tOreatment for determination of CQ and sulphadoxine concentrations. In anIoTtheYr subset of patients (n=7), treated with amodiaquine, blood and saliva samples wSere collected over 35 days. High performance liquid chromatographic techniqueEs wRere used to determine concentrations of sulphadoxine in whole blood as well as AIQV and Desethyl amodiaquine (DEAQ) in saliva. Mean maximum drug concentration (NCmax), half-life (t1/2) and area under concentration-time curve (AUC0-28d) were assessed Ufor their association and predictive value for treatment outcomes. Data were analyzed using descriptive statistics, ANOVA, Chi-square, Students’ t-test, Kruskal-Wallis and multiple regressions at p = 0.05. Age ≤ 2 years (Adjusted odds ratio [AOR] = 2.13), presence of fever (AOR = 1.33) and parasitaemia > 50,000/µl (AOR = 2.21) at enrolment were independent risk 2 factors for delay in PC, while a body temperature >38OC and parasitaemia >20,000/µl were predictors a day after treatment regardless of the drug used. Day 3 concentration ≤ 1750ng/ml and AUC0-28d ≤ 950ng/ml.h were associated with chloroquine treatment failure. In a multivariate analysis, a terminal elimination t½ ≤ 220h (AOR = 0.28) and AUC0-28d ≤ 950ng/ml.h (AOR = 4.12) were identified as independent pharmacokinetic predictors of chloroquine treatment failure. Age stratified analysis showed that SD X concentrations were significantly higher in children > 5years compared tRo cYhildren <5years: Cmax; 295 vs 125µg/ml, AUC0-28d; 1562 vs 812µg/ml.d. InA patients who received AQ, there was a rapid conversion of AQ to DEAQ, whBichR was detectable in plasma and saliva within 40 minutes of administration. The mLeanI day 7 concentration of DEAQ was significantly higher in plasma than in salivaN (24 7.8 vs 125.1ng/ml). The t1/2 of DEAQ were similar in plasma (167.25±43.4h) andA saliva (146.12±17.2h). The decline phases of DEAQ in saliva concentration-timeA cuDrves were approximately similar to that in plasma. IB Delay in parasite clearance Fis specific and related to drug resistance. In addition pharmacokinetic variability o f Osulphadoxine in children has potential impact on dosage regimen and treatmenItT outYcome. S KeywordsE: RUncomplicated malaria, Pharmacokinetics, Antimalarials, PlasImVodium falciparum. WNord Count = 499 U 3 DEDICATION To the children who participated in the studies used for this thesis. RY A BR LI DA N IB A O F SI TY R VE UN I 4 ACKNOWLEDGMENT I express my profound gratitude to my supervisor, Dr. Grace O. Gbotosho for allowing me to draw from her wealth of experience and standing by me through her concerns as well as critical reading of my thesis. I also thank Professor A. Sowunmi for co-supervising this work. I appreciate his concerns and effort during the course of my programme. I appreciate Professor AMJ Oduola for his guidance and support. IR thaYnk the Head of Department, Dr. Catherine O. Falade for her support and RadvAise during the course of this programme. I thank Dr. T.C. Happi for his assistIanBce at all time. I must record my indebtedness to Dr O.A.T. Ogundahunsi for hLis support at all times. I sincerely appreciate the moral support of ProAfessNor O.O. Akinyinka and Dr. Olanrewaju Sowunmi. D To all my Lecturers in the FBacuAlty and all staff of the Department of Pharmacology and Therapeutics, FUni vIersity of Ibadan. My colleagues at the Malaria Research Laboratories have Obeen quite inspiring and they include: Drs. Onikepe Folarin, Folusho FalaTdeY, Oyindamola Abiodun, Sogo Olalubi, Micheal Obaro and Thomas AnyorigSiyaI. I thank Mrs Amoo and Adeola Alabi for running the clinic from which all theR children enrolled in the studies were recruited. I appreciate all staffs of PostIgVradu Eate Institute for Medical Research and Training, for accommodating me at tNhe Institute for carrying out all my research work. U I also appreciate the contribution of my dear ones, the SIJUADES: Femi & Kemi, Funmilola & Bola, Oyetoso & Moji, Olanrewaju & Seyi, Dupe & Akin and Opeyemi; who have been sources of encouragement, financial and moral support for me at all times. They have made the dream of completing this research a reality. Thank you for been a wonderful family. I thank God for being part of you. 5 I sincerely appreciate the support of my in-laws Dr. and Mrs I.O. Ayeni, Tunde & Funmilola Oni-Obasa, Dr & Mrs Ambi Rukewe, and kikelomo. Late Engr. Dare Ayeni for his priceless encouragement. To my friends; Olutunde, Remi, Tubosun, Lowo, Adelekan, Pastor Tunde Adisa, Soji Adesina and Ayo Olalekan, I appreciate you. I say big thank you to all the children who have participated in these studies. I appreciate the support I received from Pastors Oyedokun and Samu el. Thank you all. RY To my dear wife, Oluwatoyin for her unending love at all tiRmesA, patience and understanding. I appreciate my lovely children FiyinfOluwa, FoIgBofOluwa and Ibukun for their understanding during my absentness at home. L Chief and Deaconess S.A.O. Sijuade; DaAd aNnd Mum I appreciate you for standing by me and encouraging me at all timesD. Thank you. I acknowledge the University of BAdoA-Ekiti for the support and time to prepare parts of this thesis despite our busFy sc hIedule. Finally, I give all gl orOy and honour to the Most High God for this privilege and being my sustaineTr, pYrovider and for giving me Christ Jesus, my friend and lover of my soul. ThanSk yIou FATHER. The sRtudies were supported by grants from UNDP/World Bank/WHO/TDR to Dr. GIVrace E O. Gbotosho. UN 6 CERTIFICATION I certify that this work was carried out by Mr SIJUADE, Abayomi Olusola of the Department of Pharmacology and Therapeutics, University of Ibadan, Ibadan. Y AR RIB ……………………………………… …L……… Supervisor N Grace O. GboDtosAho B. Pharm (Hons) (Ife), MSc, PhD PharAmacology and Therapeutics (Ibadan) Sen iIoBr Lecturer DepartmenOt of FPharmacology and Therapeutics Y University of Ibadan IT Ibadan ER S IV UN 7 Table of Contents Page Title i Abstract ii Dedication iv Acknowledgements v Y Certification A vRii Table of Contents viii List of Tables B R xiii List of Figures L I xvii Glossary of Abbreviations N xx Chapter 1: INTRODUCTION DA 1 Objectives of the research A 5 IB Chapter 2: LITERATURE REVFIE W 6 2.0 Malaria the D isOease 6 2.1 Life cycleY of malaria 8 2.2 MalaIriTa control methods 10 2.3 CShemotherapy of malaria 11 R2.3.1 Classification of antimalarial drugs 12 E 2.3.2 Pharmacology of antimalarial drugs 14 IV2.4 Review of studies on pharmacokinetics of chloroquine, N sulphadoxine-pyrimethamine and amodiaquine 23 U 2.5 Antimalarial drug resistance 36 Chapter 3: MATERIALS AND METHODS Study 1: Studies on risk factors contributing to delay in parasite clearance in acute uncomplicated malaria. 3.0 Patients and drug treatment 40 8 3.1 Sample collection 40 3.2 Assessment of parasitaemia 42 3.3 Evaluation of response to treatment 42 Study 2: Pharmacokinetic determinants of response to treatment with chloroquine in children with acute uncomplicated Plasmodium falciparum malaria 44 3.4. Study Population 4R6 Y 3.5 Drug Analysis A46 3.5.1 Reagent for drug analysis R 46 3.5.2 Preparation of stock and working solutioInB 47 3.5.3 Analysis of chloroquine from filter p aLper samples 48 Study3: Development of High Performance LiNquid Chromatographic Analytical mDethAod for measurement of sulphadoxAine concentration in filter paper sample. 52 3.6 Analysis of sul pIhaBdoxine from filter paper samples 52 Study 4: Pharmacokinetic Fdisposition of sulphadoxine in children with acute un cOomplicated malaria treated with standard dose oTf suYlphadoxine-pyrimethamine. 56 Study 5: EvalIuation of the use of saliva for therapeutic drug RmSonitoring in children with uncomplicated Plasmodium E falciparum Malaria treated with amodiaquine-artesunate. IV 57 N 3.7 Patients enrolled in the study and sample collection 57 U 3.8 Analysis of amodiaquine or desethyl amodiaquine from plasma or saliva samples 58 3.9 Statistical analysis for all studies 60 9 Chapter 4: RESULTS Study 1: Risk factors contributing to delay in parasite clearance in children with acute uncompleted malaria. 63 4.0 Study population 63 4.1 Drug treatment and delay in parasite clearance 63 4.2 Risk factors for delay in parasite clearance at enrolment 69 4.3 Risk factors for delay in parasite clearance following initiation of treatment 6 9 4.4 Delay in parasite clearance and treatment failure AR Y69 Study 2: Pharmacokinetic determinants of response to trReatment with chloroquine in children with acute uncoImBplicated Plasmodium falciparum malaria. L 73 4.5 Study population N 78 4.6 Pharmacokinetic parameters of aDll pAatients enrolled in the study 78 4.7 Comparison of blood chloroqAuine concentrations and pharmacokinetic parameItBers in children with early or delay in parasite clearance. 81 4.8 Comparison of phaFrmacokinetic parameters in children with or without pre tOreatment chloroquine concentration. 84 4.9 CompaTrisYon of clinical parameters and treatment outcome in childIren with or without pre-treatment chloroquine concentration. 87 4.10 RCSomparison of blood chloroquine concentration in children E with sensitive or resistance responses to chloroquine. 87 IV4.11 Comparison of pharmacokinetic parameters in children N whose infection responded or failed to respond to U chloroquine treatment. 90 4.12 Pharmacokinetic risk factors associated with chloroquine treatment failure. 90 Study 3: Development of a simple cost effective high performance liquid chromatography assay of sulphadoxine in whole blood spotted on filter paper for field studies. 96 4.13 Chromatographic techniques 96 4.14 Recovery, calibration curves and reproducibility 99 10 4.15 Interference 102 4.16 Clinical application 102 Study 4: Pharmacokinetic disposition of sulphadoxine in children with acute uncomplicated malaria treated with standard dose of sulphadoxine-pyrimethamine. 105 4.17 Study population 105 4.18 Pharmacokinetic parameters of sulphadoxine in children with uncomplicated falciparum malaria after administration of staRndaYrd dose of sulphadoxine-pyrimethamine. A 107 4.19 Comparison of blood sulphadoxine concentration andR pharmacokinetic parameters in children with earIlyB parasite clearance or delay in parasite clearance. L 109 4.20 Comparison of disposition of sulphadoxNine in children with and without detectable blood levels of suAlphadoxine at enrolment before treatment. A D 112 4.21 Pharmacokinetic of 61 children without pretreatment sulphadoxine at enrolm eInBt. 115 4.22 Comparison of blooFd sulphadoxine concentrations in children with sensitive aOnd resistant responses to sulphadoxine- pyrimeTthaYmine 117 4.23 CompIarison of disposition of sulphadoxine in children RwShose infection responded or failed to respond to standard E dose of sulphadoxine-pyrimethamine. 117 IV4.24 Relationship between age and sulphadoxine pharmacokinetic N parameters in children treated with standard oral dose U of sulphadoxine-pyrimethamine. 121 Study 5: Evaluation of the use of saliva for therapeutic drug monitoring in Children with Uncomplicated Plasmodium falciparum malaria treated with amodiaquine. 4.25 Patients enrolled in the study 130 4.26 Pharmacokinetic parameters of desethyl amodiaquine in children with uncomplicated falciparum malaria after administration of standard doses of 11 artesunate-amodiaquine. 132 Chapter 5: DISCUSSION 139 CONCLUSION AND RECOMMENDATION 154 REFERENCES 155 APPENDICES 174 RY RA LI B DA N BA F I Y O IT RS IV E UN 12 List of Tables Table Title Page 2.1 Classification of antimalarial drugs. 13 3.1 Antimalrial drugs and their treatment regimens with time of 41 study administered to children with acute uncomplicated malaria 3.2 Evaluation of clinical response of children to treatment with 43 antimalarial drugs Y 4.1 Baseline clinical and parasitological parameters of 2752 children R65 with acute uncomplicated falciparum malaria enrolled Rin tAhe study. 4.2 Proportion of children with delay in parasite clearancIe Bfollowing 66 treatment with standard doses of selected antimal arLial drugs. 4.3 Proportion of children with falciparum malNaria who failed to 67 respond to treatment with standard doses of antimalarial drugs. 4.4 Predictors of delay in parasite clearanAce at presentation in 70 children with acute falciparum malaria D 4.5 Predictors of delay in parasiteI Bclea Arance on day 1 after treatment 71 in children with acute falcip arum malaria. 4.6 Percentage recovery of Fchloroquine from whole blood spotted 75 on filter paper using Othe analytical method for chloroquine. 4.7 Precision ofT anYalytical method for extraction of chloroquine 76 from wholeI blood spotted on filter paper. 4.8 BasRelinSe clinical and demographic characteristics of seventy 79 four children with acute uncomplicated malaria treated with Estandard doses of chloroquine. 4.9 IV Pharmacokinetic parameters of chloroquine determined in 80 N children with falciparum malaria and treated with standard doses U of chloroquine. 4.10 Pharmacokinetic parameters of chloroquine in children with 83 acute uncomplicated malaria who had delay in parasite clearance after treatment with standard doses of chloroquine. 4.11 Pharmacokinetic parameters of chloroquine in children with and 86 without pretreatment chloroquine levels following treatment with standard doses of chloroquine (n=74). 4.12 Comparison of clinical parameters and treatment outcome in 88 13 children with acute uncomplicated malaria who had or without pretreatment chloroquine concentration (n=74). 4.13 Whole blood chloroquine concentrations in children with 89 uncomplicated falciparum malaria with sensitive or resistant response after administration of 25mg/kg chloroquine base (n=74). 4.14 Pharmacokinetic parameters of chloroquine determined in 91 children with acute uncomplicated falciparum malaria who had sensitive or resistant responses after treatment with standard doses of chloroquine (n=74). Y 4.15 Pharmacokinetic predictors of treatment failure in children wAith R93 uncomplicated malaria treated with standard doses of chloroquine. R 4.16 Pharmacokinetic risk factors of treatment failure in chIilBdren with 94 uncomplicated malaria treated with comp leLte doses of chloroquine (when considering P value 0.3). N 4.17 Percentage recovery of sulphadoxine fromA whole blood samples 100 collected on filter paper. D 4.18 Precision of analytical method foAr sulphadoxine determination 101 using whole blood spotted o nI fBilter paper. 4.19 Demographic and pharmFacokinetic parameters of a patient who 104 received a standard dOose of sulphadoxine-pyrimethamine. 4.20 Baseline cliTnicYal and demographic characteristics of children 106 with acuteI uncomplicated falciparum malaria treated with standarSd dose of sulphadoxine-pyrimethamine (n=74). 4.21 PhaRrmacokinetic parameters of sulphadoxine in seventy-four 108 children treated with standard dose of sulphadoxine-Epyrimethamine. IV 4.22 Comparison of pharmacokinetic disposition of sulphadoxine in 111 N children with and without delay in parasite clearance. U 4.23 Pharmacokinetic parameters of sulphadoxine in children with 113 and without sulphadoxine in pretreatment whole blood samples. 4.24 Clinical and laboratory data at enrolment of children with acute, 116 uncomplicated falciparum malaria treated with standard dose of sulphadoxine-pyrimethamine without pre-treatment sulphadoxine levels. 4.25 Whole blood sulphadoxine concentration in children with 118 uncomplicated falciparum malaria with sensitive or resistant 14 response after oral administration of a standard dose of sulphadoxine-pyrimethamine. 4.26 Pharmacokinetic parameters of sulphadoxine in children without 119 pre-treatment sulphadoxine with uncomplicated falciparum malaria who had sensitive or resistant responses to oral administration of sulphadoxine-pyrimethamine. 4.27 Whole blood sulphadoxine concentration in children with 123 uncomplicated falciparum malaria with sensitive or resistant responses after administration of standard oral dose sulphadoxine-pyrimethamine. Y 4.28 Relationship between age and pharmacokinetic parameters of R124 sulphadoxine in children with uncomplicated falciparum malaAria treated with standard sulphadoxine-pyrimethamine. R 4.29 Baseline clinical and demographic characteristics Lof IsBeven with 131 acute uncomplicated falciparum malaria enrolled in the study. 4.30 Saliva-plasma ratio of desethyl amodiaqAuinNe concentrations in 134 children with falciparum malaria treaDted with standard doses of artesunate-amodiaquine. 4.31 Pharmacokinetic disposition IofB de Asethyl amodiaquine in plasma 136 after standard oral doses o f artesunate-amodiaquine (30mg/kg amodiaquine) in five chilFdren with uncomplicated malaria. 4.32 Pharmacokinetic d isOposition of desethyl amodiaquine in saliva 137 after standard oYral doses of artesunate-amodiaquine (30mg/kg amodiaquine) in five children with uncomplicated malaria 4.33 Comparison of pharmacokinetic parameters of desethyl 138 amodiaSquinIe T after standard oral doses of artesunate- amoRdiaquine (30mg/kg amodiaquine) in children with uncomplicated malaria. E 4.aI VStandard curve table for extrapolation of unknown concentration of chloroquine in whole blood on filter paper. 176 UN4.b Standard curve table for extrapolation of unknown concentration of sulphadoxine in whole blood on filter paper. 177 4.c Standard curve table for extrapolation of unknown concentration of desethyl amodiaquine in plasma 178 4.d Standard curve table for extrapolation of unknown concentration of desethyl amodiaquine in saliva. 179 15 LIST OF FIGURES Figure Legend Page 2.1 Global distribution of malaria. 8 2.2 Plasmodium SPP life cycle. 9 2.3 Chemical structure of Quinine 15 2.4 Chemical structure of Mefloquine 17 2.5 Chemical structure of Halofantrine 1Y9 2.6 Chemical structure of Artemisinin derivatives AR22 2.7 Chemical structure of Chloroquine R 23 2.8 Mode of action of Chloroquine. LIB 25 2.9 Chemical structure of Sulphadoxine. N 29 2.10 Mode of action of Sulphadoxine-pyrimethAamine. 30 2.11 Chemical structure of PyrimethamAine D 32 2.12 Structures of amodiaquine a nId BN-desethyl amodiaquine 34 4.1 Numbers of children wiFth delay in parasite clearance following 65 treatment with antim Oalarial drugs. 4.2 The proportTionYs of children treated with various antimalarials 69 who had dIelay in parasite clearance (PC) and subsequently failed treatme Snt 4.3 StanRdard curve of chloroquine for extrapolation of unknown 74 Econcentrations from whole blood spotted on filter paper. 4.4 IVChromatogram showing chloroquine and desethyl chloroquine 77 N from whole blood sample collected from collected from a patient U and spotted on filter paper on day 0 and day 3. 4.5 Mean whole blood concentration-time curves of chloroquine 82 following oral administration of 25mg/kg of chloroquine base over 3 days in children infected with Plasmodium falciparum malaria with early or delay in parasite clearance (PC). 4.6 Mean whole blood concentration-time curves of chloroquine in 85 children infected with Plasmodium falciparum malaria treated with oral doses of 25mg/kg chloroquine base over 3 days with or without chloroquine in their blood at enrolment. 16 4.7 Mean concentration-time curve for all patients, those who 92 responded to treatment and those who failed to respond to oral dose of chloroquine base given over 3 days. 4.8 Decline of chloroquine concentration in children with or without 95 chloroquine in their pre-treatment whole blood samples following oral administration of chloroquine base over 3 days. 4.9 Sulphadoxine standard curve for extrapolation of sulphadoxine 97 concentrations from unknown blood sample spotted on filter paper. Y 4.10 Chromatograms showing peaks after extraction of sulphadoxineR 98 and internal standard from drug free whole blood spiked wAith 60µg/ml sulphadoxine, day 0 and day 3 of sample collecteRd from a patient who was administered with a standaIrdB dose of sulphadoxine/pyrimethamine. L 4.11 Concentration – time curve for sulphadoxine in w hole blood after 103 single oral dose of sulphadoxine-pyrimethamNine (25mg/kg body weight of sulphadoxine and 1.25mgA/kg body weight of pyrimethamine. D 4.12 Mean whole blood concentratiBon-AI time curves of sulphadoxine in 110 children infected with Plasm odium falciparum malaria with early or delay in parasite clearance. F 4.13 Mean whole blood SODX concentration - time curves in children 114 with PlasmodYium falciparum malaria with or without sulphadoxinTe in their blood at enrolment. 4.14 Mean wShoIle blood concentration-time profile of sulphadoxine in 120 chilRdren whose infection responded and whose infection failed to responded to treatment after oral administration of standard dose Eof sulphadoxine-pyrimethamine. 4.15I V Mean concentration-time curve of sulphadoxine in capillary 125 N whole blood of children younger than 5 years or older than 5 U years after oral administration of standard dose of sulphadoxine-pyrimethamine. 4.16 Standard curve of desethyl amodiaquine for extrapolation of 127 unknown concentrations of DEAQ in plasma. 4.17 Standard curve of desethyl amodiaquine for extrapolation of 128 unknown concentrations of DEAQ in saliva. 4.18 Chromatograms showing amodiaquine (AQ) and 129 desethylamodiaquine (DEAQ) in plasma and saliva of a patient at 17 4 h after the first dose of oral artesunate-amodiaquine. 4.19 Plasma and Saliva log concentration - time curves of 135 desethylamodiaquine in children infected with P. falciparum malaria who were treated with oral doses of artesunate- amodiaquine. AR Y R LI B AN BA D F I Y O T RS I E IV UN 18 Glossary of Abbreviations ACPR Adequate clinical response ADP Adenosine diphosphate AOR Adjusted odd ratio ATP Adenosine triphosphate AL Artemether plus lumefantrine Y AMQ Mefloquine plus artesunate AR ANOVA Analysis of variance R AOR Adjusted odd ratio LIB AQ Amodiaquine N AQAS Artesunate plus amodiaquine A AQPS Amodiaquine plus sulphadoxAineD-pyrimethamine AQSP amodiaquine plus sulfa lIenBe-pyrimethamine AS Artesunate F AUC0-28d Area undeYr co n Ocentration time curve from 0 to 28 day CI ConfIidTence interval Cl RCSlearance Cmax E maximum concentration COTI V Co-trimozaxole UCNQ Chloroquine CQCP chloroquine plus chlorpheniramine CQKET Chloroquine plus ketotifen CQPS Chloroquine plus sulphadoxine-pyrimethamine d Day DCQ Desethyl chloroquine 19 DEAQ Desethyl amodiaquine df degree of freedom DHA Dyhydro artemisinin DHPS Dihydropteroate synthase DHFR Dihyrdofolate reductase DNA Deoxyribonucleic acid dUMP deoxyuridine monophosphate RY dUTP deoxyuridine triphosphate RA ETF Early treatment failure IB F female L Fig. Figure AN-3 gcm Gramm per centimetre cube D -3 gdm Gramm per decimetre cubBe A GM Geometric mean F I GTP Glutathione r edOuctase h hour Y Hb HSaemIo Tglobin HCl RHydrochloric acid HPLCV EI High performance liquid chromatography INPT Intermittent preventive treatment UIS Internal standard ITNs Insecticide treated nets Kg Kilograms L Litre LTF Late treatment failure 20 M Molarity MDR Multi drug resistance ml millilitre min minutes MQ Mefloquine mV Millivolt n Sample size RY NaH2PO4.2H20 Sodium dihydrogen phosphate RA NaOH Sodium hydroxide IB ng/ml Nano gram per Millilitre L o C Degree Celsius AN OR Odds ratio D pABA Paraaminobenzoic acid BA PCT Parasite clearance Ftim eI PYR Pyrimethamin eO alone PPV PapavTerinYe RBM RSoll BIack Malaria SP R Sulphadoxine-pyrimethamine SPPBV EI Sulphadoxine-pyrimethamine plus probenecid SND Standard deviation USDX sulphadoxine alone t1/2 Half-life tmax Time at maximum concentration V/V/V Volume/volume/volume VS Versus 21 WHO World Health Organization y Year χ2 Chi square Y AR LIB R N AD A IB OF ITY S VE R I UN 22 CHAPTER ONE INTRODUCTION Significance of the project Among several tropical diseases that affect humans, malaria poses Yspec ial control problems due to increasing population at risk from the disease. In 2R008, there were approximately 232 million malaria cases and 841,000 malaria RdeatAhs, with close to 90% of these occurring in sub-Sahara Africa (WHO 2009).I BNigeria is the largest population at risk in Africa. Being a serious disease th aLt mostly affects children under the age of 5 year, it is responsible for 25% Aof iNnfant mortality and 30% of all childhood deaths in Nigeria (FMOH 2004). DGlobally, it causes 85% of death in children under 5years of age (GlobalI MBal Aaria, 2008). It is a significant cause of morbidity accounting for about 6F5% of all clinic attendance in the country (FMH 2005). Nigeria is the first aOmong the top-five countries for malaria death (Global Malaria, 2008). In tThe Yabsence of an effective antimalarial vaccine, chemotherapy remains the maiSnstaIy for control of the disease. However, the malaria parasite has developed reRsistance to nearly all the classes of antimalarials including the promising artemIVisin Ein derivatives (Dondorp et al., 2009). This could be as a result of wNidespread and indiscriminate use of antimalarials, which places a strong selective Upressure on malaria parasites to develop high levels of resistance (Olumese 2005; WHO 2006). The reports of antimalarial drug resistance in disease endemic countries, especially Africa (Sowunmi & Salako 1992; Falade et al., 1997; Wang et al., 1997; Sibley 2001; Sowunmi et al., 2004; Talisuna et al., 2004; Pitmang et al., 2005) had 23 made the WHO to recommend Artemisinin based combination therapy (ACTs) for the treatment of acute uncomplicated malaria. Between 2002 and 2005 many African countries including Nigeria adopted the use of ACTs (FMH 2004). Unfortunately, ACTs are expensive for poor people living in disease endemic countries and require longer dosage regimen which pose challenges in their use. In addition, the emergence and spread of resistance to these front-line antimalarials have necessita ted the need to continually field-monitor parasite susceptibility to coRnveYntional antimalarial drugs such as chloroquine (CQ) and sulphadoxinRe-pAyrimethamine (SDX) in order to revisit them for the treatment of acute uInBcomplicated malaria (Nkoma et al., 2007; Dondorp et al., 2009). For instan ceL, in Malawi chloroquine regained it efficacy for treatment of malaria 12 yeaArs aNfter it was withdrawn from use, suggesting that CQ might once again be considDered for treatment of malaria (Laufer et al., 2006). BA Indeed, it is very importanFt to Idetermine factors that can be observed prior to treatment that can lead to tre aOtment failure especially in children. In this perspective, it would clearly help Tif aYll, or, at least, most children at risk of drug treatment failure could be identifSied Isoon after they become ill. This would allow the planning of alternative, mRore effective treatment strategies and thus the reduction of malaria- attriIbuVtab Ele morbidity and mortality and slowing of the spread of drug resistance. N A fundamental component of the strategy for the control of malaria disease Uis based on prompt and effective treatment. In vivo tests are much more direct measurement of treatment efficacy in a target population. The assessment of in-vivo therapeutic efficacy involves clinical and parasitological outcomes of treatment over a certain period following the start of treatment in order to check for the reappearance of parasites in the blood. A significant proportion of treatment failures do not appear 24 until after day 28 in areas of low or high transmission (WHO 2006). However, it is not often clear if patients have adequate drug blood levels at the time of recrudescence of infection and this can be determined by monitoring of blood concentration after treatment which can also be used to predict treatment outcome (White et al., 2008). Pharmacokinetics can also play a major role in the development of drug resistant malaria. It has been demonstrated t hat pharmacokinetics of drugs can be altered during long time use (BousquRet, Y1970). Chloroquine and sulphadoxine-pyrimethamine have been common RdruAgs used in the treatment of malaria. The long-time use of CQ and SP might hIaBve caused changes in their kinetic disposition which may result in selective dru gL pressure (Gardella et al., 2008; Hodel et al., 2010). Thus, the current studAy wNas designed to re-assess drug resistance level and determine whether drug Dtreatment failures observed during a standard in-vivo test are related to BparAasite factors or whether they are the consequence of poor metabolism oFf C QI and SP. Although, chroma toOgraphic techniques have been developed to allow accurate and sensitiveT deYtermination of the level of most antimalarial drugs in blood (Karbwang et alS., 19I87; Walker et al., 1983; Walker & Ademowo 1996; Babalola et al., 2003; MRinzi et al., 2005; Dua et al., 2007) these techniques are quite expensive, timeI cVons Euming and require the collection and use of venous blood. The sampling tNechniques require expertise, are invasive and sampling may not be easy in children Uduring field studies. These techniques may not be very practicable in analytical laboratories in low resource areas where malaria is endemic. Efforts in this project were devoted to the optimization and development of simple and cost effective chromatographic techniques for measurement of CQ and sulphadoxine in micro blood samples collected on filter paper. The newly developed method is 25 sensitive and requires less sample which are easy to transport. This new method of analysis will provide a unique opportunity to facilitate SDX blood level determination in patients and can also be employed for therapeutic drug monitoring during Intermittent Preventive Treatment for malaria control in pregnant (IPTp) women or children (IPTc) using sulphadoxine-pyrimethamine. Besides, the determination of antimalarial drug levels has been estimated in general from whole blood, plasma or red cell (White 1992; Salako and RSowYunmi, 1992; Gbotosho et al., 2009; Obua et al., 2008). However, antimRalarAials have also been estimated in saliva, for example, quinine (Salako aInBd Sowunmi, 1992). Although, there is much difficulty in measuring artem isLinin drugs in biological samples, it is often easier to measure their paArtneNr drugs, e.g. amodiaquine or sulphadoxine plus pyrimethamine (Gitau et al.,D 2004; Gbotosho et al., 2009; Obua et al., 2008). An ideal medium from wIBhich A antimalarial drugs should be measured should be non-invasive with resFpec t to sample collection; saliva is one of such medium (Salako and Sowun mOi 1992; Wilson et al., 1993). Despite increaTsinYg drug treatment failure, there are no clear guidelines, at least in Nigeria, SaboIut the time to change antimalarial drug treatment if parasites do not clear quiRckly from peripheral blood following treatment of uncomplicated acute infecItVions E in African children. Effort in this thesis was devoted to understanding the cNlinical factors that can lead to antimalarial treatment failure and the pharmacokinetic Ubasis of treatment failure in children with acute uncomplicated P. falciparum malaria in an area of intense malaria transmission in Nigeria. Objectives of the research: 1. To determine the relationship between delay in parasite clearance and 26 antimalarial treatment failure in children with falciparum malaria. 2. To determine the pharmacokinetic risk factors and the effects of pharmacokinetic variability on treatment outcome in children with acute uncomplicated falciparum malaria treated with chloroquine. 3. To develop and evaluate a simple, cost effective and sensitive method for quantification of sulphadoxine in small capillary samples of wholeY blo od spotted on filter paper. R 4. To assess the field applicability of the developed method foRr sulAphadoxine: to study the pharmacokinetic disposition of sulphadoxine IaBnd evaluate the effect of pharmacokinetic variability on therapeutic effica cLy in children. 5. To evaluate the use of saliva for therapeAutiNc drug monitoring in patients treated with amodiaquine-artesunate comDbination. IB A O F ITYS VE R NIU 27 CHAPTER TWO LTERATURE REVIEW 2.0 Malaria the disease Malaria, the most prevalent and most pernicious disease of human, remaYins up till now a major endemic parasitic disease and a leading cause of mAorRbidity and mortality especially in sub-Sahara Africa (WHO 2006). In thBe wRorld as a whole, malaria has been a major disease of mankind for thousa nLds Iof years. The global morbidity and mortality due to malaria have not signiNficantly changed over the past 50 years (Greenwood, 2004). World HealtDh OArganization (WHO) reported an increase in malaria clinical cases from 273 Amillions in 1998 to 515 millions cases in 2002 (Snow et al., 2005). IB F O SI TY ER NI V U Figure 2.1: Global distribution of Malaria in 2003 (http://rbm.who.int/wmr2005/html/map1.htm, Adopted from World Malaria Report 2005). 28 Malaria is a hematoprotozoan parasitic infection transmitted by species of anopheles mosquitoes (White 2004). It is currently endemic in 90 countries of the world of which a substantial population is from Africa, South of Sahara (Figure 2.1) (Kondrachie & Trigg, 1997; Olumese, 2005). Malaria accounts for one in five of all childhood deaths in Africa (Kager, 2002; RBM, 2006). The disease causes over 1 million deaths, 75% of which occur in African children < 5 years. In Nigeria, mala ria accounts for 63% of the diseases reported in health care centres and preRvaleYnce of malaria among pregnant women is 48% (FMOH, 2004). Almost USR$ 3A.5 million was reported by the Nigerian government for funding of malaria coInBtrol in 2003, with an additional US$ 2.3 million from other sources (RBM, 2005 )L. Non-immune travellers visiting malaria enAdemNic areas are at risk of malaria infection because of lack of immunity which deDvelops after repeated infection (Hviid, 2005; Stevenson & Zavala, 2006). PreIgnBant A women especially primigravidae are also at risk of malaria infection (WhittFy e t al., 2005; WHO, 2003; Adam et al., 2005) due to reduced immunity in preg nOancy and the presence of the plancenta which offer a highly supportive grTowYth environment for the parasite (Serghides et al., 2001). Malaria in pregSnanIcy leads to maternal anaemia (Miaffo et al., 2004), low birth weight and Rpremature delivery, which are associated with an increased risk of neonIaVtal Edeath and impaired cognitive development. Economically, the impact of mNalaria is alarming and contributes to individual, community and country poverty Uthrough lost labour days and expenses incurred for treatment and prevention. The disease impairs physical and mental development in children (Kihara et al., 2006). 2.1 Life Cycle of Plasmodium There are various species of the genus plasmodium responsible for the disease 29 in man which includes P. falciparum, P. vivax, P. malariae and P. ovale (White, 2004; Crutcher & Hoffman, 2001). The most important of these is P. falciparum because it can rapidly cause fatal infections and it is responsible for the majority of malaria related deaths. The life cycle of the malaria parasite is split between a vertebrate host and an insect vector (Figure 2.2). The extreme rarity of P. vivax in West Africa is apparently due to prevalence of the duffy negative trait in W est Africans. This is an inherited red cell phenotype that lacks the receptor foRr inYvasion of the human red cell by the merozoites of P. vivax. RA When a mosquito infected with the malaria pIarBasite bites human, developmental stages of the parasite called sporozoites is Linjected into the human’s bloodstream (A). The sporozoites then travel toA thNe liver (B). Each sporozoite undergoes asexual reproduction, in which Aits Dnucleus splits to form two new cells, called merozoites. Merozoites enter tIheB bloodstream and infect red blood cells (C). In red blood cells, merozoites Fgro w and divide to produce more merozoites, eventually causing the red Oblood cells to rupture. Some of the newly released merozoites infect oIthTer Yred blood cells (D). Some merozoites develop into sexual stages known asS male and female gametocytes (E). When another mosquito bites the infected EhumRan, ingesting the gametocytes (F). The gametocytes mature in the mosIquVito’s stomach and undergo sexual reproduction, uniting to form a zygote (G). TNhe zygote multiplies to form sporozoites, which travel to the mosquito’s salivary Uglands. If this mosquito bites another human, the cycle begins again (H). 30 AR Y BR N LI A AD F I B O Y Figure 2.2: PlasmoIdTium spp. Life Cycle (http://encartaR.msnS.com/media_461541582/Life_Cycle_of_the_Malaria_Parasite.html) E IV UN 31 2.2 Malaria Control Methods Global Malaria Control and Disease Management Malaria control efforts are currently focused on two major interventions which include targeting the mosquitoes that transmit malaria by the use of insecticide-treated bed net (ITNs) and early diagnosis and treatment of mala ria (EDTM). These are two of the pillars of global malaria control campaiRgn Y(WHO 2000; Olumese, 2005; Barat, 2006; Killeen, 2007). PreventionA of malaria encompasses a variety of measures that may protect against inBfectRI ion or against the development of the disease in infected individuals. Me aLsures that protect against infection are directed towards the mosquito vectAor pNreventing the transmission of gametocyte. These include personal (individuDal or household) protection measures e.g. protective clothing, repellents, bBednAets or community/population protection measure e.g. use of insecticiFdes Ior environmental management to control transmission. Intermittent Opreventive treatment (IPT) with sulfadoxine- pyrimethamine (SP) in pYregnant women is the primary prevention strategy that relies on the use of mediIcTS ation (White, 2005; Yartey, 2006; Schellenberg et al., 2006; Vallely et al.R, 2007) for control of malaria among pregnant women. VSeEveral partnerships have been set-up by international organizations for eNffecItive control of infectious diseases especially malaria. These organizations Uinclude Malaria Control and Evaluation Partnership in Africa (MACEPA) and Roll Back Malaria (RBM). MACEPA’s mission is to demonstrate that scaling up malaria prevention and control under national leadership saves lives, reduces illness, and increases economic opportunity (Macepa, 2007). The Roll Back Malaria partnership is an initiative to improve malaria control in the context of health sector reform. It 32 was initiated in 1998 through a joint partnership of WHO, UNICEF, UNDP and the World Bank. However this joint partnership has now embarked on a Global Malaria Control Programme. This new programmes focused more on reducing the morbidity and mortality associated with malaria. Accordingly, the objectives of the global malaria control strategy were prioritized as follows: Y  Provision of early diagnosis and prompt treatment for the disease; R  Planning and implementation of selective and sustaiRnabAle preventive measures including vector control IB  Early detection of and response to malaria epidemi csL,  Improved prevention and treatment of malaAria iNn pregnant women.  Strengthening of local research capAacitDies to promote regular assessment of countries’ malaria situations, in parItiBcular the ecological, social and economic determinants of the disease. F Y O IT RS IV E UN 33 2.3: Chemotherapy of malaria Chemotherapy remains one of the important practicable tool to control falciparum malaria in sub- Sahara Africa where > 90% of the world’s burden of malaria mortality and morbidity occurs (Sibley, 2001). Chemotherapy in malaria as in other infectious diseases is based on preventing the growth or survival of infect ing agents, by means of drugs without damage to the host. Effective treRatmYent of falciparum malaria depends on a rapid reduction and clearance of pRarasAitaemia. The malaria parasite is however developing resistance rapidly to moIsBt of these drugs. L N 2.3.1: Classification of antimalarial drugs DA There are many antimalarial drugBs wAith specific effect on various stages of the malaria parasite lifecycle that have b eIen developed and are currently in clinical use. The stages of development ofO maFlaria parasite show varying degree of susceptibility to antimalarial drugs. YThe se antimalarial drugs can be categorized according to chemical classes orI sTtage of parasite against which they are most effective (Table 2.1). S ER NI V U 34 Table 2.1: Classification of anti-malaria drugs Class Blood schizontocide Tissue schizontocide 4-Aminoquinolines Chloroquine Amodiaquine Arylaminoalcohols Quinine Quinidine Y Mefloquine R Phenanthrene- Halofanthrine RAmethanol 8-Aminoquinoline Primaquine IB Artemisinin and L Dihydroartemisinin derivatives N Artemether D A Artesunate A Antimetabolites Proguanil IB Proguanil PyrimethamFine SufadoxOine SuYlfale ne IT Dapsone Antibiotics S Tetracycline Tetracycline R Doxycycline Doxycycline E Minocycline Minocycline V NIWHO recommended combination therapy options for Africa 1U. Artemether plus lumefantrine 2. Artesunate plus amodiaquine 3. Artesunate plus sulphadoxine-pyrimethamine 4. Dihydroartemisinin plus piperaquine 2.3.2: Pharmacology of antimalarial drugs 35 Quinine Quinine is a quinoline antimalarial (Fig. 2.3). It has been the drug of choice for the management of severe malaria in most areas of the world (WHO, 2005). Krishna et al (2001) reported that malaria parasites still remain sensitive to quinine in Africa despite resistance to commonly used quinolines although, in some part of South East Asia decrease sensitivity has been detected (RBM 2001). Recently, it was stYron gly suggested that parenteral artesunate should replace quinine as the treatmenRt of choice for severe falciparum malaria worldwide. Artesunate was well tRolerAated, with no serious drug-related adverse effects with significant reductionIs Bin parasite clearance time (Roshental., 2008; Dondorp et al., 2010). Quinoline aLntimalarials are known to have similar mechanism of action in malaria chemAothNerapy (Slater, 1993; Francis et al., 1997). They are known to inhibit digeAstioDn of haemoglobin by the parasite and thus reduce the supply of amino acidsI nBecessary for parasite viability. Salako et al. (1989) reported a significantly loFwer clearance and longer half-life in children with kwashiorkor than normal ch ilOdren. Administration of quinine plus nevirapine results in significant decreasTes oYf AUC, Cmax and t1/2 in healthy volunteers (Soyinka et al., 2009). AdversSe rIeactions are common with quinine therapy, but severe life- threateninEg tRoxicity is rare. A characteristic symptom complex known as cinchonism is caIuVsed by quinine (Powell & McNamara, 1966). These consist of tinnitus, high tNone deafness (Roche et al., 1990), nausea, uneasiness, malaise, and blurred vision. UHypoglycemia is a more commonly encountered problem with quinine treatment. Quinine clearance is increased by 36 RY RA IB Figure 2.3: Chemical structure of Quinine N L DA phenobarbitone, rifampicin (PukrittayakBameAe et al., 2003) and smoking, but reduced by cimetidine. The pharmacokiFneti cI parameters of quinine are reported to be significantly altered during m Oalaria and this alteration is in proportion to the severity of the infection. MalariaY infection causes a reduction in volume of distribution and clearance leadingS toI a T prolongation of elimination half-life as well as elevated plasma drug concentRrations (Babalola et al., 1998). VE UN I Mefloquine Mefloquine is a quinoline-methanol compound, which is structurally similar to quinine (Fig. 2.4). It was developed by the Walter Reed Army Institute of Research (WRAIR), to combat emerging strains of drug resistant P. falciparum (Bryskier et al., 1988). Structurally, it consists of 2– and piperidyl and 37 trifluoromethyl group in position 2 of the quinoline nucleus. Mefloquine is effective against all malaria species including multidrug resistant P. falciparum. Initially mefloquine against chloroquine resistant and sensitive strains of P. falciparum has been well reported (Doberstyn et al., 1997; Sowunmi et al., 1990; 1992). However significant resistance has developed in South East Asia (WHO 2000a). Oduola et al. (1987) reported a reduction in the in-vitro sensitivity of isolates of P. falcipar um from West Africa to mefloquine even before the drug was introduced into Rthe rYegion, suggesting that parasite with innate resistance to mefloquine may Rbe Apresent in the West Africa sub region. The structure-activity relation is bIaBsed on N-O of 2-  piperidyl group (Tracy et al., 1996). L The actual mechanism of action of mefloquAineN is still unknown however, it is thought to inhibit heme polymerase. MefloquDine forms toxic complexes with free heme causing damage to parasite mIeBmbr Aane and interacts with other plasmodia components (Mockenhaupt, 1995F). Mefloquine is metabolized by CYP3A4. An increase in plasma conc enOtration was reported when mefloquine was co- adminisatered with TketYoconazole, a CYP3A4 inhibitor (Ridtitid et al., 2005). Rifampicin whiSch Iinduces CYP3A4 enzyme reduced plasma concentration of mefloquine wRhen coadministered (Ridtitid et al., 2000). Mefloquine is generally wellI Vtole Erated, although nausea, abdominal discomfort, dizziness, vomiting and dNiarrhea are the frequent adverse reactions associated with it (Phillips-Howard et al., U1995; Ter Kuile et al., 1995; Sowunmi et al, 1990). There is no established biochemical basis behind neurotoxicity of mefloquine but it is found to disrupt neuronal calcium homeostasis and induce an endoplasmic reticulum stress response at physiologically relevant concentrations effect that may contribute at least in part of the neurotoxicity (Geoffrey et al., 2003). It also results in severe neuropsychiatry 38 reactions such as disorientation, seizures, encephalopathy, hallucinations and sleep disturbances (nightmares). Recently, mefloquine has been reported to induce pneumonitis (Soentjens et al., 2006). AR Y R LIB AN BA D I OF Y Figure 2.4: Che TSmicIal structure of Mefloquine ER IV UN 39 Halofantrine Halofantrine is a 9-phenanthrene-methanol antimalarial agent (Fig. 2.5). It is a schizonticidal drug that probably acts by formation of a complex with Ferriprotoporphyrin IX toxic to the parasite. Inhibition of proton pump at the host- parasite interface has also been hypothesised as an alternate mode of action (Watkins et al., 1988; Barriso & Goa 1992). It is active against all human malarial parasi tes. Minor and reversible event including nausea, diarrhea, vomiting and RabdYominal discomfort occur but are usually self-limiting. Pruritus has been reRporAted in 13% of Nigerian taking the drug (Sowunmi et al., 1989). It has no adIvBerse effect on central nervous system and is better tolerated than mefloquine L(TerKuile et al., 1993). Halofantrine prolongs Q-Tc interval (Dion et alA., 2N001; Darrel et al., 2001) and should not be administered to patients taking drDugs known to prolong the QT interval (i.e. Quinine, tricyclic antidepressant, BQuAinidine) or to those who have received mefloquine within few days (WhitFe, 1 9I96; Touze et al., 1997). Oral bioavailability is increased up to six-fold if h alOofantrine is taken with fatty meal (Shanks et al., 1992; Tracy et al., 1996). TY SI VE R I UN 40 Y AR BR LI DA N A F I B O Figure 2.5: Chemical stYruc ture of HalofantrineIT ER S V UN I 41 Artemisinin and its derivatives The family of trioxane compound derived from the plant artemisia annua (qinghaosu) and a series of synthetic derivatives are among the most potent antimalarial compounds known (Fig. 2.6). The trioxane structure contains a peroxide group essential for its antimalarial activity (Klayman, 1985; Woerdenbarg et al., 1994). These naturally occurring sesquiterpene lactones are structurally unrelated to the quinoline antimalarial drugs. Artemisinin derivatives include: ARrtemYisinin, deoxyartemisinin, dihydroartemisinin (DHA), artesunate, aRrtemAisinic acid, artemisitene, arteether and artemether respectively. Three cIoBmpounds have been extensively evaluated: the parent compound artemisinin, t heL water soluble artesunate and the oil soluble ether artemether. AN 4 Artemisinins act very rapidly reducingD parasitemia by a factor of 10 with each cycle (Baird et al., 2005; Baird, 20B05A; Wilairatana et al., 2002). Unfortunately, there are reports of reduced susceFpti biIlity of the artemisininsin the Thai/Cambodian border (Jambou et al., 2005 ; ODandorp et al., 2009). Artemisinin can be given orally and intramuscularly. TArtYemisinin, but not other derivatives (Karunajeewa et al., 2004), shows consideraSble Iautoinduction of its own metabolism so that blood concentrations after several Rdays of dosing are considerably lower than would be predicted from the initiIalV dos Ee (Ashton et al., 1998). Oral artesunate and artemether, but not artemisinin, aNre hydrolyzed rapidly back to the common metabolite DHA, which is intrinsically Umore active as an antimalarial agent (Li et al., 2007). Oral artesunate may be considered mainly as a prodrug for DHA, as the metabolite is the main contributor to overall antimalarial activity (Newton et al., 2000). 42 Y AR LIB R Artemisinin DA N IB A OF ITY RS IV E UANrtemether 43 Y R BR A LI Artesunate DA N BA F IO Figure 2.6: Chemical struYctu re of Artemisinin derivativesIT ER S NI V U 44 2.4: Review of studies on Pharmacokinetic of chloroquine, sulphadoxine- pyrimethamine and amodiaquine. Chloroquine: Chloroquine, is 7-chloro-4-(4-diethyl amino-1-methylbutyl-amino) quinoline, and a tertiary amine (Figure 2.7). It is the most important of the 4-aminoquinol ine compounds for the treatment of malaria. The 4-aminoquinoline nucleus Yhas a th chlorine atom attached to 7th carbon atom of the nucleus. The chloriAne aRtom at 7 position of the quinoline nucleus greatly confers the antimalariaRl activity on the molecules (Goodman & Gillman 1996). B N LI AD A F I B Y O SI T VE R NI Figure 2.7: Chemical structure of Chloroquine U 45 (i) Mode of action: Chloroquine is a weak base but its accumulation in the parasite lysosome is 1000-fold greater than predicted on the basis of a weak base effect (Rang et al., 1997). Chloroquine acts on the intraerythrocytic P. falciparum stages that are responsible for the clinical manifestation of the disease. These stages feed on the erythrocyte haemoglobin, in acidic vacuoles (lysosomes). Chloroquine inhib its digestion of haemoglobin by the parasite and thus reduces the supply of amRinoY acids necessary for parasite viability (Figure 2.4); it is also said to causeR fraAgmentation of the parasite RNA and intercalate DNA. The toxic haem is polyImBerized into insoluble non-toxic haemozoin (Sarchez et al., 1997). Plasmodial heLme polymerase catalyses this polymerization reaction and it is inhibited byA chlNoroquine and other quinolines (Slater, 1993; Francis et al., 1997). A highDer parasite burden is associated with increased risk of failure of treatment wiBth CAI Q and mefloquine (Sowunmi et al., 2004 & 2005). F O Pharmacokinetics ofT chYloroquine (i) Absorption: S ChIloroquine is rapidly absorbed and widely distributed after oral administratioRn in healthy adults (Gustafsson et al., 1983) and children with uncoImVpli Ecated malaria (Adelusi et al., 1982). In adults with moderately severe mNalaria, bioavailability relative to parenteral treatment was 70% compared with 75% Uin healthy subjects (Gustafson, 1983). In children with uncomplicated malaria given an initial oral treatment dose of 10mg base/kg, peak plasma concentration of approximately 250µg/L was reached in 2h (Adelusi et al., 1982). Absorption is very 46 Y RA R LI B DA N A IB OF Figure 2.8 Y M ode of action of Chloroquine IT ER S NI V U 47 rapid and relatively complete even in very severe infections (White et al., 1987b, 1988). The absorption and elimination of chloroquine in children appears to be similar to that in adults (WHO 2000). (ii) Distribution: Chloroquine is widely distributed throughout the body after absorption. It is extensively bound to tissues particularly liver, spleen, kidney, lun gs, connective tissues and tissues containing melanin such as retina resultYing in enormous apparent volume of distribution (Walker et al., 1A983R). The pharmacokinetic properties of chloroquine are complex. The totalR apparent volume of distribution is enormous (100-1000 L/kg) because of LexIteBnsive tissue binding (Frisk-Holmberg et al., 1984) whereas the volume of Ncen tral compartment is several orders of magnitude smaller (0.18 ±0.13L/kg) (LAooareesuwan et al., 1986). The process of distribution from this centrAal Dcompartment determines the blood concentration profile during the treat mIeBnt of malaria. Chloroquine concentration in red blood cells is approximatelyF 3-5 times higher than in plasma, and there is considerable concentratioYn i n O granulocytes and platelets (Nosal et al., 1988). Whole blood concentrationI iTs 6-10 times higher than plasma concentration (Gustafsson et al., 1983) and fuSrther in parasitized red blood cells (Adelusi et al., 1982; Ajayi et al., 1988). CEhloRroquine concentration in cerebrospinal fluid are very low, with a mean valuIe Vof 2.7% of corresponding whole blood concentration (White, 1988b) and UsNlightly concentrated in breast milk with area under the concentration time curve milk/plasma ratio of 2.0 - 4.3 (Edstein et al., 1986). Chloroquine in saliva has a long elimination half-life (7-20d) and there is a good correlation between the AUC values derived from saliva and plasma (Onyeji and Ogunbona 1996; Ogunbona et al., 1986). 48 (iii) Elimination: Chloroquine is eliminated slowly such that the drug and it metabolites can be detected in plasma for 21 - 60 days after single dose of 5mg/kg depending on the sensitivity of the assay method. It is 51% cleared unchanged by the kidney (Gustafsson et al., 1983; Frisk-Holmberg et al., 1984). The remainder is slowly biotransformed by side chain de-ethylation in the liver leading to the formation of the major metabolite, desethyl-chloroquine which is the primary am ine that can undergo deamination to form an alcohol - the 4-hydroxyl compoRundY which then undergoes oxidation to form the carboxylic acid derivativeRs. AThe principal metabolite, desthyl-chloroquine also exhibits potent antimaIlaBrial activity against sensitive isolates of P. falciparum (Oduola et al., 1989 ),L but less active than the parent drug and is also eliminated more slowlyA (GNustafsson et al., 1987). The terminal elimination half-life is approximatelyD 1-2months but, in terms of curative treatment (blood concentration), the IreBal Ahalf-life (t1/2) is about 6-7 days (Frisk- Homberg et al., 1984). VarFiou s half-lives have been reported after oral administration of chlroquine . O This is because the determination of the half-life of a drug depends on the TideYntification of the true terminal log linear elimination phase, which is difficulSt toI obtain with chloroquine in view of its continuous redistribution from tissuesR to plasma over weeks. It also depends on the sampling time and sensIitVivit Ey of the assay method. Drugs that inhibit the actions of liver microsomal eNnzymes prolong the half-life of chloroquine (Bowman & Rand, 1980). U (v) Toxicity: Serious adverse reaction associated with use of chloroquine is rare at therapeutic dosage. The common side effect among Nigerians is pruritus (Sowunmi et al, 1989). Some patients may vomit and may complain of blurred vision (Ferreras et al., 2007). It causes disruption of lysosome in living cells (Michihara et al., 2005). Cardiovascular abnormalities such as hypotension or cardiac arrhythmia progressing 49 to cardiac arrest and death are often observed after parenteral administration or overdose with the drug (Olatunde, 1970; Williams, 1966; Tuboku–Metzger, 1964; White, 2007). For instance, there are reports of sudden death following administration of intramuscular chloroquine to children with severe malaria (Olatunde, 1970). Since chloroquine is rapidly absorbed in the gastrointestinal tract, the use of parenteral route of administration is not encouraged. Prolonged treatm ent with chloroquine may cause a lichenoid skin eruption in some patieRnts.Y The condition is mild and subsides when the drug is discontinued. ChloRroqAuine overdose (usually self poisoning) is manifested by coma, convulsiIonB, dysrhythmias and hypotension. Diazepam is a specific antidote (Riou et aLl., 1988). Oral activated charcoal is administered when an overdose is used.A N AD Sulphadoxine B F I Sulfadoxine (SDX) i s Oa weak acid (Fig. 2.9) and 88 - 90% bound to plasma protein, mainly albumin Y(Abdi et al., 1995; Mayxay et al., 2001). Body weight and age significantlyS inIfl Tuence the pharmacokinetics of sulphadoxine (Trengue et al., 2004). SulfRonamides are structural analogues and competitive antagonists of p- aminoVbenEzoic acid (Rang et al 1995). Sulphadoxine inhibit dihydropteroate synthase (NDHIPS), a key enzyme in the folate biosynthesis (Rang et al 1995). Sulphadoxine is Ua partner drug with pyrimethamine for treating malaria. They act through a two steps synergistic blockage of plasmodial metabolism. The success of SP depends on this synergy: when either component is compromised, the effectiveness is dramatically reduced. 50 (i) Mode of action: This is the most used of a family of drug combinations which antagonize parasite folic acid synthesis. Sulphadoxine act by inhibition of dihydropteroate synthase (sulphonamide and sulphones) (Fig 2.10) This leads to a decrease level of fully reduced tetrahydrofolate, a necessary co-factor important in one-carbon transfer reactions in the purine, pyrimidine and amino acid biosynthetic pathway (Ferone, 1977). The lower level of tetrahydrofolate result in decrea sed conversion of glycine to serine, reduced methoinine synthesis, and lower thRymYidylate levels with a subsequent arrest of DNA replication (Gregson & PlowRe, 2A005). B LI DA N A IB O F TY Figure 2.9: CheSmicIal structures of Sulphadoxine. ER IV UN 51 Sulphadoxine Pyrimethamine RY Figure 2.10: Mode of action of sulphadoxine/pyrimethamine A BR Pharmacokinetics of sulphadoxine LI (i) Absorption: Sulphadoxine is well absorbed AoralNly. In healthy subjects, peak plasma concentration is reached in 2-8h (AhAmaDd & Rogers, 1980; Weidekamm et al., 1982; Dzinjalamala et al., 2005a). AbsIoBrption in uncomplicated malaria is similar to that in healthy subjects (WinstanleFy e t al., 1992). O (ii) Disposition: The apYpare nt volume of distribution of sulfadoxine (0.1-0.2L/kg) (WHO, 2000). It isI lTipophilic. Plasma protein binding of sulphadoxine in healthy subjects is RhighS (88%) (Abdi et al.,1995). Red blood cells concentration of sulphadoxEine is less than half of that in plasma (WHO, 2000). Pregnancy has been showInV to have significant effect on disposition of sulphadoxine; it reduces SDX half- UlNife, lowers the AUC and clearance is significantly greater than in non-pregnant women (Green et al., 2007). (iii) Elimination: Sulphadoxine is a slowly eliminated sulfonamide. Unlike other sulphonamides, only 5% of sulphadoxine is n-acetylated and eliminated in the urine in this form (WHO, 1984). Biotransformation and clearance is approximately 52 0.5mL/kg/h. Elimination half-life of sulphadoxine ranges from 4.8 to 10.6 day (Winstanley et al., 1992; Dzinjalamala et al., 2005). (iv) Toxicity: Sulphadoxine is very well tolerated and severe adverse effects are uncommon (WHO, 1985). It has been reported to cause kernicterus in neonates but sulphonamide treatment in a lactating woman does not pose a threat to her breast fed neonates unless there is jaundice, prematurity or G-6PD deficiency (WHO, Y2000). Life threatening erythema multiforme (Steven Johnson syndromAe) Rand toxic epidermal necrolysis has been reported in individuals taking the druRg for prophylaxis (Miller et al., 1986). LI B AN Pyrimethamine AD Pyrimethamine (PYR) is a IbBase (Bergqvist et al., 1985)(Fiig. 2.11). Pyrimethamine is a competitiOve iFnhibitor of dihydrofolate synthesis (DHFR) (Peter, 1997). Pyrimethamine aYcts through a two steps synergistic blockage of plasmodial division. The succIesTs of pyrimethamine in treatment of malarial depends on sulphadoxine-pySrimethamine synergy: when either component is compromised, the effectivenEessR is dramatically reduced. IV U(Ni) Mode of action: This is the most used of a family of drug combinations which antagonize parasite folic acid synthesis. They act by sequential inhibition dihydrofolate reductase (pyrimethamine and biguanides) enzymes in the folate pathway (Fig 2.6) (Bzik et al., 1987; Triglia and Cowman, 1994; Triglia et al., 1999; Cowman, 1997). This leads to a decrease level of fully reduced tetrahydrofolate, a necessary co-factor important in one-carbon transfer reactions in the purine, 53 pyrimidine and amino acid biosynthetic pathway (Ferone, 1977). The lower level of tetrahydrofolate result in decreased conversion of glycine to serine, reduced methoinine synthesis, and lower thymidylate levels with a subsequent arrest of DNA replication (Gregson & Plowe, 2005). RY BR A LIN DA IB A F Figure 2.11: Chemical stru ctOures of Pyrimethamine Y PharmacokinetiScs oIf T Pyrimethamine (i) AbsorEptioRn: Pyrimethamine is well absorbed orally. In healthy subjects, peak plasImVa concentration is reached in 2-8h (Ahmad & Rogers, 1980; Weidekamm et al., U1N982; Dzinjalamala et al., 2005a). Absorption in uncomplicated malaria is similar to that in healthy subjects (Winstanley et al., 1992). (ii) Disposition: The apparent volume of distribution of pyrimethamine is 2-3 L/kg (Weidekamm et al., 1987). This is considerably larger than volume of the sulfadoxine (0.1-0.2L/kg) (WHO, 2000). Both drugs are lipophilics. Plasma protein binding of pyrimethamine in healthy subjects is high (93%) (Abdi et al.,1995). 54 (iii) Elimination: Biotransformation and clearance is approximately 0.5mL/kg/h. Elimination half-life of pyrimethamine is 3.3 to 4.8 day (Winstanley et al., 1992; Dzinjalamala et al., 2005). Pyrimethamine is transformed to several unidentified metabolites and is cleared predominantly by hepatic biotransformation. (iv) Toxicity: Pyrimethamine is very well tolerated and adverse effects are r are (WHO, 1985). Prolong use of pyrimethamine may provoke folate deficieYncy in vulnerable subjects (pregnant or malnourished patients. No record of caArdioRtoxicity of pyrimethamine has been recorded either in animal or human exRperiment (White, 2007). LIB N Amodiaquine DA Amodiaquine is a 4-aminiquIiBnoli Ane similar to chloroquine (Fig 2.11). Amodiaquine was synthesized durFing the 2nd world war and has a similar action with chlroquine since both are str ucOtural analog (Brykier et al., 1988). It is made up of the same quinoline nucleTus Ywith lateral para-hydroxylphenyl group linked to a methyl- aminodiethyl chSain.I Amodiaquine is as effective as chloroquine and also used against chlorRoquine-resistant strains of P. falciparum (Van Dillen et al., 1999; Olliaro et alI.,V 199 E6). UNMode of action of amodiaquine Amodiaquine is a schizonticidal antimalarial drug. The mechanism of action of amodiaquine is not completely certain. Like other quinoline derivatives, it is thought to inhibit heme polymerase activity. This results in accumulation of free heme, which is toxic to the parasites and leads to the parasite death. 55 Amodiaquine Y R BR A LI N AD A B N-desethyl amodiaquine I O F Figure 2.12: Chemical Ystructure of amodiaquine and N-desethyl amodiaquine PharmacokinetiScs oIf T amodiaquine (i) AbsorEptioRn: Amodiaquine is rapidly absorbed, widely distributed and extensively metaIbVolized to a pharmacologically active metabolite, desethyl amodiaquine, UfNollowing oral administration and it seems that this metabolite is responsible for the antimalarial activity of the compound (Winstanley et al., 1985). The main metabolite of AQ is N-desethylamodiaquine (DEAQ) with other minor metabolites being 2- hydroxyl-DEAQ and N-bisdesethylAQ (bis-DEAQ) (Churchill et al., 1985, 1986; Mount et al., 1986). The primary route of AQ metabolism to DEAQ is via polymorphic CYP2C8 enzyme (Li et al., 2002; Adejei et al., 2008). The absorption 56 and elimination of amodiaquine in children appears to be similar to that in adults (WHO 2000). (ii) Distribution: Amodiaquine is concentrated in the red blood cells, the whole blood to plasma concentration ratio being 3.1. The AUC0-24h for DEAQ in whole blood is significantly higher than that in plasma (Winstanley et al., 1987). The to tal apparent volume of distribution of DEAQ is small compared to chloroquine (Y87.9 – 243.1L/kg) (Stepniewska et al., 2009; Orrell et al., 2008). Both amoAdiaRquine and DEAQ were found to be highly bound to plasma protein with a meRan bound fraction of 92 and 85% respectively (Li et al., 2003). LIB (iii) Elimination: The formation of DEAQ is rapid, itNs el imination is very slow with a terminal half-life of over 100 h (Winstanley et alA., 1987; Laurent et al., 1993; Adjei et al., 2008; Orrell et al., 2008). Excretion oAf DDEAQ is slow and is detectable in urine 5 months after dosing (Wintsanley et aIl.B, 1987). (iv) Toxicity: Amodiaquine at usuFal doses has similar adverse effect to chloroquine but high prevalence of aYgran u Olocytosis and hepatitis (Hatton et al., 1986) after long- term use. Hepatitis IhTas been observed to occur from as early as 3 weeks (exposure to 3 weekly dosesS) to as late as 10 months of prophylaxis. Patients with severe amodiaquEineR induced hepatitis may remain jaundiced for 3-6 months. Severe neutIroVpenia may occur if amodiaquine is used in anti-inflammatory doses for UrNheumatoid arthritis. It seems to be an unstable molecule and undergoes auto- oxidation in aqueous solution to yield a quinoneimine, which may be implicated in the drug’s toxicity (Maggs et al., 1987). However, the adverse reactions appear to be idiosyncratic and have not been described when amodiaquine is used in malaria therapy (Olliaro et al., 1996). 57 2.5: Antimalarial Drug Resistance The emergence and spread of drug resistant malaria globally has become one of the most important problems in malaria control in recent years (Bradley, 1996; Olliaro et al., 2004). The parasites that caused the disease have developed resistance to all antimalarial including artemisinin derivatives (WHO 2006; Jambou et al., 2005; Dondorp et al., 2009). Chloroquine resistance is now common in every region wh ere P. falciparum occurs (Edward & Biagini, 2006). Chloroquine was formerlRy reYplaced by sulphadoxine-pyrimethamine until when this combination succRumAbed in South- east Asia, South America and most recent, Africa (WHO 2I0B06). The impact of resistant malaria is considerable and compounds the seri oLusness of malaria related morbidity and mortality (Price et al, 2001). It is tAhe Nmost critical factor in reducing the useful life span of a drug and undermining the drug policy. AD Antimalarial drug resistance isI Bdefined as the ability of a parasite strain to survive and/ or multiply despite thFe a dministration and absorption of a drug given in doses equal to or higher than Othose usually recommended but within the tolerance of the subject; and the IdTrugY in question must gain access to the parasite or the infected red blood cell foSr the duration of time necessary for its normal action (WHO, 2001). The mainE mRechanism underlying the development of resistance includes, naturally occuIrVring genetic mutations in the malaria parasite. These mutations result in a dNecline in drug sensitivity depending on the class of antimalarial drug (Olliaro, 2004). UInadequate treatment (e.g. sub-therapeutics dose, sub-optimal drug) of a high biomass infection will not kill the mutant parasites and is the main selective pressure for resistance. Resistant parasites are then transmitted to other individuals by mosquitoes. In addition, drugs with long half-lives are more likely to select for resistance because low drug concentrations linger on and are only able to kill sensitive parasites (Nzila 58 et al., 2000). The need to extend the clinical use of chloroquine has become important for several reasons including the socioeconomic situation in endemic areas of Africa and South America. The availability, tolerability and it low cost which had been the main cause to retain it in malaria control efforts. In Nigeria, chloroquine and sulphadoxine-pyrimethamine were antimalarial drugs of choice prior to chaYnge of treatment policy to artemisinin based combination therapy (ACTs) in 20R04 (FMH, 2004). In few malaria endemic areas such as Malawi, the efficacyR ofA CQ has been renewed through withdrawal of CQ from circulation to reduceI dBrug pressure (Laufer et al., 2006). Chloroquine has also been combined wit hL artesunate yielding high efficacy (Fehintola and Balogun 2010). SulphadAoxiNne-pyrimethamine is currently employed as Intermittent Preventive TreaAtmeDnt in children (IPTc) and pregnant women (IPTp). It is essential toI Bstudy the effect of drug resistance on pharmacokinetics of CQ or SP duFrin g acute infections in children. With respect to drug level estimation, the Odetermination of antimalarial drug levels has been estimated in generalI fTromY whole blood, plasma or red cell (White., 1992; Gbotosho et al., 2009; Obua eSt al., 2008). HEoweRver, antimalarials have also been estimated in saliva, for example, quinIinVe (Salako and Sowunmi 1992; Wilson et al., 1993, Babalola et al 1996). UANlthough, there is much difficulty in measuring artemisinin drugs in biological samples, it is often easier to measure their partner drugs, e.g. amodiaquine or sulphadoxine plus pyrimethamine (Gitau et al., 2004; Gbotosho et al., 2009; Obua et al., 2008). An ideal medium from which antimalarial drugs should be measured should be non-invasive with respect to sample collection; saliva is one such medium (Salako and Sowunmi 1992; Wilson et al., 1993). This will provide useful 59 information that may guide drug combination strategies and drug therapeutic monitoring in order to revisit existing but abandoned drugs as a result of drug resistance to all antimalarial drugs (Dondorp et al., 2009). RY RA LI B DA N BA F I Y O IT RS IV E UN 60 CHAPTER THREE MATERIALS AND METHODS Study 1: Studies on risk factors contributing to delay in parasite clearaRnceY in children with acute uncomplicated falciparum malaria. A Drug resistance in Plasmodium falciparum is commoBn iRn many endemic countries but there is no clear recommendation on whe n Lto Ichange therapy when there is delay in parasite clearance after initiation of thNerapy in African children. This study reports the relationship between delay iDn paArasite clearance and anti-malarial treatment failure in children with fBalciAparum malaria in an area of intense transmission in south-western Nigeria ,I where resistance in Plasmodium falciparum to CQ and SP has increase steadiOly oFver the past ten years. Y 3.0: Patients andI dTrug treatment. The sRtudSies were conducted between April 2002 and July 2010 in patients presenVtinEg at the Malaria Research Laboratories Clinic, University College Hospital, INbadIan. Ibadan is an hyper-endemic area for malaria in southwestern Nigeria (Salako Uet al., 1990). Ethical clearance was provided by University of Ibadan/University College Hospital ethics committee and Oyo State Ministry of Health, Secretariat Ibadan. During the period, a series of antimalarial drug efficacy studies were conducted to evaluate the efficacy and safety of different treatment regimens (Table 3.1). The study was conducted with the assistance of a physician (Professor A. Sowunmi). 61 Briefly, children with symptoms compatible with acute uncomplicated falciparum malaria who fulfilled the following criteria were enlisted in the study: age ≤12 years, pure P. falciparum parasitaemia greater than 2000 asexual forms/µl blood, negative urine tests for antimalarial drugs (Dill-Glazko and lignin tests), absence of concomitant illness, no evidence of severe malaria (WHO, 2000) and written informed consent given by parents or guardians. Clinical and parasitologi cal evaluation was done at enrollment (day 0) and on days 1-7, 14, 21 and Rday Y28. In patients who received ACTs, follow-up was for 42 d. Clinical evaluRatioAn consisted of a general clinical examination including measurement of weIigBht, core temperature and physical examination. L AN 3.1: Sample collection. D Prior to treatment, 100µl of caIpBilla Ary blood sample was obtained from each patient and spotted on filter paperF (1 10mm Whatman Filter paper) for determination of chloroquine or sulphadox inOe levels. During follow-up, on day 1, 2, 3, 4, 5, 6, 7, 14, 21 and 28 thick bTlooYd films were prepared and 100µl of capillary blood samples were obtained anSd sIpotted on filter paper. The filter paper samples were stored in desiccated rReseal able plastic bags at room temperature for measurement of chlo EIroVquine and sulphadoxine concentrations. UN 62 Table 3.1: Antimalarial drugs and their treatment regimens with time of study administered to children with acute uncomplicated falciparum malaria. Treatment Drugs* Regimens† No of Years Group patients of study Monotherapy AQ 30 mg/kg of amodiaquine base over 3 days, that is, 10 mg/kg 573 2005/6 daily AS Artesunate given as 28 mg/kg over 7 days, that is, 4 mg/kg 120 2006 daily CQ 30 mg/kg of chloroquine base over 3 days, that is, 10 mg/kg 388 2002-4 daily MQ Mefloquine given as 25mg/kg at presentation 176 2007/8 Y Artemisinin AQAS Artesunate given as 4 mg/kg daily for 3 d plus amodiaquine 142 2004/5/ Combination given as in AQ above R 10 Therapy AMQ Mefloquine given as 25mg/kg at presentation plus artesunAate 174 2006 as given in AQAS above R AL Artemether (20mg) plus lumefantrine (120mg) given thus: 5- 90 2007/8 14kg received 1 tab., 15-24kg received 2 taIbB., 25-34kg received 3 tab., > 34kg received4 tab. at pres enLtation, 8 h later and at 24, 36, 48 and 60 h after first dose Non- AQSP Amodiaquine given as in AQ abAoveN plus sulphadoxine- 69 2002-4 Artemisinin pyrimethamine given as 25 mg/kg of the sulphadoxine Combination component at presentation Therapy AQSFP Amodiaquine given as inA ADQ above plus sulfalene- 91 2006 pyrimethamine given as 25 mg/kg of the sulfalene component at presentation B COT Co-trimoxazole giFven Ias 20 mg of the sulphamethoxazole 104 2003 component twice daily CQCP 30 mg/kg of cOhloroquine base over 3 days, that is, 10 mg/kg 315 2003 daily plus chlorpheniramine 8mg start and 4 mg 8 hourly for 5 d. CQKET 30 mg/kYg of CQ base over 3 days, i.e., 10 mg/kg daily plus 70 2004 ketoItTifen 25mg/kg start followed by 0.125 mg/kg 8 hourly for S4 d. CQSPR 30 mg/kg of chloroquine base over 3 days, that is, 10 mg/kg 107 2007/8 daily plus sulphadoxine-pyrimethamine given as 25 mg/kg of E the sulphadoxine component at presentation VSP Sulphadoxine-pyrimethamine given as 25 mg/kg of the I 291 2003 sulphadoxine component at presentation N Sulphadoxine-pyrimethamine given as in SP above plus U SPPB probenecid at 20-25mg/kg in two divided doses daily for 3 day 42 2003 † All drugs were administered orally. AQ, amodiaquine; AQAS, amodiaquine plus artesunate; AQSP, amodiaquine plus sulphadoxine-pyrimethamine; AQSFP, amodiaquine-sulfalene-pyrimethamine; AMQ, mefloquine plus artesunate; AL, artemether plus lumefantrine; AS, artesunate; COT, co-trimoxazole; CQ, chloroquine; CQCP, chloroquine plus chlorpheniramine; CQKET, chloroquine plus ketotifen; CQSP, chloroquine plus sulphadoxine- pyrimethamine; MQ, mefloquine; SP, Sulphadoxine-pyrimethamine; SPPB, sulphadoxine-pyrimethamine plus probenecid; 63 3.2: Assessment of parasitaemia. Thick and thin blood films prepared from a finger prick were Giemsa-stained and were examined by light microscopy under an oil-immersion objective, at X100 magnification and re-examined by an independent microscopist. Parasitaemia in thick films was estimated by counting asexual parasites relative to 1000 leukocytes, or 500 asexual forms, whichever occurred first. From this figure, the parasite dens ity was calculated assuming a leukocyte count of 6000/L of blood. GametocyteYs were also counted in thick blood films against 1000 leukocytes assuminAg aRn average leukocyte count of 6000/L of blood (Shaper & Lewis, 1971; EzBeiloR, 1971; Sowunmi et al., 1995). LI AN 3.3: Evaluation of response to drug treatment. D Response to drug treatment wIasB as Asessed using World Health Organization (WHO) criteria (WHO, 1973 or 20F03 ) as shown in Table 3. 2. In those with sensitive or RI response, parasite clea raOnce time (PCT) was defined as the time elapsing from drug administration uTntilY there was no patent parasitaemia for at least 72 h. Delay in parasite clearancSe wIas defined as a parasite clearance time > 2 d, and was based on the asexual liRfe cycle of 48 h in the infected erythrocyte (White, 1997). E V UN I 64 Table 3.2: Evaluation of clinical response of children to treatment with antimalarial drugs Treatment Response Description S (sensitive) clearance of parasitaemia without recurrence RI (mild resistance) parasitaemia disappears but reappears within 7 to 14 days; RII (moderate resistance) decrease of parasitaemia but no complete clearance from peripheral blood; RY RIII (severe resistance) no pronounced decrease or increase inR parAasitaemia at 48 hours after treatment B LI OR N ACPR (adequate clinical and if there was no reappAearance of parasites or fever by parasitological response) day 14 D ETF ( Early treatment failure) if dangerI sBign As or severe malaria developed on days 1, 2F and 3 in the presence of parasitaemia, or Oparasitaemia on day 2 higher than day 0 irrespective Y of temperature, or parasitaemia on day 3 with fever T o≥37.5 C, or day 3 parasitaemia ≥25% of the count on SI day 0 LCF (Late cliniRcal failure) if danger signs or severe malaria developed after day E 3 in the presence of parasitaemia or presence of IV oparasitaemia and fever (≥37.5 C) on any day between N day 4 and day 14; ULPF (Late parasitological if there was parasitaemia on day 14 and axillary o failure) temperature less than 37.5 C without meeting any of the previous conditions 65 Study 2: Pharmacokinetic determinants of response to treatment with chloroquine in children with acute uncomplicated Plasmodium falciparum malaria. Resistance to antimalarial has become an important issue in infectious diseases research. The usefulness of the common antimalarials, such as chloroquine and sulphadxoine-pyrimethamine is fading out as a result of resistance to the drugs. Apart from the issue that the parasites are resistant to these drugs, the way indivYidu als respond to drug therapy varies considerably within a population. RThere are interindividual differences in absorption, distribution, metabolism aRnd Aelimination of drug. LIB Monitoring of efficacy and evaluation of thNe p harmacokinetic parameters requires routine collection of whole blood for drugA analysis. The use of whole blood collected through finger prick and adsorbAed Don filter paper has reduced risk of infection (Green et al., 2002) and se mIi-Bskilled field workers can collect the samples after minimal training with little Fdiscomfort or risk to the patient or volunteer. In addition, blood collectioYn o n O filter paper reduces the need to provide facilities for separating and storIinTg blood samples in the field. However, collection of field samples for phaSrmacokinetic studies in children has particular challenges since frequent EsamRpling over a follow up period is required. Children tend to be less williInVg to have repeated finger pricks. When such situations arise, it may be difficult UtNo obtain the correct blood volumes especially using capillary tubes. Different methods for measuring drug levels on filter paper have been developed (Minzi et al., 2003; Gitau et al., 2004; Hoegberg et al., 2005). The pharmacokinetic determinants of antimalarial treatment failure are important in chemotherapy. This will help in prompt treatment of drug failure especially in children and pregnant women who are the vulnerable groups. It is 66 important to recommend correct optimal dosage regimen where it is insufficient especially in children and pregnant women in who low drug levels have been clearly documented (Barnes et al., 2008; Gbotosho et al 2009). Sub-therapeutic concentrations and variability in pharmacokinetic disposition to antimalarial drugs contribute to poorer responses to treatment and increase the spread of antimalarial drug resistance. Suboptimal drug concentrations which could result from inadequ ate drug absorption, an unusually large apparent volume of distribution or duRe toY rapid clearance of the drug. Response to treatment is ensured if aRntimAalarial drugs concentrations produce maximum effect until all malaria parasiIteBs are eliminated. The use of Pharmacokinetic parameters to pred iLct treatment outcome is important and this has not been utilised in antimaAlarNial drug clinical trials to asses progression of responses to drug resistAancDe. Routine measurement of drug concentrations of a slowly eliminatedI aBntimalarial drug such as CQ can be used to determine the minimum therapeFutic concentration during treatment. For a drug eliminated slowly, the area uOnder blood or plasma concentration time curve (AUC) could be a useful phIaTrmYacokinetic predictor of treatment outcome in uncomplicated malaria becauseS it captures both the drug concentration and duration of exposure (White etE alR., 2008). The AUC comprises both the absorption and the elimination phasIeVs of a drug and provides a measure of parasite exposure to the anitmalarial dNrugs. U 3.4: Study population From the delay in parasite clearance study, it was shown that children who were treated with chloroquine and sulphadoxine-pyrimethamine had highest proportions of delay in parasite clearance 70.8% (CQ) and 63.9 % (SP). A cohort of children was randomly selected during enrolment for identification of 67 pharmacokinetic determinants of responses to chloroquine and assessment of pharmacokinetic disposition of sulphadoxine. In this study, children, aged between 6 months – 12years were treated with standard doses of chloroquine (30 mg/kg of chloroquine base over 3 days, that is, 10 mg/kg daily). Children had microscopically confirmed infection with pure P. falciparum at parasiteamia greater than or equal to 2000 asexual parasites perY mi cro liter of blood before enrollment. Prior to treatment, 100µl of capillary bloRod sample was obtained from each patient and spotted on filter paper (110mmR WAhatman filter paper) for determination of chloroquine or sulphadoxine level.I BDuring follow-up, on day 1, 2, 3, 4, 5, 6, 7, 14, 21 and 28 thick blood films w eLre prepared and 100µl of capillary blood samples were obtained and spottedA onN filter paper. The filter paper samples were stored in desiccated resealabAle Dplastic bags at room temperature for measurement of chloroquine and sulphIadBoxine concentrations. F 3.5: Drug analysis O 3.5.1: Reagent for aInTalyYsis. Chloroquine, DeSsethyl chloroquine and Papaverine reference standard were obtained from WEalteRr Reed Army Institute, USA; Diethyl ether, Sodium hydroxide, HydIroVchloric acid and perchloric acid were obtained from BDH chemical England; ANcetonitrile, Methanol and Water were HPLC grade obtained from LiChrosolv®, E. UMerck, Germany. All other reagents were analytical grade. 3.5.2: Preparation of Stock and Working solutions. 1. Sodium hydroxide (NaOH, 5M) working solution was prepared by weighing 3 40g of NaOH pellets and dissolving in 200cm of distilled water in a volumetric flask. 68 2. The working solution of Sodium dihydrogen phosphate (NaH2PO4.2H2O, 0.02M) was made by weighing 3.1202g of the compound and dissolving in 3 1000cm of distilled water. 3. Stock solution of chloroquine, desethyl chloroquine and papaverine (internal standard) were prepared by weighing 5mg of each of the drugs and dissolving in 5ml of 0.1M HCl. The working solution (100µg/ml) was prepared fr om 1mg/ml stock solution by making 1:10 dilution of 1mg/ml. OneR mYilliliter (1ml) of 1mg/ml stock was made up to 10ml with 0.1M HCRl inA a volumetric flask or centrifuge tube. IB 4. Preparation of 0.1M HCl was made from conc enLtrated hydrochloric acid (HCl). The concentrated acid used hadA peNrcentage purity of 35% with specific gravity of 1.18g/ml. The mDolarity of the concentrated HCl was calculated as follows; BA % purity of acid =35%F I molar mass of HCl =36.5 Concentration Oof HCl in 1.18g/ml of the acid content = % purity x specific gIrTaviYty = (35x1.18)/100g/ml = 0.413g/ml i.eS. 413 g of HCl in 1000ml (A) ERWeight equivalent to molar mass of any compound in 1000ml of distilled wIVater = 1M of the compound. UN If 36.5g of HCl (was dissolved in 1000ml solvent) = 1M HCl (B) Therefore if A=B then, 413.0g (in 1000ml solvent) = (413g x 1M)/36.5g = 11.315M (C1). The molarity of the concentrated acid was 11.315M (C1). To prepare 1000ml (V2) of 0.1M HCl (C2). The volume (V1) of concentrated acid required can be 69 obtained from the formula, C1V1 = C2V2, where C1 is initial concentration of the acid, C2 is final concentration, V1 volume of initial concentrated acid require and V2 is final volume respectively. V1= (C2V2)/C1 = (0.1M X 1000ml)/11.315M = (100ml)/11.315 = 8.837ml of concentrated acid. Therefore, 0.1M was prepared by adding 8.837ml of concentrated acid to dYistilled water in volumetric flask and making up to 1000ml with distilled water. RA R IB 3.5.3: Analysis of chloroquine from filter paper samples. LN (a) Instrumentation. A Chromatographic separations were carried AoutD on a Cecil Adept, High performance liquid chromatography unit. The unit cIoBnsists of Cecil 4100 solvent delivery system with a Rheodyne valve injectioFn system and a Cecil 4200 variable wavelength ultraviolet detector operatin g Oat 254nm with a Cecil 4900 chromatographic system manager. The sepaIrTatioYn was carried out on a Waters C18 Reversed-Phase 10m Bondapak coluSmn of 3.9mm X 300mm dimension stainless steel maintained at room temEperRature. IV U(Nb) Mobile Phase Composition. The mobile phase used for the analysis consisted of 0.02M phosphate buffer: methanol: acetonitrile (58:27:15 V/V/V) adjusted to pH 2.5 with perchloric acid. The mobile phase was delivered to the system at a flow rate 1ml/min. 70 (c) Extraction of chloroquine whole blood filter paper samples. Whole blood concentrations of chloroquine were measured from filter paper blood spots using a modified HPLC analysis (Ogunbona et al, 1986). Briefly, filter paper sample was cut into pieces and the pieces were transferred into clean extraction tubes. The pieces of filter paper were soaked in 200µl of 0.1M HCl for 30 minutes. Two hundred nanogram (40µl of 5µg/ml) of papaverine as internal standard was add ed. Two hundred microlitres (200µl) of 5M NaOH was added to the content ofR eacYh tube to basify the sample. A fixed volume (2ml) of diethyl ether was RalsoA added as the extraction solvent. The content was vortexed for sixty secondIsB and then centrifuged for 10 minutes at 2000g. After centrifugation the organic sLupernatant of the mixture was transferred into a clean centrifuge tube. One hAundNred microlitre (100µl) of 0.1M HCl was added to the organic supernatant andD vortexed for 2 minutes. The mixture was thereafter centrifuged for ten mi AInButes at 2000g. The organic supernatant was removed carefully and discarded. FTw enty microlitre (20µl) of the aqueous phase was injected onto the column for aOnalysis. TY (d) Calibration CSurvIes. CEalibRration curve is a plot of detector response (peak/ratio) with change in concIeVntration of known sample. It is a general method for determining the UcNoncentration of a substance in an unknown sample (blood) by comparing the unknown to a set of standard samples of known concentrations. Calibration curves based on peak-height ratios were prepared by spiking drug free blood sample with working solutions of chloroquine and desethyl-chloroquine to yield final concentration of 0 – 2000ng/ml for chloroquine and desethyl-chloroquine on filter paper. Chloroquine metabolite, desethyl-chloroquine was not considered because 71 desethyl-chloroquine standard powder was not available. The peak-height ratio for both the drug and the internal standard were recorded. Concentration of drug in the blood samples was extrapolated from the calibration curve as shown in Figure 4.4. (e) Precision and percentage recovery of the analytical method. Filter paper samples (drug free) were spiked with known concenRtratiYon of chloroquine (between 0 - 2000ng/ml of chloroquine). A 40µl (of 5µg/Aml) aliquot of internal standard was added to the cut filter paper in extractioBn tRube. The samples were prepared (n=5), taken through the extraction procedu rLe aIs described previously and injected onto the column. Another set of standaNrd chloroquine solutions were prepared to yield actual concentration in the spiDkedA filter papers. An aliquot (20µl) of the solution was injected directly into the cAolumn. The peak height of the extracted standard samples and the solutions in jeIcBted onto the system were compared to obtain the percentage recovery of theO meFthod, intra-assay (within day) and inter-assay (day- to-day) coefficient of vaYria tion (C.V.). Limit of detection of the drug was also determined. IT RS IV E UN 72 Study 3: Development of High Performance Liquid Chromatography analytical method for measurement of sulphadoxine concentrations in filter paper sample. Sulphadoxine-pyrimethamine ( SP) analysis from filter paper has been a very difficult analysis because of the nature of chemical composition for sulphadoxine and pyrimethamine. Sulphadoxine is both an acid and a weak base while pyrimethamine is a weak base (Green et al., 2002). This method was developed in order to Yredu ce cost of analysis and the burden of sample storage especially during fRiled trials. Although this method analysed both sulphadoxine-pyrimethamine (RSP) Abut only SDX was considered. It has been reported that there is a correlIaBtion between plasma concentration of SDX and PYR (Bergvist et al., 1 LN987 ), however knowledge of sulphadoxine concentrations provides some inforAmation of the magnitude of the accompanied pyrimethamine concentrationA asD well. The ratio of combination of sulphadoxine-pyrimethamine is 2I0:B1 (500mg of sulphadoxine:25mg of pyrimethamine) in a tablet. OF Y 3.6: Analysis of sulpIhTadoxine from filter paper samples. a. InstrumenRtatiSon. The IsVame E HPLC unit as in CQ analysis was used for sulphadoxine analysis but the cNolumn used was different. The eluent was monitored using a UV detector operated Uat 240nm with a Cecil 4900 chromatographic system manager. The separation was carried out on a Beckman Coulter ODS 5µm column of 4.6mm X 15cm dimension stainless steel maintained at room temperature. 73 b. Preparation of Stock and Working solutions. Stock solution of sulphadoxine, pyrimethamine and sulisoxazole (internal standard) were prepared by weighing 5mg of each of the drugs and dissolving in 5ml of 70% ethanol prepared with 0.1M HCl solution. Working solution (100µg/ml) was prepared from 1mg/ml stock solution by making 1:10 dilution of 1mg/ml. One milliliter (1ml) of 1mg/ml stock was made up to 10ml with 0.1MHCl in a voluYme tric flask or centrifuge tube. AR R c. Mobile phase composition. IB The mobile phase used for the analysis consisted of 1L% triethylamine solution containing 0.05M phosphate buffer: methanol: acetAonitNrile (70:17:13 V/V/V) adjusted to pH 3.4 with phosphoric acid. The mobiAle pDhase was delivered to the system at a flow rate of 0.9ml/min. F I B d. Measurement of sulphado xOine from whole blood on filter paper samples. Sulphadoxine (SDXI) TexYtraction was performed by cutting filter paper samples into pieces and transSferring the pieces into clean extraction tubes. The pieces of filter paper weEre sRoaked in 200µl of 0.1MHCl for 30 minutes and twenty microlitre (20µl) of 5I0V0µg/ml sulisoxazole as internal standard was added. Two millitres (2ml) of aNcetonitrile was also added as an extracting solvent. The content was vortexed for Utwo minutes and then centrifuged for 10 minutes at 2000g. After centrifugation the acetonitrile supernatant of the mixture was transferred into a clean centrifuge tube O and dried under a stream of nitrogen gas in a water bath at 50 C. One hundred microlitres (100µl) of 0.1M HCl was added to reconstitute the dried extracted sample, vortexed for 2 minutes and centrifuged for ten minutes at 2000g. Twenty microlitres 74 (20µl) of the aqueous phases was injected onto the column for analysis. The retention time for sulphadoxine and sulisoxazole were determined. e. Calibration curve for sulphadoxine. The calibration curve for sulphadoxine was prepared by spiking drug free blo od sample with working solutions of SDX to yield final concentration of 0 – 60µgY/ml for sulphadoxine. Using an adjustable pipette, 100µl of drug-spiked donoAr wRhole blood o was adsorbed onto the filter paper. The samples were placed in an Rincubator at 37 C and allowed to dry overnight. The spiked filter paper bLlooIdB sample were taken through the normal assay procedure as discussed abovNe, a nd calibration graphs were constructed using the peak-height ratio of SDAX to internal standard against concentration. AD B F I f. Precision and Recovery. O To assess precision, cToefYficient of variation (CV) was determined for intra- and inter assay variabilityS. TIhis was achieved by carrying out repeated analysis of drug-free whole bloodR spiked with different concentrations of sulphadoxine (n>5 for each concIeVntra Etion) on each of six days. Calibration curves were prepared from the mNeasurement of peak height ratios of the SDX and internal standard. Extraction Urecovery was determined for each concentration by comparing peak heights ratio of the extracted known standards with the directly injected standard concentrations. Commonly used antimalarial drugs including chloroquine, mefloquine, quinine, sulphamethoxazole and trimethoprim were studied for interference by spiking the drugs in the blank whole blood. The drugs were extracted according to the method 75 described above. The presence of peaks was monitored after injection of 20µl of the reconstituted sample. Y R RA LI B AN BA D I OF ITY ER S NI V U 76 Study 4: Pharmacokinetic disposition of sulphadoxine in children with acute uncomplicated malaria treated with standard dose of sulphadoxine- pyrimethamine. Sulphadoxine-pyrimethamine is currently recommended by WHO as a partner drug with artesunate in the chemotherapy of malaria and it is also being used alone for intermittent preventive treatment for malaria in pregnancy and in infants Y(WH O 2006; WHO 2007; May et al., 2008). However, information on pharmacoRkinetics of SDX-pyrimethamine in children with acute uncomplicated malaria Ris liAmited (Barnes et al., 2006; Dzinjalamala et al 2005; Obua et al 2008; IHBellgren et al 1990; Winstanley et al., 1992 and Aubouy et al 2003). ENffor ts L in this study were thus devoted to studying the pharmacokinetic dispositioAn of SDX and evaluating the effect of pharmacokinetic variability on therapeutiAc efDficacy in children between the ages of 6 months and 12 years with acute uIncBomplicated falciparum malaria treated with standard dose of SP. F Seventy-four paYtien t Os treated with standard dose of sulphadoxine- pyrimethamine (25 ImTg/kg of the sulphadoxine component) were randomly selected from cohort of cShildren who had highest proportion of delay in parasite clearance. The simpEle cRost-effective high performance chromatographic analytical method that was IdVeveloped for the analysis of sulphadoxine in capillary whole blood collected on UfNilter paper in study 3 was used for the analysis of these samples. Samples were collected before treatment (day 0), 1, 2, 3, 4, 5, 6, 7, 14, 21 and day 28 post treatment respectively. Reagent and chromatographic conditions are as stated in study 2 and study 3. 77 Study 5: Evaluation of the use of saliva for therapeutic drug monitoring in children with uncomplicated Plasmodium falciparum malaria treated with amodiaquine-artesuante. Saliva offers an easily accessible and sometimes useful body fluid for therapeutic drug monitoring and for pharmacokinetic and pharmacodynamic sYtudies. Saliva is limited to the unbound fraction of the drug. Thus, saliva levelsA maRy be more reflective of drug concentrations at the site of action thanR are total drug concentrations in plasma or whole blood. It has beenL sIhBown that the saliva concentrations of particular drug can be used to predicNt how much of those drugs is in the blood (Fatah and Cohn, 2003). A BA D 3.7: Patients enrolled in the studFy a nId sample collection. Patients were random Oly selected from cohort of children who received oral doses of artesunate-amTodYiaquine (4mg/kg artesunate plus 30mg/kg amodiaquine base over 3 days). SSeveIn children were selected from this group of children. Venous blood (5ml) Rwas collected at 0hr before treatment, 4hr post treatment and thereafter on dIaVy 7, E 14, 21, 28 and 35. Blood samples were collected into heparinised tube and wNere immediately centrifuged at 2000g for 10 minutes to separate the plasma and red U Oblood cells, which were stored at -20 C until analysed. Saliva samples were taken at the same times that venepuntures were done. The samples were collected after rinsing the mouth with water and without any special stimulus to increase saliva flow. The saliva (3-5ml) was immediately centrifuged and the clear fluid was removed and O stored at -20 C until analysed. 78 3.8: Analysis of amodiaquine or desethyl amodiaquine from plasma or saliva samples. (a) Instrumentation. Chromatographic separations were carried out on the same HPLC unit as in CQ study (study 2). Y (b). Preparation of Stock and Working solutions. R Stock solution of amodiaquine, desethyl amodiaquine and quRinidAine (internal standard) were prepared by weighing 5mg of each of the drugsI aBnd dissolving in 5ml of 70% ethanol prepared with distilled water. WorNking Lsolution (100µg/ml) was prepared from 1mg/ml stock solution by makingA 1:10 dilution of 1mg/ml. One milliliter (1ml) of 1mg/ml stock was madAe uDp to 10ml with distilled water in a volumetric flask or centrifuge tube. IB F (c) Mobile Phase Compositi oOn. The mobile phase IcoTmpYrised 0.02M potassium diphosphate buffer, methanol and acetonitrile in thSe ratio 75:23:2 (V/V/V) adjusted to pH 2.65 with phosphoric acid. The separEatioRn was carried out on a Hypersil Reversed-Phase 5µ column of 4.6 mm x 250 ImVm dimension stainless steel maintained at room temperature. The UV detector wNas set at 254nm. The mobile phase was delivered to the system at a flow rate U1ml/min. ( d ) Extraction of amodiaquine/desethyl amodiaquine from plasma or saliva sample. Amodiaquine and desethylamodiaquine in plasma and saliva was assayed by a specific high performance liquid chromatography (HPLC) with ultraviolet detector 79 using a modification of a method described by Gitua and others (2004). Briefly, 0.5 ml of plasma or saliva was transferred into clean extraction tubes. Two hundred and fifty nanogramm (5µl of 50µg/ml) of quinidine was added as internal standard (IS). A fixed volume (5 ml) of diethylether was also added as the extraction solvent. The content was vortexed for sixty seconds and then centrifuged for 10 minutes at 2000g. After centrifugation the organic supernatant of the mixture was transferred into a O clean centrifuge tube and dried in a fume cupboard at 40 C. The reRsiduYe was reconstituted in sixty-five microlitre (65µl) of mobile phase anRd fiAfty microlitre (50µl) injected onto the column. LIB N (e) Measurement of saliva pH. DA The pH of saliva was measured immediAately after collection using a pH meter (Mettler-Toledo GmbH, Sonnenbergs trIaBsse, Switzerland) and recorded immediately. O F (f) Calibration CurvesT. I Y Stock solutionsS of amodiaquine, desethyl amodiaquine (1mg/ml) and internal standard EquinRidine (1mg/ml), was prepared in 70% methanol. Drug free plasma and salivIaV was spiked with the working solution containing both AQ and DEAQ to yield fNinal concentrations ranging from 600 – 100ng/ml of AQ and DEAQ. The samples Uwere assayed as described above and this was used for calibration curve. Concentration of drug in the plasma or saliva samples was extrapolated from a calibration curve. (g) Precision and percentage recovery of the analytical method. Drug free plasma or saliva samples were spiked with known concentration of AQ or 80 DEAQ (between 100 -800ng/ml) and 5µl of 50µg/ml aliquot of internal standard was added. The samples (n=5), were taken through the extraction procedures as described previously and injected onto the column. Another set of standard AQ and DEAQ solutions were prepared to yield actual concentration in the spiked plasma or saliva sample. An aliquot (20µl) of the solution was injected directly onto the column. The peak height of the extracted standard samples and the solutions inYjec ted onto the system were compared to obtain the percentage recovery of thRe method, intra-assay (within day) and inter-assay (day-to-day) coefficient of vRariaAtion (C.V.). LIB 3.9: Statistical analysis for all studies. N Data were analysed using version 6 ofD theA Epi-Info software (Anon., 1994), and the statistical program SPSS for WAindows version 10.01 (Anon., 1999). Proportions were compared by calc uIlaBting 2 with Yates' correction. Normally distributed, continuous data Ower Fe compared by Student's t-tests and analysis of variance (ANOVA). DatYa not conforming to a normal distribution were compared by the Mann-Whitney IUT-test and the Kruskal-Wallis test (or by Wilcoxon rank sum test). The vRalueSs presented are generally means and standard deviations (s.d.) or standaVrd Eerror of mean (s.e.m.). NI In order to determine the risk factors for delay in parasite clearance and Upharmacokinetic factors associated with CQ treatment failure, the relationship between each of the clinical or parasitological data at enrolment and pharmacokinetic parameters following treatment was investigated by univariate analysis. A multiple logistic regression model was used to test the association between response to drug treatment (sensitive or resistant) and the factors that were significant in the univariate 81 O analysis: age, presence of fever (temperature ≥ 37.5 C) at presentation, PC > 2 d (yes or no at presentation or during follow up): presence of fever, asexual parasitaemia at presentation or during follow-up, a history of vomiting, and drug treatment. All these factors were used in multiple logistic regression analysis models to test the association with delay in parasite clearance. GraphPad Prism (version 3.02) and Microsoft Excel statistical softwar e’s were used for collation of all the drug concentrations obtained from each pRatieYnt and used for test of linearity of the standard curves. PharmacokineticR parAameters were obtained using Turbo Ken software (designed by the DIepBartment of Clinical Pharmacology, University of Southampton, United Kingd oLm). P values < 0.05 were considered significant for all statistical analysis exAcepNt in multivariate analysis of t1/2 where P < 0.3 was ‘falsed’ for the analysiDs. In comparison of disposition of amodiaquine and desethylamodiaquiIneB in A plasma and saliva in children with uncomplicated Plasmodium falciFpar um malaria, plasma and saliva concentration- time curve was plotted for eOach subject. Oral clearance was calculated from the equation CL= D/AUCT, DY is the dose given. The apparent volume of distribution (Vd) was calculated frSomI the equation Vd=CL/β. VE R I UN 82 CHAPTER FOUR RESULTS Study 1: Risk factors contributing to delay in parasite clearance in uncomplicated falciparum malaria in children. 4.0: Study Population Y Between April 2002 and July 2010, 2,752 children (1,342 femaAles Rand 1,410 males) were enrolled into the antimalarial drug studies. ABll cRhildren who were recruited into these studies had primary infections wLithI P. falciparum. There were 1,716 under five-year olds. The geoNmetric mean parasitaemia at enrolment was 34,044 asexual parasites/µDl oAf blood (95% CI 30,400 – 33,664) as shown in Table 4.1. IB A 4.1: Drug treatment andO delFay in parasite clearance Overall, delay in pYara site clearance occurred in 1,237 of the 2,752 children (45%) (FigureI 4T.1 and Table 4.2). The highest proportions of children showing delay iRn paSrasite clearance were found in those treated with chloroquine (CQ), sulpEhadoxine-pyrimethamine (SP), chloroquine plus chlorpheniramine (CQCP), IVN co-trimoxazole (COT), amodiaquine (AQ), chloroquine plus ketotifen U (CQKET), chloroquine plus sulphadoxine-pyrimethamine (CQSP), amodiaquine plus sulphadoxine-pyrimethamine (AQSP), sulphadoxine- pyrimethamine plus probenecid (SPPB) and mefloquine (MQ). The proportions of children with delay in parasite clearance were significantly lower in those treated with AS, AQAS, AL and AMQ when compared with the latter 2 group above (χ = 447.91 df = 8, P < 0.0000001). There was no significant 83 difference in the proportions of children with delay in clearance in those treated 2 with AS (3 of 120), AQAS (14 of 142), AL (6 of 90) and AMQ (10 of 174) (χ = 6.1, df =3, P = 0.10). Infection in 291 of the 2,752 (10.6%) children failed to respond to treatment (Table 4.3). RY RA LI B AN BA D OF I ITYS VE R NIU 84 Table 4.1: Baseline clinical and parasitological parameters of 2752 children with acute uncomplicated falciparum malaria enrolled in the study. Variables Mean ± SD (range) 95% CI Age (year) 6.1 ± 3.0 (0.5-12) 6.0-6.2 No. < 5 years 1,716 (63%) Weight (kg) 17.3 ± 6.4 (5-47) 17.0-17.6 O Axillary temperature ( C) Y (n = 2428) 38.3 ± 1.2 (34-42) 38.2-38R.3 o No. with > 40 C 210 A Haematocrit (%) (n = 994) 30.5 ± 4.8 (10-51) 3R0.1-30.7 No. with < 30% 380 IB Parasitaemia (/µl) GM 34,044 L 30,400-33,664 Range 2009-1,19N4,285 No with > 100,000 (/µl) 638 (2A3.2%) No. with > 250,000 (/µl) 18D7 (6.8%) Gametocytaemia (/µl) GM BA27 (6 - 4188) Duration of illness (d) F I 3.0 ± 1.4 (1-14) 2.9-3.0 Duration of vomiting (Od) 1.3 ± 1.4 (1-9) 1.2-1.3 ____________________Y___ _______________ _________________ GM, geometriTc mean; parasitaemia= asexual parasites/µl of blood SI VE R I UN 85 Table 4.2: Proportions of children with Delay in Parasite Clearance following treatment with standard doses of selected antimalarial drugs. Drug Proportion with delay in PC (%) Total with delay in PC 45 (1237) Chloroquine (CQ) 70.8 Sulphadoxine-pyrimethamine (SP) 63.9 Chloroquine + chlorpheniramine (CQCP) 60.0 RY Co-trimoxazole (COT) 58.6 A Amodiaquine (AQ) 52.0 BR Chloroquine + Ketotifen (CQKET) 51.4 LI Chloroquine + Sulphadoxine- 42N.0 pyrimethamine(CQSP) A Amodiaquine + sulfalene-pyrimethamineA D 37.6 (AQSFP) IB Sulphadoxine-pyrimethamine F + 30.9 probenecid (SPPB) O Mefloquine (MQ) Y 22.7 Artesunate + amIoTdiaquine (AQAS) 9.9 ArtesunRate +S lumefantrine (AL) 6.6 VArteEsunate + Mefloquine (AMQ) 5.7 NIArtesunate (AS) ( monotherapy) 2.5 U† All drugs were administered orally. AQ, amodiaquine; AQAS, amodiaquine plus artesunate; AQSP, amodiaquine plus sulphadoxine-pyrimethamine; AQSFP, amodiaquine-sulfalene-pyrimethamine; AMQ, mefloquine plus artesunate; AL, artemether plus lumefantrine; AS, artesunate; COT, co- trimoxazole; CQ, chloroquine; CQCP, chloroquine plus chlorpheniramine; CQKET, chloroquine plus ketotifen; CQSP, chloroquine plus sulphadoxine- pyrimethamine; MQ, mefloquine; SP, Sulphadoxine- pyrimethamine; SPPB, sulphadoxine-pyrimethamine plus probenecid. 86 Table 4.3: Proportion of children with falciparum malaria who failed to respond to treatment with standard doses of antimalarial drugs. Day Total failure Failure rate (%) Y 7 57 2.1 AR 14 136 5.3 R 21 291 L I 1B0.6 2 χ for trend = 158, p < 0.000001; Infection in 291N of the 2,752 (10.6%) children failed to respond to treatment. DA IB A O F ITYS ER NI V U 87 Δ Parasite clearance > 2 d who proceded to failure. Y Δ No. with parasite clearanRce > 2 d Δ Total enrolled BR A N LI DA BA OF I SI TY R Figure 4.1: NEumbers of children with delay in parasite clearance (PC > 2 d) following treatment wIiVth antimalarial drugs. UN 88 4.2: Risk factors for delay in parasite clearance at enrolment The following were found to be independent risk factors for delay in parasite clearance at enrolment (Table 4.4): age ≤ 2 years (Adjusted odds ratio [AOR] = 2.13, 95% confidence interval [CI]1.44-3.15, P < 0.0001), presence of fever (AOR = 1.33, 95% CI = 1.04 –1.69, P = 0.019) and parasitaemia >50,000 asexual parasite/µl of blood (AOR = 2.21, 95% CI = 1.70 - 2.75, P < 0.0001). A history of vomiting w as associated with an increased risk of delay in clearance (crude odds ratio = R1.34Y, 95% CI = 1.07 -1.58, P = 0.009). A IB R 4.3: Risk factors for delay in parasite clearance following iLnitiation of treatment o Following treatment, a body temperature > 38 C aAnd Nparasitaemia > 20,000 asexual parasite/µl of blood a day after treatment begDan, were independent risk factors for delay in clearance (Table 4.5). Non aBrtemAisinin monotherapy was associated with delay in clearance. I O F 4.4: Delay in parasitTe clYearance and treatment failure Figure 4.2 showSs thIe proportions of children treated with various antimalarials who had delay in Rparasite clearance and who subsequently failed treatment. Overall, 291 childIrVen f Eailed treatment. Of these, 211 (72%) had delay in parasite clearance. The lNatter represent 17% of the total children (1237) with delay in parasite clearance. In Usummary 13 of 20, 7 of 10, 119 of 146, 14 of 15, 5 of 19 and 50 of 60 children treated with AQ, COT, CQ, CQCP, MQ, and SP respectively were those who failed treatment and previously had delay in parasite clearance. 89 Table 4.4: Predictors of delay in parasite clearance at presentation in children with acute falciparum malaria treated with antimalarial drugs. Variables Number PC Crude OR P Adjusted OR (95% P enrolled >2 d (95% CI) Value CI) Value Age (years) >2 2446 1076 1 1 ≤2 267 140 1.40 (1.09 –1.80) 0.008 2.13(1.44 –3.15) <0.000 Gender 1 Female 1342 607 1 Y Male 1240 557 0.98 (0.84 –1.15) 0.870 - R - A Fever* Absent 686 252 1 1 R Present 1743 842 1.6 (1.34 -1.93) <0.0001 B1.33 (1.04-1.69) 0.019 I Duration of L illness (d) N < 3 2085 926 1 > 3 521 246 1.12 (0.90 – 1.3D0) A0.225 - - Haematocrit (%) A ≥ 30 614 182 1 B <30 380 114 1.01 (0I.76 – 1.34) 0.904 - - Parasitemia (/µl O F blood) < 50,000 1607 634 1 1 > 50,000 1145 IT60Y3 1.70 (1.46 –1.99) <0.0001 2.21 (1.77 – 2.75) < 0.0001 Gametocytemia S Absent E2R086 896 1 Present V 232 90 0.84 (0.63 – 1.10) 0.224 - - VomitingN I No 1005 519 1 1 Yes U 665 387 1.34 (1.07-1.58) 0.009 1.21 (0.90-1.51) 0.089 Hepatomegaly Absent 471 237 1 Present 798 430 1.54 (0.91 – 2.44) 0.219 - - o CI, confidence interval; OR, odd ratio; PC, parasite clearance, *Body temperature > 37.5 C 90 Table 4.5: Predictors of delay in parasite clearance on day 1 after treatment in children with acute falciparum malaria. Variables Number PC Crude OR P Adjusted OR P enrolled >2 d (95% CI) Value (95% CI) Value Axillary O temperature ( C) <38.0 2326 1012 1 1 Y ≥38.0 228 139 2.02 (1.53 -2.67) <0.0001 1.80 (1.3R0-2.50) < 0.001 Parasitaemia (/µl A blood) B R ≤ 20,000 1328 463 1 1 >20,000 683 467 5.25 (4.20 – 6.48) <0. 0L001I 5.13 (4.14 – 6.35) < 0.001 Drug treatment * CQ 388 275 1 AN1 1 AQ 573 298 0.44 (0.33 – 0D.58) <0.0001 0.79 (0.63 – 0.98) < 0.031 AQAS 142 14 0.05 (0.03A – 0.08) <0.0001 0.24 (0.10 – 0.57) < 0.0001 AQSP 69 26 0.24 (0.14 – 0.42) <0.0001 0.44 (0.27 – 0.73) 0.002 AQSF 91 34 0.25 (0.15 – 0.39) <0.0001 0.43 (0.29 – 0.68) < 0.0001 AMQ 174 10 0.03 F I (0B.01 – 0.05) <0.0001 0.04 (0.02 – 0.09) < 0.0001 AL 90 6 0.03 (0.01 – 0.07) <0.0001 0.05 (0.02 – 0.12) < 0.0001 AS 120 3 O0.01 (0.00 – 0.03) <0.0001 0.02 (0.00 – 0.06) < 0.0001 COT 104 61 0.58 (0.37 – 0.91) 0.017 1.03 (0.68 – 1.36) 0.877 CQCP 315 189Y 0.61 (0.45 - 0.84) 0.002 1.09 (0.83 – 1.42) 0.512 CQKET 70 IT36 0.43 (0.25 – 0.73) 0.001 0.77 (0.47 – 1.25) 0.298 CQSP 107 46 0.31 (0.19 – 0.48) <0.0001 0.54 (0.36 – 0.82) 0.004 MQ 176 S 40 0.12 (0.08 – 0.18) <0.0001 0 . 2 1 ( 0 . 1 5 - 0.31) < 0.0001 SP 29R1 186 0.72 (0.52 – 1.00) 0.055 - - SPPB E42 13 0.18 (0.09 - 0.37) <0.0001 0.33 (0.17 -0.98) 0.001 CI, confidence interval; OR, odd ratio; PC, parasite clearance, * Values of OR represent chances of delay in parasite clearance. † All drugs were IadVministered orally. AQ, amodiaquine; AQAS, amodiaquine plus artesunate; AQSP, amodiaquine plus sulphadoxine-pNyrimethamine; AQSFP, amodiaquine-sulfalene-pyrimethamine; AMQ, mefloquine plus artesunate; AL, artemetherU plus lumefantrine; AS, artesunate; COT, co-trimoxazole; CQ, chloroquine; CQCP, chloroquine plus chlorpheniramine; CQKET, chloroquine plus ketotifen; CQSP, chloroquine plus sulphadoxine- pyrimethamine; MQ, mefloquine; SP, Sulphadoxine-pyrimethamine; SPPB, sulphadoxine-pyrimethamine plus probenecid; 91 Δ Delay in PC Δ Total failure RY A LIB R AN AD F I B O ITYS VE R I FigUureN 4.2: The proportions of children treated with various antimalarials who had delay in parasite clearance (PC) and subsequently failed treatment. (AQ, amodiaquine; AQAS, amodiaquine plus artesunate; AQSP, amodiaquine plus sulphadoxine-pyrimethamine; AQSFP, amodiaquine-sulfalene-pyrimethamine; AMQ, mefloquine plus artesunate; AL, artemether plus lumefantrine; AS, artesunate; COT, co-trimoxazole; CQ, chloroquine; CQCP, chloroquine plus chlorpheniramine; CQKET, chloroquine plus ketotifen; CQSP, chloroquine plus sulphadoxine- pyrimethamine; MQ, mefloquine; SP, Sulphadoxine-pyrimethamine; SPPB, sulphadoxine-pyrimethamine plus probenecid) 92 Study 2: Pharmacokinetic determinants of response to treatment with chloroquine in children with acute uncomplicated Plasmodium falciparum malaria. Chloroquine was well resolved in the analysis using this method. There was a 2 good linearity (r = 0.9992; Figure 4.3 and Appendix Table 4.a) in the standard cu rve obtained for the analysis of chloroquine (CQ). In addition, the percentage rRecovYery of CQ as well as the precision and accuracy of the analytical method for CAQ are shown in Table 4.6 and 4.7. Figure 4.4 shows the chromatogramBs Rfor CQ, desethyl chloroquine (DCQ) or papaverine (PPV), internal standar dL obItained during analysis. There was no interference with other drugs (e.g. paracNetamol, amodiaquine and other commonly used drugs) with the peaks obtaineDd (FAigure 4.4). The limit of detection for chloroquine was 5ng/ml. The retention Atime for DCQ, CQ and the PPV were 5.5 minutes, 6.5 minutes and 10.5 minute F sI rBespectively. O TY RS I VE NIU 93 10.0 7.5 5.0 RY 2.5 RA 0.0 IB 0 500 1000 1500 L2000 2500 Concentration (AngN/ml). AD Figure 4.3: Standard curve of chloroq uIiBne for extrapolation of unknown 2 concentrations from whole blood sFpotted on filter paper. (r = 0.9992) O TY RS I E IV UN 94 Peak height ratio Table 4.6: Percentage recovery of chloroquine from whole blood spotted on filter paper using the analytical method for chloroquine. Concentration % Recovery n (ng/mL) (S.D) Y Whole blood 200 85.65.9 5 400 87.52.5 4 R 800 89.61.9 4 A R B S.D= Standard deviation N LI DA BA OF I SI TY R IV E UN 95 Table 4.7: Precision of analytical method for extraction of chloroquine from whole blood spotted on filter paper. Concentration C.V (%) n (ng/mL) Intra-assay Chloroquine Y 100 2.9 5 200 1.9 5 R 1000 2.5 4 A R Inter-assay Chloroquine B 100 3.2 L5I 200 3.1 6 1000 2.8 4 C.V= coefficient variation AN BA D OF I ITY ER S NI V U 96 A IS AR Y BB R N LI IS DA BACQ IDCQ O F SI TY VE R NI Time (min) UFigure 4.4: Chromatograms showing chloroquine (CQ) and desethyl chloroquine (DCQ) from whole blood sample collected from a patient and spotted on filter paper on day 0 (A) and day 3 (B). 97 4.5: Study Population The characteristics of the 74 children who were randomly selected into the study are shown in Table 4.8. Thirty eight (51.4%) children were aged < 5 years. The geometric mean parasite density was 52,467 (95% confidence interval, 38,194 – 71,944 asexual parasite/µl of blood). Seventy eight percent (58/74) of the children were febrile at enrolment. Y 4.6: Pharmacokinetics parameters of all patients enrolled in the study R All the patients or guardians gave a negative history of chloRroqAuine ingestion in the preceding two week but analysis of the pre-treatment IbBlood sample showed substantial chloroquine levels in 43 (58.1%) of the 7N4 ch i Lldren enrolled. The mean CQ concentration was 916.86±89.79ng/ml (range,A 25.6 – 2643.0ng/ml) (Table 4.8). The children were therefore divided into 4 AgrouDps for the analysis of the data; all the 74 children that were enrolled into tIhBe study; 48 children with delay in parasite clearance, 43 children with pre-dFosin g detectable chloroquine concentration in their blood and 31 children with n oO chloroquine in the pre-treatment blood sample. Drug conceInTtratYion before drug administration on day 2 (48h) was 1740.81±137.69nSg/ml and the decline of chloroquine concentration after day 2 was mono expEonRential with a terminal half-life (t1/2) of 306.3±20.99h (95% CI, 264.4 - 348.I1Vh). Table 4.9 is the summary of pharmacokinetic parameters of all patients iNrrespective of pre-dosing chloroquine blood concentration at enrolment. The AUC0-U28d and CL were 920.6±69.6ng.h/ml and 633.6±60.2ml/h respectively. 98 Table 4.8: Baseline clinical and demographic characteristics of seventy-four children with acute uncomplicated malaria treated with standard doses of chloroquine. Parameters Mean±SD 95% CI Number enrolled 74 - Age (y) 5.2 ± 2.9 4.5 - 5.9 Range Y0.5 - 12.0 - R No with age < 5 y 38 (51.4%) - A Gender ( M:F) 38:36 B -R Weight (kg) 16.8 ± 6.5 LI15.2 - 18.3 Range 5.0 - 42.0 N - O Axillary Temperature ( C) 38.4 ± 1.1A 38.1 – 38.6 Range 36A.3 -D 40.5 - No. with Fever IB58 (78.4%) - Haematocrit (%) F 30.6±5.9 29.2 – 32.1 Range O 20.0 – 46.0 - No. with PCV < T30%Y 27 (36.5%) - ParasitaemSia/µIl (GM) 52,467 38,194 – 71,944 Range R 2,156 - 404,666 - VCQE concentration (ng/ml) on 916.8±89.7* 735.6 – 1098.0 NI day 0 (n=43) U Range 25.6 – 2643.0 O Fever, temperature ≥ 37.5 C; * Mean±s.e.m; CI, confidence interval; GM, geometric mean 99 Table 4.9: Pharmacokinetic parameters of chloroquine determined in children with falciparum malaria and treated standard doses of chloroquine. Parameters Mean ±s.e.m Y t1/2 (h) 306.3 ± 20.9 R 95% CI 264.4 – R348.A1 Range 97I.2B – 958.3 AUC0-28d (ng.h/ml) L920.6 ± 69.6 95% CI AN 781.9 – 1059.3 Range D 185.8 – 3509.2 CL (mL/h) BA 633.6 ± 60.2 95% CI I 513.6 – 753.5 Range F O 69.0 – 2579.0 Cl= clearance, AUC0-28d =Y area under concentration-time curve from day 0 -28, t 1/2= half-life IT ER S IV UN 100 4.7: Comparison of blood chloroquine concentrations and pharmacokinetic parameters in children with early or delay in parasite clearance. Figure 4.5 illustrates whole blood chloroquine concentrations versus time in those children with early parasite clearance or delay in parasite clearance. In these patients day 1 chloroquine concentrations were 1169.06±167.21 and 1958.23±213.80ng/ml, respectively. The difference between mean day 1 chlorYoqu ine concentration was not significant (P = 0.061) but higher in children witRh delay in parasite clearance. There was no correlation between delay Rin Aparasite and chloroquine concentration (P = 0.084, r = 0.237). Table 4.1I0 Bis a summary of the pharmacokinetic parameters of CQ in the children with eLarly parasite clearance or delay in parasite clearance. There was no differAentN between the pharmacokinetic parameters of chloroquine in children who hadD delay in parasite clearance compared to those whose parasites cleared earIlyB fr Aom the peripheral blood. Although the chloroquine half-life in childreFn w ith delay in parasite clearance was higher compared to those that clea reOd parasite early (321.3±27.1 versus 219.6±25.3h, P = 0.065). ITY ER S IV UN 101 2500 Early PC Y 2000 DelaAy PRC 1500 R 1000 LI B N 500 DA 0 A 0 5 10 B15 20 25 30 F I Time (days) Figure 4.5: Mean wholeY bl o Ood concentration-time curves of chloroquine following oral administration ofT 25mg/kg of chloroquine base over 3 days in children infected with PlasmodiumS faIlciparum malaria with early or delay in parasite clearance (PC). VE R NIU 102 Chloroquine concentration (ng/ml) Table 4.10: Pharmacokinetic parameters of chloroquine in children with acute uncomplicated malaria who had delay in parasite clearance after treatment with standard doses of chloroquine RY Parameters Children with Children withR AP value PC ≤ 2d PC > 2d IB n=13 n= 48 L t1/2 (h) 219.69 ± 25.35 3N21.35 ± 27.13 0.065 Range 108.0 – 379.9D A97.2 – 958.3 AUC0-28d (ng/ml.h) 728.58B ± A122.99 1011.92 ± 95.17 0.150 Range F198 .7I – 1577.5 185.8 – 3509.1 CL (mL/h) 616.84 ± 92.37 612.15 ± 75.65 0.976 Range OY 215.0 – 1505.1 69.0 – 1952.2 Mean±ISTEM, *significant value ER S IV UN 103 4.8: Comparison of pharmacokinetic parameters in children with or without pretreatment chloroquine concentration. Figure 4.6 illustrates whole blood chloroquine concentrations versus time in those children with or without CQ in their blood at enrolment. In these patients day 1 chloroquine concentrations were 1741.87±209.16 and 1640.35±226.95ng/ ml, respectively. The difference between mean day 1 chloroquine concentratioRn wYas not significant (P = 0.749). Day 2 concentrations were also similar: 1703.9A8±172.92 and 1796.90±231.34ng/ml, respectively. The difference betweenB daRy 2 chloroquine concentration was not significant (P = 0.715). Ta blLe I4.11 summarizes the pharmacokinetic parameters of children with and wNithout detectable chloroquine concentration pre-dosing. Despite presence ofD CQA in their blood pre-treatment there was no difference in the pharmacokinBeticA parameters between patients with pre-treatment CQ levels and those withou t Idetectable pre-dosing chloroquine. OF ITY ER S IV UN 104 2500 RY CQ absent 2000 ACQ presBentR 1500 LI 1000 AND 500 BA I0 0 5 10 F 15 20 25 30 O Time (days) TY Figure 4.6: MeaSn wIhole blood concentration-time curves of chloroquine in children infected withR Plasmodium falciparum malaria treated with oral doses of 25mg/kg chloroquiEne base over 3 days with or without chloroquine in their blood at enrolment. IV UN 105 Chloroquine concentration (ng/ml) Table 4.11: Pharmacokinetic parameters of chloroquine in children with an d without pretreatment chloroquine levels following treatment with stRandYard doses of chloroquine (n=74). RA Parameters Children without Children with IBP value chloroquine on chloroquine oLn day 0 day 0 N n=31 n=43 A t1/2 (h) 276.30 ± 23.94 A32D7.90 ± 31.57 0.228 Range 97.2 – 666.4 IB 108.0 – 958.3 AUC0-28d (ng/ml.h) 877.64 ± 1F25.86 951.60 ± 79.25 0.648 Range 1Y85.8 O– 3509.1 227.9 – 2640.0 CL (mL/h) IT666.77 ± 103.53 610.41 ± 73.09 0.604 Range S 143.1 – 2579.0 69.0 – 1952.2 Pre-dosing R CQ 916.86 ± 89.79 concIeVntr Eation - - (ng/ml) 25.6 – 2643.0 URNange Mean±s.e.m, *significant value 106 4.9: Comparison of clinical parameters and treatment outcome in children with or without pretreatment chloroquine concentration. Table 14.12 shows, the clinical characteristics and response to treatment in children with or without pretreatment CQ. Of the 43 or 31 children with or without CQ before treatment, 23 (53.5%) or 16 (51.6%) responded to treatment, respectively. In children with or without pretreatment CQ, 11 (25.6%), 6 (13.9%) and 3 (7.0%Y) o r 9 (29.0%), 4 (13.0%) and 2 (6.4%) had RI, RII and RIII responses, reRspectively. Response to treatment in children with or without pretreatment CQR werAe thus similar 2 (χ =0.01, P=0.9389). B N LI A 4.10: Comparison of blood chloroquine concDentrations in children with sensitive and resistant responses to chloroquineB. A I Table 4.13 is a summary of the wFho le blood CQ concentrations following treatment in the children with sensiti veO or resistant responses to chloroquine. Mean whole blood CQ concentraItiTon Yon day 3 was significantly higher in children with sensitive response compaSred with children with resistant responses (1856.7±204.7 vs 1276.3±1E38.R2ng/ml, P = 0.011). IV UN 107 Table 4.12: Comparison of clinical parameters and treatment outcome in children with acute uncomplicated malaria who had or without pretreatment chloroquine concentration (n=74). Parameters Children without Children with P value chloroquine on day 0 chloroquine on day 0 Y n= 31 n= 43 R Age (y) 5.0 ± 2.6 5.3 ± 3.1 0.A64 Range R 1.0 – 12.0 0.5 – 12.0 IB Sex (M:F) 17:14 21:22 L Weight (Kg) 15.9 ± 5.2 17.3 A± 7.N2 0.38 7.5 – 30.0 6.D0 – 42.0 FCT (d) 1.8 ± 1.1 BAI 1.9 ± 1.0 0.56 1 – 5 F 1 – 5 PCT (d) 3.3 ± 1. 1 O 3.5 ± 1.3 0.56 2.0 –Y 7.0 2.0 – 6.0 Parasitaemia/µl TS I50,839 53,987 0.65 (GMPD) R 3,125 – 313,538 2,156 – 404,666 Range RespIoVnse E SNensitive 16 (51.6%) 23 (53.5%) 0.77 URI 9 (29.0%) 11 (25.6%) RII 4 (13.0%) 6 (13.9%) RIII 2 (6.4%) 3 (7.0%) GMPD, geometric mean parasite density, Mean±s.d 108 Table 4.13: Whole blood chloroquine concentrations in children with uncomplicated falciparum malaria with sensitive or resistant response after administration of 25mg/kg chloroquine base (n=74). *Mean Time Whole blood chloroquine concentration (ng/ml) whole (days) blood Sensitive n=39 Resistant n=35 P value chlor oqu 0 496.47 ± 97.33* 573.22 ± 114.33 0.609 iYne 0.0 – 2643.0** 0.0 – 2119.0 Rconcentr ation 1 1814.96 ± 219.88 1560.11 ± 212.00 0.687A ±s.e.m, 375.7 – 5192.0 74.6 – 4680.8 BR ** 2 1873.61 ± 223.98 1592.08 ± 148.76 I 0.312 Range; 500.7 – 5378.1 732.9 – 4166.0 L RI= 3 1856.65 ± 204.70 1276.32 ± 1A38N.22 0.011 parasitaeD mia 472.5 – 5776.3 135.0 – 3086.2 disappea 7 1427.26 ± 254.75 910A.20 ± 89.14 0.087 ra but 43.7 – 5920.3 IB103.6 – 1892.4 reappear 14 1214.36 ±170.27 1131.82 ± 244.01 0.789 s within 335.8 – 2319.6 O F 254.5 – 1625.6 7 to 14 days; 21 1090.23 ± Y259.37 757.24 ± 147.57 0.308 RII= 77.7 – 2T994.4 248.5 – 1625.9 decrease 28 86S9.94I ±165.21 609.50 ± 120.68 0.328 of R34.6 – 2096.3 151.6 – 1067.0 parasitae OutcomeE mia but no SensIitVive 39 complet RNI - 20 (57.1%) e URII - 10 (28.6%) clearanc RIII - 5 (14.3%) e from peripher al blood; RIII= no pronounced decrease or increase in parasitaemia at 48hr after treatment; S= sensitive. 109 4.11: Comparison of chloroquine pharmacokinetic parameters in children whose infection responded or failed to respond to treatment. Table 4.14 is a summary of the pharmacokinetic parameters of the children with sensitive or resistant responses to chloroquine. The AUC0-28d of chloroquine was significantly higher in children with sensitive response compared with those with resistant responses (1052.1 ± 116.3 vs 774.1± 62.9 ng/ml.h, P = 0.01). The AUYC0 -28d values in children with sensitive response was approximately 1.5 fold Rthose with resistance response (Figure 4.7 and 4.8). RA LI B 4.12: Pharmacokinetic risk factors associated Nwith chloroquine treatment failure DA Table 4.15 shows the univariate and mBultAivariate analysis of pharmacokinetic risk factors for chloroquine treatmenFt fa ilIure. Day 3 CQ concentration ≤ 1750ng/ml {Crude odd ratio (COR), 4. 0O8; 95% CI, 1.13 – 14.64; P = 0.027} and AUC0-28d ≤ 950ng/ml.h (COR, 2.8T9; Y95% CI, 1.05 –7.93, P = 0.037) were significantly associated with a risk of treSatmIent failure. R HowIeVver E, if a P value less than 0.3 was taken (‘falsed’) to indicate significant dNifference in a univariate analysis, a terminal elimination half-life less than 220h U{Adjusted odds ratio (AOR), 0.28; 95% CI, 0.08 – 0.98, P = 0.047}, and an AUC 0- 28d less than 950ng/ml.h (AOR, 4.12; 95% CI,1.09 – 15.52, P = 0.036} were independent predictors of chloroquine treatment failure (Table 4.16). 110 Table 4.14: Pharmacokinetic parameters of chloroquine determined in children with acute uncomplicated falciparum malaria who had sensitive or resistant responses after treatment with standard doses of chloroquine (n=74). Parameters Sensitive n=39 Resistant n=35 P value t1/2 (h) 293.8 ± 28.8 320.2 – 31.0 0.97 Y 95% CI 235.5 – 352.0 257.2 – 383.2 R Range 97.2 – 958.3 114.0 – 875.0 A BR AUC0-28d (ng/ml.h) 1052.1 ± 116.3 774.1 ± 62.9L I 0.01* 95% CI 816.6 – 1285.0 646.33 – 9 01.8 Range 185.8 – 3509.2 202 D A .9 –N 1890.8 CL (mL/h) 647.9  84.5 A 617.1 ± 86.7 0.46 95% CI 476.9 – 818 .9I B 440.7 – 793.6 Range 69.01 – 1F952.2 107.2 – 2579.0 Mean±s.e.m, * significan tO SI TY R IV E UN 111 2500 Y All patients R RespondedA to treatment 2000 Fail LIB ed Rtreatment N 1500 DA 1000 A IB 500 O F 0 SI TY 0 5 10 15 20 25 30 ER Time (days) V FNiguIre 4.7: Mean concentration-time curve of chloroquine for all patients, whose Uinfection responded or failed to respond to treatment after oral standard doses chloroquine base given over 3 days. 112 Chloroquine concentration (ng/ml) Table 4.15: Pharmacokinetic predictors of treatment failure in children with acute uncomplicated malaria treated with standard doses of choroquine. No of children Univariate analysis Logistic regression analysis Examined Failing Crude odds ratio p value Adjusted odds P treatment (95% confidence ratio (95% CRI) YValue by day 14 interval) CQ A concentration R (ng/mL) on day 3 IB >1750 18 4 1 L1 ≤1750 39 21 4.08 (1.13 – 14.64) 0.0N27 3.25 (0.86 – 12.23) 0.081 t1/2 (h) A >220 42 22 1 D ≤ 220 32 13 0.62 (0.24 – 1A.57) 0.300 - - AUC0-28d (ng/ml.h) B I >950 F 26 8 1 1 ≤ 950 48 27 O2.89 (1.05 –7.93) 0.0307 3.25 (0.93 – 11.27) 0.063 ITYS VE R NIU 113 Table 4.16: Pharmacokinetic risk factors of treatment failure in children with uncomplicated malaria treated with standard doses of chloroquine (P value 0.3 is considered). No of children Univariate analysis Logistic regression analysis Examined Failing Crude odds p Adjusted odds P treatment ratio (95% value ratio (95% CI) Value by day 14 confidence interval) CQ Y concentration R (ng/mL) on day 3 A >1750 18 4 1 1 R ≤1750 39 21 4.08 (1.13 – 14.64) 0.027 3.057I B(0.77 - 12.13) 0.112 t1/2 (h) N L >220 42 22 1 1 @ ≤ 220 32 13 0.62 (0.24 – 1.57) 0.3A00 0.28 (0.08 – 0.98) 0.047* AUC0-28d (ng/ml.h) D A >950 26 8 1 IB 1 ≤ 950 48 27 2.89F (1. 05 –7.93) 0.030 4.12 (1.09 – 15.52) 0.036* @false to MultiOple regression; * significant ITY S VE R I UN 114 . CQ present CQ absent AR Y BR LI DA N A IB TimFe (d ays) Figure 4.8: Decline of chloroqOuine concentration in children with or without chloroquine in their pre-tYreat ment whole blood samples following oral administration of 25mg/kg of chlorIoqTuine base over 3 days. S VE R UN I 115 Chloroquine concentration (ng/ml) Study 3: Development of a simple cost effective high performance liquid chromatography assay of sulphadoxine in whole blood spotted on filter paper for field studies. 4.13: Chromatography techniques Standard curves from sulphadoxine spiked blood added to filter paper wereY linear over the concentration ranged studied. Linear regression analysis yieldAed cRorrelation coefficient r > 0.99 (n = 6, Figure 4.9). Sulphadoxine peak waBs wRell resolved from the internal standard (sulizoxasole) at the calibration rLangIes of 0 – 60 µg/ml (Appendix Table 4.b). The retention times (tR) of sulpNhadoxine and internal standard (IS) were 6.3 and 7.2min respectively (Figure D4.10A). The separation chromatograms of sulphadoxine and the internal standaArd from spiked whole blood samples corresponded with those of blood sa mIpBles obtained from a patient at time 0 before drug administration and day O3 afFter an oral standard single dose of sulphadoxine- pyrimethamine (SP) (25Ymg /kg body weight of sulphadoxine and 1.25mg/kg body weight of pyrimethaImTine (Figure 4.10 A, B and C respectively). S ER NI V U 116 3 2 Y R 1 BR A N LI 0 0 10 20 30 4D0 A 50 60 70Sulphadoxine concAentration (µg/ml)B F I Figure 4.9: Sulphadoxine Ostandard curve for extrapolation of sulphadoxine 2 concentrations from unknYown blood sample spotted on filter paper (r =0.99518). IT ER S NI V U 117 Peak height ratio Chromatogram - Cgm006 30-11-05 cali .20.0ug ( \siju\ ) 1125 984 A 844 2 1 703 Y 562 R 00:00 02:24 04:48 07:12 09:36 12:00 Chromatogram - Cgm015 11-01-06 mc125d0 ( \siju\ ) Time [mm:ss] A 640 R B 2 B 600 N LI 559 A 518 D 478 A 00:00 02:24 04:48 07:12 09:36 12:00 Time [mm:ss] B Chromatogram - Cgm010 06-01-06 mc135d1 ( \siju\ ) F I713 C O 630 ITY 2 546 RS 1 462 VE N37I8 00:00 02:24 04:48 07:12 09:36 12:00U Time [mm:ss] Figure 4.10: Chromatograms showing peaks after extraction of sulphadoxine (1) and internal standard (2) from drug free whole blood spiked with 60µg/ml sulphadoxine (A), day 0 (B) and day 3 (C) of sample collected from a patient who was administered with a standard dose of sulphadoxine/pyrimethamine. 118 Signal [mV] Signal [mV] Signal [mV] 4.14: Recovery, calibration curves and reproducibility The extraction recoveries for 25µg/ml, 60µg/ml and 100µg/ml of sulphadoxine were 82.66±4.0 (n=6), 81.02±3.24 (n=5) and 85.60±1.9 (n=5) per cent respectively (Table 4.17). The intra-day recovery deviation at 60µg/ml and 100µg/ml of SDX were 3. 7% and 4.6% respectively (n=5) (Table 4.18). The intra-day precision was <5.0Y% and inter-day accuracy ranged from 4.1 to 5.3% respectively. The limit oAf deRtection of sulphadoxine defined as a concentration giving a peak four times theR baseline noise in all biological fluids was 120ng/mL at 0.05 absorbance units full Bscale (aufs). N LI A BA D F IO ITY RS VE UN I 119 Table 4.17: Percentage recovery of sulphadoxine from whole blood samples collected on filter paper. Concentration % Recovery N (µg/mL) S.D Y Whole 25 82.64.0 6 R blood A 60 81.03.2 5B R 100 85.61.9 LI5 SD= standard deviation AN AD F I B O SI TY ER NI V U 120 Table 4.18: Precision of analytical method for sulphadoxine determination using whole blood spotted on filter paper. Concentration C.V (%) n (µg/mL) Intra-Assay Sulphadoxine 60 3.73.0 5 Y 100 4.62.5 5 AR R Inter-Assay Sulphadoxine IB 10 6.02.0 LN 5 100 4.12.0A 6 CV= coefficient of variation D BA OF I ITYS VE R NIU 121 4.15: Interference There was no interference from endogenous compounds in the biological sample. There was no interference with the peaks of sulizoxasole (IS) and other commonly used antimalarial, analgesic and anti-infective drugs. 4.16: Clinical application RY The concentration-time curve in whole blood of a patient who was admAinistered with a standard single oral dose of sulphadoxine-pyrimethamine RI B(SP) (25mg/kg body weight of sulphadoxine and 1.25mg/kg body weight of py rLimethamine i.e 500mg of sulphadoxine and 25mg of pyrimethamine) is shNown in Figure 4.11. The pharmacokinetic parameters of SDX in the indiDvidAual are shown in Table 4.19. Peak blood concentration of 212.02µg/ml BwasA reached after day 1. The calculated elimination half-life was 3.50d wFhil e Ithe areas under the concentration time curve (AUC0-28d) were 884.84µg/m lO.d. The profile show the applicability of the method for measuring SDX in whoYle blood dried on filter. The pharmacokinetic result agrees with previous reSportIs T (Dzinjalamala et al., 2005). The method was not sensitive to detect pyrimRethamine, although pyrimethamine peak was detected and separated duringV thEe standard calibration preparations. This insensitivity may be due to the fact tNhat Iamount of pyrimethamine in tablet was small (1:20 of sulphadoxine) and only a Usmall volume of blood used for the analysis. 122 250 RY 200 BR A 150 LIN 100 DA 50 IB A 0 OF0 ITY 10 20 30 Time (days) Figure 4.11: CoSncentration–time curve for sulphadoxine in whole blood following single oraEl doRse of sulphadoxine-pyrimethamine (25mg/kg body weight of sulpIhaVdoxine and 1.25mg/kg body weight of pyrimethamine). UN 123 Sulphadoxine concentration (g/ml) Table 4.19: Demographic and pharmacokinetic parameters of a patient who received a standard dose of sulphadoxine-pyrimethamine. Parameters Mean Age (year) 6.0 Y Weight (Kg) 16.0 R A t 1/2 (d) 3.50 BR Tmax (d) 1.00 LI Cmax (µg/ml) 212.02 N A AUC0-28d (µg/ml.d) 88D4.84 Cl (ml/d) A IB 524.50 AUC F0-28, area under concentratioOn time –curve from time 0 to day 28, Cmax, peak blood concentration; Tmax, time tYo p eak blood concentration; t1/2 elimination half-life; Cl, clearance.T RS I IV E UN 124 Study 4: Pharmacokinetic disposition of sulphadoxine in children with uncomplicated P. falciparum malaria treated with standard dose of sulphadoxine-pyrimethamine. 4.17: Study population A total of 74 patients with acute uncomplicated malaria were randomly selected i nto the study. The clinical and demographic parameters of the children areR shoYwn in Table 4.20. The geometric mean parasite density was 48,953 (RangRe, 2A011- 461,333 asexual parasite/µl of blood). The mean age of the children waIsB 5.07±3.02 years and their mean weight was 16.55±7.7kg. L AN AD F I B O ITY RS VE UN I 125 Table 4.20: Baseline clinical and demographic characteristics of children with acute uncomplicated falciparum malaria treated with standard dose of sulphadoxine-pyrimethamine (n=74). Parameters Mean±SD 95% CI Number enrolled 74 - Y Age (y) 5.2 ± 3.0 4.5 - 5.9 R Range 0.5 - 12.0 R- A No with age < 5year 38 (51.4%) IB - Gender ( M:F) 35:39 L - Weight (kg) 16.9 ± 7.A6 N 15.1 - 18.7 Range 4.0 –D 38.0 - O Axillary Temperature ( C) B3A8.2 ± 1.0 37.2 – 38.4 Range F I 36.1 - 40.5 - No. with fever O 57 (77.0%) - Haematocrit (%T) Y 30.7±4.0 29.1 – 32.3 Range SI 25.0 – 38.0 - No. wiRth PCV < 30% 27 (36.5%) - IVPar Easitaemia/µl (GM) 48,953 N Range 2011- 461,333 - U SDX Concentration (µg/ml) on 8.9 ± 3.5* (n=13) day 0 Range 2.6 – 168.3 O Fever, temperature >37.5 C; * Mean±s.e.m; CI, confidence interval; GM, geometric mean 126 4.18: Pharmacokinetic parameters of sulphadoxine in children with uncomplicated falciparum malaria after administration of standard dose of sulphadoxine-pyrimethamine. Thirteen patients (17.6%) had pretreatment levels of SDX and their data were excluded from the final analysis. The mean SDX concentration was 8.9±3.5µg/ml (range, 2.6 – 168.3µg/ml) (Table 4.20). The patients were therefore divided Yinto 4 groups for the analysis of the data; the 74 children that were enrolled intoR the study; 45 with delay in parasite clearance; 13 with detectable preR-treAatment SDX concentration and 61 children without SDX before treatment. LIB N Table 4.21 is the summary of the pharmacokDineAtic parameters of all the patients irrespective of pre-treatment SDX blood coAncentration at enrolment. The mean tmax and elimination half-life of SDX in ca pIiBllary whole blood were 2.1±0.1 and 5.5±0.3d. The peak whole blood conceOntraFtion (Cmax) ranged from 25.5 to 745.69µg/ml with mean of 214.9±16.9µg/mYl. The AUC0-28d varied between 91.19 and 3768.62µg/ml.d (mean 1252.9±106.1IµTg/ml.d). The clearance was 437.1±39.9ml/d (ranged, 86.2 – 1555.3). RS IV E N U 127 Table 4.21: Pharmacokinetic parameters of sulphadoxine in seventy-four children treated with standard dose of sulphadoxine-pyrimethamine. Parameters Mean ±s.e.m Cmax (µg/ml) 214.9 ± 16.9 Range 25.5 – 745.69 Y Tmax (d) 2.1 ± 0.1 AR Range 1 – 5 R t1/2 (d) 5.5 ± 0.3I B Range 1.1 – 1L7.0 AUC0-28d (µg/ml.d) A125N2.9 ± 106.1 Range AD 91.1 – 3768.6 CL (mL/d) IB 437.1 ± 39.9 Range F 86.2 – 1555.5 Day 3 SDX concentration (µOg/ml) 184.9 ± 20.0 Range TY 19.7 – 856.9 Day 7 SDX concentIration (µg/ml) 85.0 ± 11.2 Range RS 3.0 – 368.9 DayI 1V4 S EDX concentration (µg/ml) 43.9 ± 6.6 RNange 3.7 – 151.6 U 128 4.19: Comparison of blood sulphadoxine concentrations and pharmacokinetic parameters in children with early parasite clearance or delay in parasite clearance. Figure 4.12 illustrates whole blood sulphadoxine concentrations versus time in th ose children with early parasite clearance or delay in parasite clearance. In these pYatients day one sulphadoxine concentrations were 127.0±19.5 or 215.2A±25R.5 µg/ml, respectively. The difference between mean day 1 SDX cRoncentration was significantly (P = 0.049) higher in children with delay in LparIasBite clearance. Table 4.22 is a summary of the pharmacokinetic parameterNs of SDX in the children with early parasite clearance or delay in parasite cleaArance. There was no difference between the pharmacokinetic parameters ofA suDlphadoxine in children who had delay in parasite clearance compared to th oIsBe that their parasites cleared early from the peripheral blood. Afterwards, thFe concentrations of SDX in children who cleared parasite early were higheYr, a lt Ohough not statistically significant. The mean maximum concentration of SDIXT (Cmax) obtained in children who cleared parasite early was high but not significanSt. ER NI V U 129 300 Early PC Y Dealy PC AR 200 LIB R N 100 A BA D 0 I 0 3 6 9 F 12 15 18 21 24OTime (days) ITY Figure 4.12: Mean whole blood concentration-time curves of sulphadoxine in children infeRctedS with Plasmodium falciparum malaria with early or delay in parasite clearance. VE UN I 130 Sulphadoxine concentration (µg/ml) Table 4.22: Comparison of pharmacokinetic disposition of sulphadoxine in children with and without delay in parasite clearance (n=65) Parameters Children with PC Children with P value ≤ 2d PC > 2d (n=20) (n= 45) RY tmax (d) 1.95 ± 0.23 2.09 ± 0.15 0A.611 Range R 1.0 – 4.0 1.0 – 4.0 IB Cmax (µg/ml) 218.49 ± 38.65 194.1N0 ± L18.64 0.522 Range 137.5 – 299.4 D1A56.4 – 231.7 t1/2 (d) 5.01 ± 0.53 BA 5.59 ± 0.55 0.524 Range 1.4 – 1F1.0 I 1.16 – 17.0 AUC0-28d (µg/ml.d) 1 1O02.45 ± 187.78 1011.92 ± 95.17 0.150 Range ITY91.1 – 2568.9 185.8 – 3509.1 CL (mL/d)R S 616.84 ± 92.37 612.15 ± 75.65 0.976 Range IV E 215.0 – 1505.1 69.0 – 1952.2 Mean±s.e.m, *significant value UN 131 4.20: Comparison of disposition of sulphadoxine in children with and without detectable blood levels of sulphadoxine at enrolment before treatment. Figure 4.13 illustrates whole blood sulphadoxine concentrations versus time in those children with or without SDX in their blood at enrolment. The mean concentration of SDX at presentation in 13 patients who had detectable SDX level before treYatm ent was 8.90±3.5µg/ml (range 2.99 - 168.3µg/ml). In children with or withoRut SDX in their blood at enrolment, day 1 SDX concentrations were R18A8.94±22.2 or 274.04±71.4µg/ml (P=0.225) respectively. Day 2 concentratiIoBns were also similar; 211.57±18.1 or 261.69±45.4µg/ml (P=0.343) betweenN chi ld Lren with or without SDX in their blood at enrolment. Comparison of dispAosition of SDX in children with pretreatment SDX levels and those without ASDXD showed a similar disposition profile; Cmax (P=0.325), Tmax (p=0.128), Cl (PI=B0.377), day 3 concentration (P=0.343), day 7 concentration (P=0.249), and daFy 14 SDX concentration (P=0.245). Table 4.23 summarizes the pharmacoki neOtic parameters of children with and without detectable sulphadoxine concentTratiYon pre-treatment. However, a trend towards a shorter mean elimination half-SlifeI (3.91±0.40d versus 5.85.69±0.44d, P=0.063) and a higher AUC0- 28d (1660E.33R±172.59 versus 1161.62±121.39 µg/ml.d, P=0.069) was observed in patieInVts with pre-treatment SDX levels. UN 132 Table 4.23: Pharmacokinetic parameters of sulphadoxine in children with and without sulphadoxine in pretreatment whole blood samples. Parameters Children without Children with P values sulphadoxine on sulphadoxine on day 0 (n=61) day 0 (n=13) t1/2 (d) 5.85±0.44 3.91±0.40 0.063 Range 1.16-17.01 2.31-7.04 Cmax (µg/mL) 207.75±19.31 254.10±27.24 0.325 Range Y25.5-856.9 112.6-413.6 R t 0.128 max (d) 2.19±0.14 1.66±0.28 A Range 1.0-5.0 0.0-3.0 R -1 AUC0-28d (µgmL .d) 1161.62±121.39 1660.33±17I2.B59 0.069 Range 91.1-3768.6 221.6-2 5L40.9 Cl (mL/d) 454.26±47.19 36N3.16±59.03 0.377 Range 86.2-1555.3 DA96.0-936.58 Geometric mean 47081.70 59293.28 0.925 (parasite/µL blood) A Range 2011 -4 6IB 1333 6973-33684 Day 0 Conc. (µg/ml) - OF 8.90±3.54 - Range 2.62-168.32 Day 3 Conc. (µgT/mlY) 175.81±22.59 225.17±42.67 0.343 Range I 19.71-856.98 60.45-636.54 Day 7 ConcS.(µg/ml) 78.86±13.07 112.74±47.44 0.249 Range ER 19.71-856.98 66.00-205.32 IRVesponse ACPR 50 (82.0%) 6 (46.2%) N ETF 2 (3.2%) 2 (15.4%) U LCF 9 (14.8%) 5 (38.4%) Values are in Mean±s.e.m 133 Y 350 300 SDX abseRnt A R 250 SDXI pBresent L200 N 150 DA 100 50 IB A 0 F 0 5 O 10 15 20 Time (days) ITY Figure 4.13: MeSan whole blood sulphadoxine concentration-time curves in children with PlasEmoRdium falciparum malaria with or without sulphadoxine in their blood at enroIlmVent. UN 134 Sulphadoxine concentration (µg/ml) 4.21: Pharmacokinetics of sulphadoxine in 61 children without pretreatment sulphadoxine at enrolment. The characteristics and demographic parameters of the 61 children without pretreatment SDX are shown in Table 4.24. Thirty one (51.6%) children were aged < 5 years. The geometric mean parasite density was 472,467 (range 2011 - 461,333 asexual parasite/µl). Infection in 50 (81.9%) of the patients exhibited adequ ate clinical response (ACPR), whereas 3.3% (2/61) of the patient exhibRitedY early treatment failure (ETF) and 14.8% (9/61) had late parasitological faiRlureA (LPF). B LI DA N A F I B O SI TY R IV E UN 135 Table 4.24: Clinical and laboratory data at enrolment of children with acute, uncomplicated falciparum malaria treated with standard dose of sulphadoxine- pyrimethamine without pre-treatment sulphadoxine levels. Parameters Sulphadoxine Number enrolled 61 Y Gender (no. of males: no of females) 31:30 R Age (yr) 5.16±3.0 A Range 0.5-12.0 BR Weight (kg) 16.04±6.9 LI Range 4.0-38.0 O Axillary Temperature ( C) 38.16±1.1 N Range 36.1D-40A.5 Geometric Mean (Asexual 4A7,081 parasite per µl of blood) B Range F I 2,011-461,333 Cure rate on day 14 (% ) O ACPR Y 81.9 (50/61) ETF IT 3.3 (2/61) LPF S 14.8 (9/61) Values are inR Mean±Standard deviation, IV E UN 136 4.22: Comparison of blood sulphadoxine concentrations in children with sensitive and resistant responses to sulphadoxine-pyrimethamine. Table 4.25 is a summary of the whole blood SDX concentrations following treatment in children whose infection were sensitive or resistant to sulphadoxine- pyrimethamine. There was no difference in whole blood sulphadoxine concenYtrat ion in children whose infection were sensitive or resistant to treatment. FRigure 4.14 shows the mean concentration-time curves for the disposition of RSDAX in children whose infections were sensitive or resistant to SP treatmeInBt. There was inter- individual variation in the achieved sulphadoxine concNentr at Lion in the children. DA 4.23: Comparison of disposition of sulAphadoxine in children whose infection responded or failed to respo nIdB to standard dose of sulphadoxine- pyrimethamine. F O Table 4.26 is a summarYy of the pharmacokinetic parameters of the children whose infection were s TSensIitive or resistant to sulphadoxine-pyrimethamine. The t1/2 of sulphadoxineR in children whose infections were sensitive or resistant to SP was 5.98±V0.5 Eor 5.26±1.0d respectively. The Cmax of SDX ranged from 42.1 to 7N45.I69µg/mL and 25.5 to 565.12µg/mL in patients whose infections were sensitive Uand resistant to sulphadoxine-pyrimethamine. There was no difference between the mean peak sulphadoxine concentration (Cmax) in patients who failed to respond to treatment (163.09±30.3µg/mL) and those whose infection were sensitive (217.78±22.5µg/mL) to treatment. 137 Table 4.25: Whole blood sulphadoxine concentration in children with uncomplicated falciparum malaria with sensitive or resistant response after oral administration of a standard dose of sulphadoxine-pyrimethamine (n=61). * Time Whole blood sulphadoxine concentration (µg/ml) Mean± (days) s em Sensitive (n=50) Resistant (n=11) P value RYwhole 0 0.0 0.0 - A blood 1 191.03 ± 23.43* 171.65 ± 81.15 RIB0.791 SDX 22.3 – 565.1** 62.1 – 413.1 L concen 2 215.60 ± 20.37 192.72 ± 42 82.3 – 421.3 49.5 – 2D70.A .73N 0.644 tration, 9 ** 3 179.06 ± 25.83 B157A.53 ± 38.23 0.737 Range 19.7 – 856.9 F I39.2 – 347.9 7 84.24 ± 14.92 O 51.95 ± 23.32 0.365 3.0 – 368.9Y 3.7 – 155.5 14 50.21 I±TS 8.85 14.59 ± 5.95 0.151 R4.7 – 151.6 3.7 – 24.1 21 VE 35.09 ± 11.17 8.40 ± 6.25 0.310 NI 2.6 – 81.8 2.16 – 14.66 U 138 Table 4.26: Pharmacokinetic parameters of sulphadoxine in children without pre-treatment sulphadoxine with uncomplicated falciparum malaria who had sensitive or resistant responses to oral administration of sulphadoxine- pyrimethamine (n=61). Parameters Sensitive (n=50) Resistant (n=11) P value Cmax (µg/ml) 217.78 ± 22.5 163.09 ± 30.35 0.277 RY Range 42.1 – 745.6 25.5 – 347.9 A T Rmax (d) 2.10 ± 0.14 2.63 ± 0.41 IB 0.148 Range 1.0-5.0 1.0 – 5.0 L t1/2 (d) 5.98 ± 0.50 5.2A6 ± N1.01 0.540 Range 1.1 – 17.0 D1.2 – 13.3 AUC0-28d (µg/ml.d) 1196.18 ± 131.B83 A 1013.93 ± 314.00 0.561 Range 91.1 – 36F79 .1I 125.4 – 3768.6 CL (mL/d) 437 .O45  49.16 531.61 ± 140.81 0.450 Range Y86.2 – 1356.1 98.3 – 1555.3 Parasite dSensIit Ty 47260 46311 0.830 (GM) R RangeV E 2011 – 461,333 2574 – 231,333 NIU 139 300 Responded to treatmenRt YFailed treatment RA 200 B N LI A 100 AD F I B 0 O 0 5 10 15 20 ITY Time (days)S Figure 4.14.: MEeanR whole blood concentration-time profile of sulphadoxine in children whose infecItVion responded and whose infection failed to respond to treatment after oral adminisNtration of standard dose of sulphadoxine-pyrimethamine (25mg/kg S-1.25mg/kg P). U 140 Concentration of Sulphadoxine (µg/ml) 4.24: Relationship between age and sulphadoxine pharmacokinetic parameters in children treated with standard oral dose of sulphadoxine-pyrimethamine. Figure 4.15 shows the mean concentration-time profile of sulphadoxine in children < 5 years and ≥ 5years who took oral dose of sulphadoxine-pyrimethamine for the treatment of uncomplicated malaria during infection. The mean SDX concenYtration on day 1 (126.9±30.5 versus 235.9±27.9µg/ml, P=0.015), day 3 (98.A1±1R5.9 versus 245.1±35.7µg/ml, P=0.001) and day 7 (54.1±10.5 versus 122.5±27R.7µg/ml, P=0.01) were significantly higher in children older than 5 years coLmpIarBed to those less than 5years of age (Table 4.27). N DA Table 4.28 describes the effect of ageB onA disposition of sulphadoxine in children infected with Plasmodium falciparFum . I A significant difference in disposition of SDX was observed between child reOn younger and older than 5 years of age. The Cmax of SDX in ≥5 years age groYup (295.7 ± 28.7 µg/ml) was significantly (p<0.001) higher than age group Sof cIh Tildren <5years (125.4 ± 15.2µg/ml). The time (tmax) taken to reach this peRak concentration were similar (2.0 ± 0.18d versus 2.4± 0.2d, p=0.160). The eVxteEnt of exposure (AUC0-28d) of SDX obtained in children ≥5 years of age (N156I2.9 ± 202.3µg/ml.d) was significantly higher (p<0.001) compared to the age Ugroup less than 5 years (812.0 ± 112.7µg/ml.d) respectively. The values of Cmax and AUC in older children were twice as high as those in younger children. In contrast, the mean clearance of SDX was significantly lower in older children (334.3 ± 45.8 versus 574.1±76.8 ml/d, P=0.010). However, response to treatment in the two age 141 groups was not significantly different (P=0.352) though a higher proportion of children below 5 years failed treatment with SP. RY RA LI B AN BA D F I O SI TY VE R UN I 142 Table 4.27: Whole blood sulphadoxine concentration in children with uncomplicated falciparum malaria with sensitive or resistant responses after administration of standard oral dose sulphadoxine-pyrimethamine (n=61). Whole blood sulphadoxine concentration (µg/ml) Time (days) <5 years (n= 31) ≥ 5 years (n=30) P value Y 0 0.0 0.0 - R 1 126.99 ± 13.09* 235.93 ± 27.92 0.R015A 36.1 – 413.1** 98.7 – 565.1 IB 2 182.57 ± 40.75 225.36 ± 20.39 L 0.344 49.1 – 312.1 103.2 – 421A.3 N 3 98.10 ± 15.94 245.19 D± 35.79 0.001 22.1 – 347.9 IB 191A.7 – 856.9 7 54.17 ± 10.59 F 122.54 ± 27.70 0.010 3.0 – 217.7 O 17.3 – 368.9 14 38.64T ± Y9.01 57.61 ± 15.52 0.268 S3.7I – 115.3 12.3 – 151.6 21 R 28.28 ± 13.62 30.67 ± 11.69 0.930 VE 14.6 – 41.9 10.2– 81.6 NI *Mean±sem whole blood SDX concentration, ** Range U 143 Table 4.28: Relationship between age and pharmacokinetic parameters of sulphadoxine in children with uncomplicated falciparum malaria treated with standard sulphadoxine-pyrimethamine (n=61). Parameters <5 year ≥5 Year P value (n=31) (n=30) Cmax (µg/ml) 125.45 ± 15.22 295.72 ± 28.72 0.0Y00 Range 25.5 – 347.9 98.0 – 745.6 R Tmax (d) 2.00 ± 0.18 2.40 ± 0.21 A 0.160 Range 1.0-4.0 1.0 – 5.0B Rt1/2 (d) 5.61 ± 0.58 6.10 ± 0I.70 0.593 Range 1.2 – 13.8 1.1 –L 17.0 AUC0-28d (µg/ml.d) 812.09 ± 112.77 AN1562.93 ± 202.3 0.001* Range 91.1 – 2194.5 222.9 – 3768.6 Cl (mL/d) 574.14  7A6.84D 334.39 ± 45.88 0.010* Range 86.2 – 1B555.3 86.6 – 867.9 Parasite density 3854 0I 57133 0.891 (GM) F Range OY 2011 – 461333 2574 – 231,333 IT Parasite S reduction 2.29 ± 0.98 3.32 ± 0.62 0.001* ratio (PRR) REespRonse IVACPR 24 (77.4%) 26 (86.7%) N ETF 1 (3.2%) 1 (3.3%) U LPF 6 (19.4%) 3 (10.0%) 0.352 144 300 Y children >A 5 RyearschildreRn < 5 years200 B LI 100 N DA 0 A 0.0 2.5 5.0 B 7.5 10.0 12.5 15.0 OF ITime (days) Figure 4.15: Mean conYcen tration-time curve of sulphadoxine in capillary whole blood of children yoIuTnger than 5 years or older than 5 years after oral administration of standard dose Sof sulphadoxine-pyrimethamine. VE R UN I 145 Concentration of sulphadoxine (g/ml) Study 5: Evaluation of the use of saliva for therapeutic drug monitoring in children with uncomplicated Plasmodium falciparum malaria treated with amodiaquine-artesuante. Amodiaquine (AQ) and desethyl amodiaquine (DEAQ) were well resolved in the analysis using the modified method of Gitau and others (Gitau et al., 2004).Y Th ere 2was a good linearity (r = 0.9907 plasma; Figure 4.16 and appendix TabRle 4.c; and 2 saliva, r =0.9838, Figure 4.18 and appendix Table 4.d) in the standaRrd cAurve obtained for desethyl amodiaquine in plasma and saliva. In additioInB, the recoveries for amodiaquine and desethylamodiaquine over the concentra tiLon range of 100 ng/ml to 600 ng/ml were between 69.7 and 79.4% for AplaNsma and saliva, respectively. Coefficient of variation within samples wereD 5.7% - 2.0%, and between samples 7.2% - 3.5%, for concentrations of I1B00 Ang/ml to 800 ng/ml, respectively. The compound eluted from the systemF in the order of quinidine, desethylamodiaquine and amodiaquine. Figure 4.18 sOhows the chromatograms for IS, DEAQ and AQ in plasma and saliva. ThereY was no interference with other drugs (e.g. paracetamol and other commonlyS useId T drug such as antimalarial, analgesic and anti-infective drugs) with the peaRks obtained (Figure 4.18). The lower limit of detection for desethyl amoIdViaqu Eine was 15ng/ml at 0.05 absorbance units’ full scale (aufs). The retention tNime for IS, DEAQ and AQ were 5.5, 9.5 and 11.5 minutes respectively. U 146 0.75 Y 0.50 RA R 0.25 LI B N 0.00 A 0 250 500 D 750 1000 ConcentrationA (ng/ml) F I B Figure 4.16: Standard curve oOf desethyl amodiaquine for extrapolation of unknown 2 concentrations of DEATQY in plasma (r = 0.9907). RS I IV E UN 147 Peak height ratio 2.5 2.0 RY 1.5 BR A 1.0 I 0.5 L 0.0 N 0 250 500 750 A 1000 Concentration (ng/Aml)D B Figure 4.17: Standard curve of dFese thIyl amodiaquine for extrapolation of unknown 2 concentrations of desethyl am Oodiaquine in saliva (r = 0.9838). SI TY ER IV UN 148 Peak height ratio IS A IS B AR Y BR AQ DEAQ LI DEAQ AQ N AD A IB Time (min.) F Time (min.) Figure 4.18: Chromatograms shOowing amodiaquine (AQ) and desethylamodiaquine (DEAQ) in plasma (A) IaTnd Ysaliva (B) of a patient at 4 h after the first dose of oral artesunate-amodiaquiSne. Quinidine is the internal standard (IS) VE R UN I 149 4.25: Patients enrolled in the study The characteristics of seven children who were randomly selected from the group of children treated with artesunate-amodiaquine combination (30mg/kg of amodiaquine base over 3 days that is 10mg/kg daily) are shown in Table 4.29. The seven children enrolled in the study aged between 6-13 years. The mean age was 10.8 ± 2.8 years and mean weight was 26.2 ± 5.9kg. The geometric mean parasite densitYy w as 43797/µl of blood. The pH of saliva generally ranged between 6.8 andR 7.8 in all children. RA LIB AN D IB A O F SI TY VE R UN I 150 Table 4.29: Baseline clinical and demographic characteristics of seven children with acute uncomplicated falciparum malaria enrolled in the study. Y Parameters Mean±SD R Age (y) 10.8±2.8 A Range 7 – 13 R Weight (kg) 26.2±5.9 LIBRange 16 – 33 Gender (F:M) 4:3 N O Axillary Temperature ( C) 38.2±0.65 A Range 36.9 – 38.6D % PCV 32I.6B±4 A Range 2 9 – 39 Geometric Mean of parasiteF 43797 density /µl of blood O Range Y 19,009 – 150,000 SD= standardI Tdeviation, PVC= pack cell volume, ER S V UN I 151 4.26: Pharmacokinetic parameters of Desethyl amodiaquine in children with uncomplicated falciparum malaria after administration of standard doses of artesunate-amodiaquine Two of the seven children who were enrolled had AQ and DEAQ in their blood pre- dosing and were excluded from the analysis. Therefore data from 5 children out of the 7 who were enrolled were used for the pharmacokinetic analysis. Amodiaqu ine was rapidly converted to desethylamodiaquine, which appeared in the pRlasmYa and saliva within 40 minutes in all subjects. All children had detRectaAble levels of amodiaquine in plasma and saliva up to 40 h. The plasma anId Bsaliva concentration- time curve for desethylamodiaquine for the five children aLre shown in Figure 4.19. The concentration on day 7 was 210.3ng/ml in AplaNsma and 131.8ng/ml in saliva (Table 4.30). The decline phases of the desethyDlamodiaquine in saliva concentration- time curves were approximately paralleBl toA that in plasma (Figure 4.19). The saliva concentration was approximately aF qu aIrter and two-fifth that in plasma. The pharmacokinetic param eOters derived from the oral plasma concentration-time curve are summarizeTd iYn Table 4.31. The saliva pharmacokinetic parameters are summarized in TSablIe 4.32. The plasma half-life (t1/2) was 156.7±20.6h (s.e.m). The elimination Rhalf-life from saliva was also similar at 139.1±8.3h. There was no signIifVican Et difference between plasma and saliva half-life (P = 0.16) (Table 4.33). TNhe mean AUC were 96003.6 ± 12344.6 ng/ml.h (s.e.m) for plasma and 74004.2 ± U9514.3 ng/ml.h for the saliva and were similar (P = 0.196) (Table 4.33). Comparison of other pharmacokinetic parameters in the 5 patients gave a mean volume of distribution (Vd) of 74.1 ± 16.2 L/kg (s.e.m) for plasma and 91.5±21.8 L/kg for DAQ in saliva. The plasma oral clearance calculated from the expression CLp=fD/AUC, 152 was found to be 325.4±48.8ml/h/kg (s.e.m). This, too, was not significantly different from the clearance of 443.7±85.1ml/h/kg obtained from saliva data. Y AR BR N LI DAA F I B O ITY RS IV E UN 153 Table 4.30: Saliva-plasma ratio of desethyl amodiaquine concentrations in five children with falciparum malaria treated with standard oral doses of artesunate-amodiaquine. Desethyl amodiaquine concentration (ng/ml) Time Plasma (n=5) Saliva (n=5) Saliva/plasma P value (days) ratio Y 0 0 0 ND NAD R 7 210.3±31.59* 131.8±19.5 0.626 R0.068 121.9 – 314.9** 75.5 – 176.1 IB 14 193.9±47.1 141.0±24.0 0.727 L 0.358 98.7 – 293.3 92.5 – 183.4 AN 28 111.3±48.1 41.4±24.0 D 0.119 0.237 63.1 – 159.4 9.0 – 8B8.5 A 35 27.6±5.4 6F.4± 3I.1 0.233 0.064 19.2 – 37.7 O3.3 – 9.6 *Mean±sem, ** Range ITY RS IV E UN 154 Y Plasma Saliva R 2.25 A LIB R 1. 25 N 0. 25 DA 0 100 200 300 400 A500 600 700 800 900 T IimBe (h) OF Figure 4.19: PlasmYa and Saliva log concentration-time profile of desethylamodiaquinIe T(DEAQ) in children infected with P. falciparum malaria who were treRatedS with oral doses of artesunate-amodiaquine.E IV UN 155 DEAQ concentration (ng/ml) Table 4.31: Pharmacokinetic disposition of desethyl-amodiaquine in plasma after standard oral doses of artesunate-amodiaquine (30mg/kg amodiaquine) in five children with uncomplicated malaria. Subject Weight AUC0-35d YExtrapolated (Kg) -1 -1 -1 -1(n=5) t1/2 (h) Vd (Lkg ) Cl (mlh kg ) (ng.ml h) R AUC (%) 1 27 132.9 87.2 454.8 71370.0 A 2.6 2 29 233.3 131.2 389.7 708I5B9.4 R 11.0 3 20 128.8 43.5 234.0 L124593.5 2.7 4 29 121.5 62.4 355.9 AN 87015.4 0.2 5 33 167.2 46.5 192.D8 126179.8 10.7 Mean ±sem 27.6±2.1 156.7±20.6 74.1±16.2 B3A25.4±48.8 96003.6±12344.6 5.4±2.2 s.e.m= Standard error of mean OF I TY RS I IV E UN 156 Table 4.32: Pharmacokinetic disposition of desethylamodiaquine in saliva after standard oral doses of artesunate-amodiaquine (30mg/kg amodiaquine) in five children with uncomplicated malaria. Subject Weight AUC0-35d Extrapolated (Kg) -1 -1 -1 -1(n=5) t1/2 (h) Vd (Lkg ) Cl (mlh Kg ) (ngml h) A UC (%) 1 27 133.6 66.7 346.0 94390.0 RY2.0 2 29 135.8 119.2 608.1 50376.9 A 1.2 3 20 114.5 52.3 316.7 907B43.9R 4.1 4 29 165.7 164.2 686.6 L42I804.1 5.2 5 33 146.1 55.0 260.9 N 85843.5 17.8 Mean±sem 27.6±2.1 139.1±8.3 91.4±21.8 443.D7±8A5.1 72831.7±10864.5 6.1±3.0 s.e.m= Standard error of mean BA F I O ITY ER S IV UN 157 Table 4.33: Comparison of pharmacokinetic disposition of desethyl amodiaquine in plasma and saliva after standard oral doses of artesunate-amodiaquine (30mg/kg amodiaquine) in children with uncomplicated malaria (n=5). Parameters Plasma n=5 Saliva n=5 P value t1/2 (h)* 156.7 ± 20.6 139.1 ±8.3 0.453 95% CI 99.3 – 214.2 115.8 – 162.4 Range** 121.5 – 233.3 114.5 – 165.7 Y AR -1 AUC0-35d (ngml .h) 96003.6±12344.661 72831.7±10864B.54R0.196 95% CI 729.3 – 130277.9 2666.8–1 02L99I6.6 Range 87015.4 – 126179.9 42804N.1 – 94390.0 D A -1 -1 CL (mLh Kg ) 325.4±48.8 A 443.7±85.1 0.302 95% CI 189.4-461.1 B 207.8-680.2 Range 192O.8 –F I 454.8 260.9 – 680.1 Y -1 Vd (LKg ) IT 74.1  16.2 91.5 ± 21.8 0.542 95% CI RS 29.1 – 119.2 30.8 – 152.1 RangVe E 43.5 – 131.2 52.3 – 164.2 *NMeIan±sem, ** Range U 158 REFERENCES Abdi, Y. A., Gustafsson, L.L, Ericsson, O. and Hellgreen, U. (1995). Hand book of nd Drugs for Tropical Parasitic Infections 2 Edition Taylor and Francis, p.155-159. Adam, I., Khamis, A.H. and Elbashir, M.I. (2005). Prevalence and risk factors for Plasmodium falciparum malaria in pregnant women of eastern Sudan. Malaria Journal, 4:18, 4-18. Adelusi, S.A., Dawodu, A.H., and Salako, L.A. (1982). Kinetics of the uRptakYes and elimination of chloroquine in children with malaria. British Journal of Pharmacology, 14: 483-487. A Adjei, G.O., Kristensen, K., Goka BQ., Hoegberg, L.C.G., AlifrangRis M., Rodrigues, O. P. and Kurtzhals, J.A.L. (2008). Effect of CoInBcomitant Artesunate Administration and Cytochrome P450 2C8 LPolymorphisms on the Pharmacokinetics of Amodiaquine in NGhanaian Children with Uncomplicated Malaria. Antimicrobial Agent and Chemotherapy, 52(12), 4400–4406. A Adjuik, M., Agnamey, P., Babiker, A., (20D02). Amodiaquine–artesunate versus amodiaquine for uncomplicated PAlasmodium falciparum malaria in African children: a randomized, multiIcBentre trial. Lancet, 359:1365–1372. Ahmad, R. A., and H. J. RogerFs. (1980). Pharmacokinetics and protein binding interactions of dapsOone and pyrimethamine. British Journal of Clinical Pharmacology, 10: 519– 524. Ajayi, F.A., Salako, L.AY., and Kuye. J. O., (1988). Comparison of the partitioning in-vitro of cIhTloroquine and its desethyl metabolites between the erythrocytes and plasSma of healthy subjects and those with falciparum malaria. African JourRnal of Medical Science, 430:926–927. AkinyinkEa, O.O., Sowunmi, A., Honeywell, R. and Renwick, A.G. (2000). The IVeffect of acute falciparum malaria on the disposition of caffeine and the comparison of saliva and plasma-derived pharmacokinetic parameters in N adult Nigerians. European Journal of Clinical Pharmacology, 56:159-165. UAnonymous (1994). Epi info Version 6. A word processing data base and statistical program for public health on IBM- compatible microcomputer. Atlanta, (GA). Centers for Disease and Control and prevention and Geneva: World Health Organisation. Anonymous (2005). Global distribution of malaria; Adopted from World Malaria Report (http://rbm.who.int/wmr2005/html/map1.htm, Retrieved on March 2006. 159 Ashton, M., Hai, T.N., Sy, N.D., Houng, D.X., Vn Huong, N., Nieu, N.T. and Cong, L.D. (1998). Artemisinin pharmacokinetics is time-dependent during repeated oral administration in healthy male adults. Drug Metabolism and Disposition, 26, 25-27 Aubouy, A., Bakary, M., Keudjian, A. (2003). Combination of drug level measurement and parasite genotyping data for improved assessment of amodiaquine and sulphadoxine-pyrimethamine efficacies in treatment Plasmodium falciparum malaria in Gabonese children. Antimicrobial Agents and Chemotherapy, 47:231-237. Babalola, C.P., Bolaji, O.O., Ogunbona, F.A., Dixon, P.F. (1996) Relations hip between plasma and saliva quinine levels in humans. TherapeuticsY Drug Monioringt, 18:30-33. AR Babalola, C.P., Bolaji, O.O., Ogunbona, F.A., Sowunmi, A and WRalker, O. (1998). Pharmacokinetics of quinine in African patients wIiBth acute falciparum malaria. Pharmaceutical World Science , 20 (3):118-122. Babalola, C.P., Scriba, G.K.E., Sowunmi, A., Alawo dLe, O.A. (2003). Liquid chromatographic determination of pyronaridine in human plasma and oral dosage form. Journal of Chromatography AB, 7N95 265–272. Barat, L.M. (2006). Four malaria succAessD stories: how malaria burden was successfully reduced in brazil, eritrea, india, and Vietnam. The American Society of Tropical Medicine aInBd Hygiene., 74(1), 2006, pp. 12–16. Baird, L.K. (2005). EffectivenesFs o f antimalarial drugs. New England Journal of Medicine, 352;1565-O1577 Barnes, K.I., Little, F.,Y Sm ith, P.J., Evans, A., Watkins, W.M. and White, N.J. (2006). SuTlphadoxine-pyrimethamine pharmacokinetics in malaria: Paediatric Idosing implications. Pharmacokinetic and drug disposition. JournaSl of American Society for Clinical Pharmacology and Therapeutics. 80(6):582-96. Barnes, KE.I.R, Watkins, W.M. and White, N.J. (2008). Antimalarial dosing regimens V and drug resistance. Cell, Vol. 24(3)127-134. BNayIoumi, R.A., Dar, F.K., Tamira, M.O.M., Stephen, R.S., Hussein, M.M., Hammo, U N.E., Omer, R.I., Beidas, M.F., Shalabi, A. and El-Wasila M. (1997). Effect of previous chloroquine intake on in-vivo P. falciparum. East African Medical Journal, 74(5):278-282 Bergqvist, Y. and Eriksson M. (1985). Simultaneous determination of pyrimethamine and sulfadoxine in human plasma by high performance liquid chromatography. Transaction of Royal Society of Tropical Medicine and Hygiene. 79; 297-301. Bousquet, E. (1970) Asthma and glomectomy. Laval Med. 41(10):1016-1017. 160 Bradley, T., (1996). Malaria and drug resistance. Special Study Module Project Report. Department of Microbiology and Immunology, University of Leicester, USA. Bradbury, J. (2007). Malarial sensitivity to chloroquine returns to Malawi. The Infection Lancet, l7 January. Bryskier, A. and Labro, M.T. (1988). Paludisme et medicaments. Edition. Brohult, J., Rombo L., Sirleaf, V., Bengtsson, E. (1979). The concentration of chloroquine in serum during short and long term malaria prophylaxis w ith standard and 'double' dosage in non-immunes: clinical implications. Annals Tropical Medicine Parasitolology,73(5):401-5. RY Brown-Awala, E.A., Thomas, W.O., Essien, E.E., and Ette, E. I., (1989). Determination of saliva: total plasma chloroquine levels relatioAnship, by high performance liquid chromatography. Journal of ClinicaBl PRharmacology, 29: 1135-1139 I Butos, D.G., Lazaro JE., Gay F., Pottier A., Laracas C.J. , LTraore B., and Diquet B. (2002). Pharmacokinetics of sequential and siNmultaneous treatment with the combination chloroquine and sulfadAoxine-pyrimethamine in acute uncomplicated Plasmodium falciparumD malaria in the Philippines. Tropical Medicine and International Health, A7: 584-591. Bzik, D.J., Li, W.B., Horil, T. and IIBnselberg, J. (1987). Molecular cloning and sequence analysis of the Pl asmodium falciparum dihydrofolate reductase-thymidylate synthase genFe. Proceedings of the National Academy of Sciences of the United States oOf America, 84: 8360-8364. Checchi, F., Piola P., FoYgg C. (2006). Supervised versus unsupervised antimalarial treatment wTith six-dose artemether-lumefantrine: Pharmacokinetic and dosage regimen findings from a clinical trial in Uganda. Malaria Journal, 5:59. SI Churchill, FR.C., Mount, D.L, and Patchen, L.C. (1986) Isolation, characterization aEnd standardization of a major metabolite of amodiaquine by Vchromatographic and spectroscopic methods. Journal of Chromatography. I 377:307–318. UCNhurchill, F.C., Patchen, L.C., Campbell, C.C., Schwartz, I.K., Nguyen-Dinh, P. and Dickinson, C.M. (1985) Amodiaquine as a pro-drug: the importance of metabolite(s) in the antimalarial effect of amodiaquine in humans. Life Science. 36:53–62. Cowman, A.F (1997). The mechanism of drug action and resistance in malaria . Molecular Genetics in Drug Resistance. 3, 221-246. Crutcher, J.M. and Hoffman, S.L. (2001). Malaria. Medimicro Chapter 83. Dondorp, A.M., Nosten, F., Yi, P., Das, D., Phyo, D., Tarning, J., Lwin, K.M., Ariey F., Hanpithakpong W., LeeSJ., Ringwald P., Silamut K., Imwong M., 161 Chotivanich K., Lim P., Herdman T., Sam An S., Yeung S., Singhasivanon P., Day NPJ., Lindegardh N., Socheat D.and White NJ (2009). Artemisinin Resistance in Plasmodium falciparum Malaria. The New England Journal of Medicine, 361; 455-469. Dondorp AM., Fanello CI., Hendriksen ICE., Gomes E., Seni A., Chhaganlal KD, Bojang K., Olaosebikan R, Anunobi N., Maitland K., Kivaya E., Agbenyega A., Nguah SB., Evans J., Gesase S., Kahabuka C., Mtove G., Nadjm B., Deen J., Mwanga-Amumpaire J., Nansumba M., Karema C., Umulisa N., Uwimana A., Mokuolu OA., Adedoyin OT., Johnson WBR., Tshefu AK., Onyamboko MA., Sakulthaew T., Ngum WP., Silamut K., Stepniewska K., Woodrow CJ., Bethell D., Wills B., Oneko M., Peto TE., von Seidlein L., Day NPJ., White NJ (2010). Artesunate versus quinine in the treatmYent of severe falciparum malaria in African children (AQUAMAT): an oRpen-label, randomised trial. The Lancet, 376; 9753, 1647 – 1657 A Dua, V.K., Gupta, N.C., Kar, P.K., Nand, J., Edwards, G., SRharma, V.P. and Subbarao, S.K. (2000). Chloroquine and desethylchLloIroBquine concentrations in blood cells and plasma from Indian patients infected with sensitive or resistant Plasmodium falciparum. Annals of Tropical Medicine and Parasitology, 94(6) 565 – 57. N Doberstyn, E.B., Phintuyothin, P., NoeypatimaAnordh, S., Teerakiartkamjorn, C. (1997). Single dose therapy of AfalDciaprum malaria with mefloquine or pyrimethamine-sulphadoxine. BBulletin of World health Organisation. 57:275. Dorsey, G., Kamya, M.R., NdeF Iezi, G., Babirye, J.N., Phares, C.R., Olson, J.E., Katabira, E.T., RoseOnthal, P.J. (2000). Predictor of chloroquine treatment failure in children and adults with falciparum malaria in Kampala, Uganda. American JournYal of Tropical Medicine and Hygiene, 62(6):686-92. Dzinjalamala, F.KI.T, Macheso, A., Kublin, J.G., Taylor, T.E., Barnes, K.I., MolyneuSx, M.E., Plowe, C.V. and Smith, P.J. (2005). Blood folate concRentrations and invivo Sulphadoxine-pyrimethamine failure in Malawian cEhildren with uncomplicated Plasmodium falciparum Malaria. American Journal of Tropical Medicine and hygiene, 72(3): 267-272. DzinIjValamala, F.K., Macheso, A., Kublin, J.G., Taylor, T.E., Barnes, K.I., N Molyneux, M.E., Plowe, C.V. and Smith, P.J. (2005a). Association between U the pharmacokinetics and in vivo therapeutic efficacy of Sulphadoxine-pyrimethamine in Malawian children. Antimicrobial Agent and Chemotherapy, 49 (9):3601-3606. Edstein, M.D., Veenendaal, J.R., Newman, K., and Hyslop, R. (1986). Excretion of chloroquine, dapsone and pyrimethamine in human milk. British Journal of Clinical Pharmacology, 22: 733-735. Ehrhart, S., Mockenhaupt, F.P., Eggelte, T.A., Agana-Nsiire, P., Stollberg, K., Anemana, S. D., Otchwemah, R.N. and Bienle, U. (2003). Chloroquine 162 blood concentration-resistant Plasmodium falciparum in febrile children in northern Ghana. Ekanem, O.J., Weisfeld, J.S., Salako, L.A., Nahlen, B.L., Ezedinachi, E.N., Walker, O., Breman, J.G., Laoye, O.J., Hedberg, K. (1990). Sensitivity of Plasmodium falciparum to chloroquine and sulfadoxine/pyrimethamine in Nigerian children. Bulletin World Health Organanization, 68(1):45-52.. Ette, E.I., Essien, E.E., Brown-Awala, E.E., (1986). Pharmacokinetics of chloroquine: saliva and plasma levels relationship. European Journal of Drug Metabolism and Pharmacokinetics, 11(4):275-81. Ewards, G. and Biagini, G.A. (2006). Resisting resistance: dealing wiYth the irrepressible problem of malaria. British Journal of Clinical PharRmacology, 61: 6, 690-693. A Ezeilo, G.C. (1971). Neutropenia in Africans. Tropical aRnd Geographical Medicine, 23, 264-267. IB Falade, C.O., Salako, L.A., Sowunmi, A., Oduola, A.M .LJ. and Larcier, P. (1997). Comparative efficacy of chloroqNuine, halofantrine and Sulphadoxine/pyrimethamine in treatment Aof acute uncomplicated falciparum malaria in Nigeria. Transaction of RoDyal Society of Tropical Medicine and Hygiene, 91, 58-62. Fehintola, F.A. and Balogun, S.TI. B(20 A10). Comparative study of efficacy of artesunate plus cotrimoxaz ole and artesunate plus chloroquine in the treatment of malaria in NFigerian children: a preliminary report. Journal of Vector Borne DiseaseO, 47: 145–150 Ferreira, I.D., NogueiraY, F. , Borges, S.T., do Rosario, V.E., Cravo, P. (2004). Is the expression oTf genes encoding enzymes of glutathione (GSH) metabolism involved inI chloroquine resistance in P. Falciparum chabaudi parasites? MoleculSar Biochemistry and Parasitology, 136, 43-50 Ferone (197R7). Folate metabolism in malaria. Bulletin W.H.O. 55: 291-298 FMHV (20E04). National Antimalarial Treatment Policy. Federal Ministry of Health I Abuja, Nigeria. UFNMH (2005). National Antimalarial Treatment Policy. Federal Ministry of Health Abuja, Nigeria. Francis, S.E., Sullivan, D.J., Goldberg, D.E., (1997). Haemoglobin metabolism in the malaria parasite P. falciparum. Annals Rev. of Microbiology. 51: 97- 123. Fontanet, A.L. and Walker, A.M. (1993). Predictors of treatment failure in multiple drug resistant falciparum malaria: results from a 42-day follow-up of 224 patients in eastern Thailand. American Journal of Tropical Medicine and Hygiene, 49, 465-472. 163 Frisk-Holmberg, M., Bergqvist., Termond, E. and Domeij-Nyberg, B., (1984). The single dose kinetics of chloroquine and its major metabolites desethylchloroquine in healthy subjects. European Journal of Clinical Pharmacology, 26: 521-530. Gardella, F., Assi, S., Simon, F., Bogreau, H., Eggelte, T., Ba V.., Foumane V., Henry, M., Traore, P., Basco, L., Trape, J., Lalou R., Martelloni, M., Desbordes M., Baragatti M., Briolant S., Almeras L., Pradines B., Fusai T and Rogier C (2008). Antimalarial drug use in general populations of tropical Africa. Malaria Journal Vol. 7, 124 Gbotosho, G.O., Happi, T.C., Sijuade, A.O., Ogundahunsi, O.A.T., Sowunmi, A. and Oduola, A.M.J. (2008). Comparative study of interaction beYtween chloroquine and chlorpheniramine or promethazine in healthyA voRlunteers: a potential combination-therapy phenomenon for resuscitating chloroquine for malaria treatment in Africa. Annals of Tropical Medicine Rand Parasitology, 102(1): 3-9. B Gbotosho, G.O., Happi, C.T., Sijuade, A., Sowunmi, A. a nLd OIduola, A.M.J. (2009). Pharmacokinetic disposition of sulfadoxiNne in children with acute uncomplicated falciparum malaria treated with sulfadoxine–pyrimethamine in South West Nigeria. American Journal Aof Therapeutics, 2009; Nov 14. E pub ahead of print. D Geoffrey, S. D., Hudson, T.H., VaheyB, MA. and Koenig, M. L (2003). The acute neurotoxicity of mefloquin e Imay be mediated through a disruption of calcium homeostasis and ER function in vitro. Malaria Journal. 2: 14. Ginsburg, H. (2005). Should OchloFroquine be laid to rest? Acta Tropica 96:16–23. Gitau, E.N., Muchohi, S.N. , Ogutu, B.R., Githiga, I.M. and Kokwaro, G.O. (2004). Selective andT seYnsitive liquid chromatographic assay of amodiaquine and desethylamoIdiaquine in whole blood spotted on filter paper. Journal of ChromaStography B, 799:173-177. Global MalRaria (2008). Malaria Key facts. Adopted from Global Malaria Action PElan, (http://www.rollbackmalaria.org/keyfacts.html, Retrieved on August IV2010. GN thoodman, A.G. (2006). The pharmacological basis of therapeutics. 9 Edition. U McGraw-Hill, New York. GraphPad Prizm. (2003). H.J. Motulsky, Prism 4 Statistics Guide −Statistical analyses for laboratory and clinical researchers. Graph Pad Software Inc., San Diego CA. Greenwood, B. (2004). Malaria: between hope and hard place. Nature. 430(7002):926-7 Green, M.D., Dwight, L.M. and Netty, H. (2002). High performance liquid chromatographic assay for the simultaneous determination of sulphadoxine 164 and Pyrimethamine from whole blood dried onto filter paper. Journal of Chromatography B. 767:159-162. Green, M.D., van Eijik, A.M., van te Kuile, F.O., Ayisi, J.G., Parise, M.E., Kager, P.A., Nahlen, B.L., Steketee, R. and Netty H. (2007). Pharmacokinetics of sulphadoxine-pyrimethamine in HIV-Infected and uninfected pregnant women in Western Kenya. The Journal of Infectious Diseases. 196:1403-8 Gregson A and Plowe CV. (2005). Mechanisms of resistance of malaria parasites to antifolates. Journal of Pharmacological Review, 57:17-45 Gustaffson, L.L., Lindstrom, B., Grahnen, A., and Alavan, G.,(1987). Chloroqu ine excretion following malaria prophylaxis. British Journal of CYlinical Pharmacology 24, 221-224. AR Gustaffson, L.L., Walker, O., Alavan, G., Beerman, B., EstevezR, F., Gleisner, L., Lindstrom, B., and Sjoqvist, F (1983). The dispositionI oBf chloroquine in man after single intravenous doses. British Journal of CLlinical Pharmacology 15, 481-489. Happi, C.T., Gbotosho, G.O., Sowunmi, A., Falade, C.O., Akinboye, C.O., Gerena, L., Kyle, D.E., Milhouse, W., Wirth, DA.F.,N and Oduola, A.M.J. (2004). Molecular analysis of PlasmodiumD falciparum recrudescence malaria infections in children treated with cAhloroquine in Nigeria. American Journal of Tropical Medicine and HygieBne, 70,20-26 Happi, CT., Gbotsho, G.O., Folarin , IO.A., Bolaji, O.M., Sowunmi, A., Kyle D.E., Milhouse, W., Wirth, D.FF., and Oduola, A.M.J. (2006) Association between mutations in PlasmodOium falciparum chloroquine resistance Transporter and P. falciparum multidrug resistance 1 genes and in vivo amodiaquine resistance in P. Yfalc iparum malaria-infected children in Nigeria. American Journal of TrTopical Medicine and Hygiene, 336: 1201-1204. Hammer, D.H.,S MIacleod W.B., Addo-Yobo E., Duggan, C.P., Estrella, B., Fawzi W.WR., Kondelule J.K., Mwanakasale, V., Premji, Z.G., Sempertegui F., Ssengooba, F.P., Yeboa-Antwi, K. and Simon JL.. (2003). Age, temperature, aEnd prasitaemia predict chloroquine treatment failure and anaemia in IVchildren with uncomplicated Plasmodium falciparum malaria. Transaction of the Royal Society of Tropical Medicine and Hygiene, 97, 422-428. UHNappi, C.T., Thomas, S.M., Gbotosho, G.O., Falade, C.O., Akinboye, C.O., Gerena, L., Hudson, T., Sowunmi, A., Kyle, D.E., Milhouse, W., Wirth, D.F., and Oduola, A.M.J. (2003). Point mutation in the pfcrt and pfmdr1 genes of Plasmodium falciparum and clinical response to chloroquine, among malaria patients from Nigeria. American Journal of tropical Medicine and Parasitology. 97, 439-451. Hatton, C.S., Peto, T.E.A., Bunch, C., Pasvol, G., Russel, S.J., Singer, C., Edward, G., and Winstanley, P. (1986). Frequency of severe neutropenia associated with amodiaquine prophylaxis against malaria. Lancet i: 411-413. 165 Hayward, R., Saliba, K.J. and Kirk, K. (2006). The pH of the digestive vacuole of Plasmodium falciparum is not associated with chloroquine resistance. Journal of Cell Science. Feb 21. Hellgren, U., Kihamia, C.M., Mahikwano, L.F., Bjorkman, A., Eriksson, O., Rombo, L. (1989). Response of Plasmodium falciparum to chloroquine treatment: relation to whole blood concentrations of chloroquine and desethylchloroquine. WHO Bulletin, 67:197-202. Hellgren, U., Angel, V.H., Bergqvist, Y., Arvidsson, A., Forero-Gomez, J.S. and Rombo, L. (1990). Plasma concentration of sulphadoxine-pyrimethamine and mefloquine during regular long term malaria prophylaxis. Transaction of Royal Society of Tropical Medicine and Hygiene.84:46-49 RY Hodel, E.M., Genton, B., Zanolari, B., Mercier, T., Duong, S., Beck, HA., Olliaro, P., Decosterd, L.A. and Ariey, F. (2010). Residual antimalarial concentrations before treatment in patients with malaria from Cambodia: IRndication of drug pressure. The Journal of Infectious Diseases. 202(7):1I0B88–1094 Hoegberg, L.C.G., Khalil, I.F., Angelo, H.R., BygbjeNrg, I. LC., Ronn, A.M (2005). A reversed-phase high-performance liquid chromatography method for the determination of amodiaquine and the maiAn metabolites in small volumes of blood and plasma. Acta Tropica 955 SID-S506. Hombhanje, F.W., Hwaihwanje, I., TsukaAhara, T., Saruwatari, J., Nakagawa, M., Osawa, H. (2005). The dispIoBsition of oral amodiaquine in Papua New Guinean children with falciparum malaria. British Journal of Clinical pharmacology, 59(3):298F–30 1 Hviid, L. (2005). Naturally acOquired immunity to Plasmodium falciparum malaria in Africa. Acta TroYpica 95:270-275. Jambou, R., LegraIndT, E., Niang, M., Khim, N., Lim, P., Volney, B., Ekala, M.T., BouchierS, C., Esterre, P., Fandeur, T., Mercereau-Puijalon, O. (2005). ResisRtance of Plasmodium falciparum field isolates to in-vitro artemether and point mutations of the SERCA-type PfATPase6. Lancet, 366 (9501):1960-3. KageVr, PE.A. (2002). Malaria control: constrains and opportunities. Tropical I Medicine and International Health. 7:12, 1042-1046. UKNarbwang, J., Looareesuwa, S., Phillips, R.E. Wattanagoon, Y. Molyneux, M.E., Nagachinta, B., Back, D.J. and Warrell D.D. (1987). Plasma and whole blood mefloquine concentrations during treatment of chloroquine-esistant falciparum malaria with the combination mefloquine-sulphadoxine- pyrimethamine. British Journal of Clinical Pharmacology. 2, 47-481. Karunajeewa, H.A., Ilett, K.F., Dufall, K., Kemiki, A., Bockarie, M., Alpers, M.P., Barrett, P.H., Vicini, P. and Davis T.M.E. (2004). Disposition of Artesunate and Dihydroartemisinin after Administration of Artesunate Suppositories in Children from Papua New Guinea with Uncomplicated Malaria. Antimicrobial Agent and Chemotherapy, 8:2966–2972. 166 Kihara, M., Carter, J.A., Newton, C.R.J.C. (2006). The effect of Plasmodium falciparum on cognition: a systematic review. Tropical Medicine and International Health. 11(4):386-397 Klayman, D.L. (1985). Qinghaosu (artemisinin): An antimalarial drug from china. Science, 228:1049-1055. Kondrachine, A.V. and Trigg, P.I. (1997). Global overview of malaria. Indian Journal of Medical Research, 106:36-52. Krishna, S. and White, N.J. (1996). Pharmacokinetics of quinine, chloroquine and amodiaquine: Clinical implication. Journal Clinical Pharmacokinet ics. 30(4): 236-299. RY Krishna S., Webb, R., Woodrow, C. (2001).Transport proteins of Plasmodium falciparum: defining the limits of metabolism. InternationaAl Journal of Parasitology, 31(12):1331-42. BR Laufer, M.K.,, Thesing, P.C., Eddington, N.D., Masong aL, RI., Dzinjalamala, F.K., Takala, S,L., Taylor, T.E., Plowe, C.V. (2006). Return of chloroquine antimalarial efficacy in Malawi. New EnglaNnd Journal of Medicine, 355: 1959-1966. A Laurent, F., Saivin, S., Chretien, P., MagnAavaDl, J.F., Peyron, F., Sqalli, A., Tufenkji, A.E., Coulais, Y., Baba, H. and Campistron, G. (1993). Pharmacokinetic and pharmacodynamic study of aImBodiaquine and its two metabolites after a single oral dose in human vo lunteers. Arzneim-Forsch, 43:612–616 Li, X-Q., Bjorkman, A., AnOdersFson, T.B., Ridderstrom, M. and Masimerembwa, C.M. (2003). Ident ification of human cytochrome P450’s that metabolise anti-parasitic druYgs and predictions of in vitro drug hepatic clearance from in-vitro data. T European Journal of Clinical Pharmacology, 59: 429-442. Li, X-Q., BjorkSmanI, A., Andersson, T.B., Ridderstrom, M. and Masimerembwa, C.M. (2002). Amodiaquine clearance and its metabolism to N-desethyl amodiaquine is mediated by CYP2C8: a new highly affinity and turnover eEnzyRme-specific probe substrate. Journal of Pharmacology and Experimental VTherapeutics, 300: 399-407. LNi, IQ., Xie, L.H., Johnson, T.O., Si, Y., Haeberle, A.S., and Weina, P.J. (2007). U Toxicity evaluation of artesunate and artelinate in Plasmodium Berghei-infected and uninfected rats. Transaction of Royal Society of Tropical Medicine and Hygiene 101, 104-112. Looareesuwan, S., White, N.J., Chanthanvanich, P., Edwards, G., Nicholl, D.D., Bunch, C. and Warrel, D.A. (1986). Cardiovascular toxicity and distribution kinetics of intravenous chloroquine. British Journal of clinical Pharmacology, 22, 31-36. Maggs, J.L., Kitteringham, N.R., Breckenridge, A.M. and Park, B.K. (1987). Autoxidative formation of a chemically reactive intermidiate from 167 amodiaquine, a myelotoxin and hepatoxin in man. Journal of Biochemical Pharmacy. 36(13): 2061-2062. Mansor, S.M., Taylor, T.E., McGrath, C.S., Edward, G., Wirima, J.J., Molyneux M.E (1990). The safety and kinetics of intramuscular quinine in Malawian children with moderately severe falciparum malaria. Transaction of Royal Society of Tropical Medicine and Hygiene, 84:482-487. May, J., Adjei, S., Busch, W (2008). Therapeutic and prophylactic effect of intermittent preventive antimalarial treatment in infants (IPTi) from Gh ana and Gabon. Malaria Journal 7:198-205. Mayxay, M., Chotivanich, K., Pukrittayakame, S., (2001). Contribution oRf huYmoral immunity to the therapeutic response in falciparum malaria. American Journal of Tropical Medicine and Hygiene, 65:918-923 A McCombie, S. C. (2002). Self-treatment for malaria: BthRe evidence and methodological issues. Health policy and Planning, 17I(4):333-344. Miaffo, C., Some, F., Kouyate, B., John, A. and Muell erL, O. (2004). Malaria and anemia prevention in pregnant women of rural Burkina Faso BMC Pregnancy and Childbirth, 8, 4-18 AN Michihara, A., Toda, K., Kubo, T., FujiwarDa, Y., Akasaki, K., Tsuji, H. (2005). Disruptive effect of chloroquine on lysosomes in cultured rat hepatocytes. Journal of Antimicrobial ChemoBtheArapy, 55(5):788-91. Minzi, O.M.S., Massele, A.Y., GFus taIfsson, L.L. and Ericsson, O. (2005). Simple and cost effective lOiquid chromatographic method for determination of pyrimethamine in w hole blood samples dried on filter paper. Journal of chromatographyY B, 814: 179-183. Minzi, O.M.S., RaiIs,T M., Svensson, J.O., Gustafsson, L.L. and Ericsson, O. (2003). High peSrformance liquid chromatographic method for determination of amodiaquine, chloroquine, and their monodesethyl metabolites in biological sampRles. Journal of Chromatography B, 783:473-480. MockVenhEaupt, F. P., (1995). Mefloquine resistance in P. falciparum. Parasitology I Today, 11: 248-253. UMNount, D.L, Patchen, L.C., Nguyen-Dinh, P., Barber, A.M., Schqartz, I.K. and Churchill, F.C. (1986). Sensitive analysis of blood for amodiaquine and three metabolites by high-performance liquid chromatography with electrochemical detection. Journal of Chromatography, 383:375–386. Na-Bangchang, K. and Congpuong, K. (2007). Current malaria status and distribution of drug resistance in East and Southeast Asia with special focus to Thailand. Journal of Experimental Medicine, 211; 99–113. NCID (2004). Retrieved from: www.cdc.gov/malaria/biology/mosquito/index.htm, National Centre for Infectious Diseases, Division of Parasite Diseases, USA. 168 Nkoma, S., Molyneux, M. and Ward, S. (2007). In vitro antimalarial susceptibility profile of and Pfct/mdr-1 genotypes of Plasmodium falciparum filed isolates from Malawi. The American Society of Tropical Medicine and Hygiene, 76(6), 1107–1112. Newton, P., Suputtamongko, Y., Teja-isavadharm, P., Pukrittayakamee, S., Navaratnam, V., Bates, I. and White, N. (2000), Antimalarial Bioavailability and Disposition of Artesunate in acute falciparum malaria, Antimicrobial agents and chemotherapy 972–977. Nosál, R., Ericsson, O., Sjöqvist, F., Durisová, M. (1988). Distribution of chloroquine in human blood fractions. Methods Find Experimental Clini cal Pharmacology, 10(9):581-7 RY Nzila, A.M., Mberu, E.K., Sulo, J., Dayo, H., Winstanley, P.A., SibAley, C.H. and Watkins, W.M. (2000). Towards an understanding of thRe mechanism of pyrimethamine-sulphadoxine resistance in PlasImBodium falciparum: Genotyping of Dihydrofolate Reductase and Dihydrofolate synthase of Kenyan parasites. Journal of Antimicrobial Agent and Chemotherapy, 44(4): 991-996. L Obua, C., Hellgren, U., Ntale, M., Gustafsson L.LA., ONgwal-Okeng, J.W., Gordi, T., Jerling, M. (2008). Population phDarmacokinetics of chloroquine and sulfadoxine and treatment responseA in children with malaria: suggestions for an improved dose regimen. BrBitish Journal of Clinical Pharmacology; 65: 493–501. I Oduola, A.M., Milhous, W.K., Sala ko, L.A., Walker, O., Desjardins, R.E. (1987). Reduced in-vitro susOceptFibility to mefloquine in West African isolates of Plasmodium falcipa rum. Transaction of Royal Society of Tropical Medicine and Hygiene, 2(8Y571):1304-5. Oduola, A.M.J., MToya-Some, R.S., Kyle, D.E., Martin, S.K., Gerena, L., and Milhous, WI.K. (1989). Chloroquine resistance P. falciparum in indigenous residRent Sof Cameroon. Transaction of Royal Society of Tropical Medicine and Hygiene, 83: 308-310. OguInVbon Ea FA, Onyeji CO, Lawal AA, Chukwuani CM, Bolaji OO (1986) Liquid chromatographic analysis of chloroquine and desethylchloroquine in human N plasma, saliva and urine. Journal of Chromatography, 380:425-430. UOgunbona, FA., Lawal, A.A. and Onyeji, C.O. (1986). Saliva secretion of chloroquine in man. Journal of Pharmacy and Pharmacology, 38(7):535-7. Okocha, E.C., Ibeh, C.C., Ele, P.U. and Ibeh, N.C. (2005). The prevalence of malaria parasitaemia in blood donor in Nigerian teaching hospital. Journal of Vector Borne Diseases. 42 (1):21-4 Olatunde, I.A. (1970). Parenteral chloroquine in children. West. African Medicine Journal. 19: 93-99. 169 Olliaro, O., Nevill, C. and Lebras, J. (1996) Systematic review of amodiaquine treatment in uncomplicated malaria. Lancet, 348: 1196-1201. Olumese, P. (2005). Epidemiology and surveillance: Changing the global picture of malaria—myth or reality?. Acta Tropica 95: 265-269. Olliaro, P. (2004). Congenital toxoplasmosis. Clinical Evidence. 12:1058-61. Onyeji, C.O. and Ogunbona, F.A. (1996). Time-dependent variability of chloroquine secretion into human saliva. Pharmaceutical World Science. 18(6):211-6. Orrell, C., Little, F., Smith, P., Folb, P., Taylor, W., Olliaro, P. and BarneYs, K .I. (2008). Pharmacokinetics and tolerability of artesunate and amodiaquine alone and in combination in healthy volunteer. 64:683-690. R Panisko, D. M., and Keystone, J. S (1990). Treatment of malariRa, DArugs 39:160–189. B Philips-Howard, P.A. and ter Kuile, F.O. (1995). CNS ad vLersIe event associated with antimalarial agents. Fact or Fiction? Drug Safety, 12:370-383. Pitmang, S.L., Thacher, T.D., Madaki, K.J.A., EgAah, ND.Z. and Fischer, P.R. (2005). Comparison of Sulphadoxine-pyrimethaDmine with and without chloroquine in uncomplicated malaria in Nigeria. American Journal of Tropical Medicine and Hygiene, 72(3), 263-266. A Plowe, C.V. (2007). Combination IthBerapy for malaria: mission accomplished?. Clinical Infectious DiseaseFs. 44(8):1075-7. Powell, R.D. and McNama rOa, J. (1966). Quinine: side effects an plasma levels. Proceedings of thYe Helminthological Society of Washington, 30, 331-338 Pukrittayakamee, ITS., Prakongpan, S., Wanwimolruk, S., Clemens, R., LooareesSuwan S., and White N. J.. (2003). Adverse effect of rifampin on quinine efficacy in uncomplicated falciparum malaria. Antimicrobial Agents and CRhemotherapy, 47:1509–1513. QuasVhie,E N.B., Akanmori, B.D., Goka, B.Q., Ofori-Adjei and Kurzhals, J.A.L. I (2005). Pretreatment blood concentration of chloroquine in patients with N malaria infection: Relation to response to treatment. Journal of Tropical Pediatrics, 51(3):149-163. U rdRang, H.P., Ritter, J.M., and Dale, M.M., (1997). Basic Pharmacology. 3 Edition Churchill Livingstone, Pp 765-766. Ridtitid, W, Wongnawa, M., Mahatthanatrakul, W., Raungsri, N., Sunbhanich, M. (2005). Ketoconazole increases plasma concentrations of antimalarial mefloquine in healthy human volunteers. Journal of clinical Pharmacology and Therapeutics. ;30(3):285-90. Rio, B., Barriott, P., Rimailho, A. and Baud, F.J. (1988). Treatment of severe chloroquine poisoning. New England Journal of Medicine. 318, 1-6. 170 Roche, R.J., Silamut K., Pukrittayakamee, S., Looareesuwan, S., Molunto, P., Boonamrung, S. and White, N.J. (1990). Quinine iduces reversible high-tone hearing loss. British Journal of Clinical Pharmacology, 29:780-782. Rosenthal PJ (2008). Artesunate for the treatment of severe falciparum malaria. New England Journal of Medicine, 358:1829-36. RBM. (2005). Roll Back Malaria: Overview of malaria control activities and programme progress in Nigeria. RBM, (2006). Roll back Malaria: http://www.rollbackmalaria.org/keyfacts.html Ronn, A.M., Lemnge, M.M., Angelo, H.R., (1995). High performanRce Yliquid chromatography determination of dapsone, monoacetyldAapsone, and pyrimethamine in filter paper blood spots. Therapeutic Drug Monitoring, 17:79-83. R Safeukui-Noubissi I., Rangue S., Poudiougou B., (2004). RiBsk factors for severe malaria in Bamako, Mali: a matched case-control sLtudIy. Microbes infection, 6: 572-278. Salako, L.A., Sowunmi, A. and Akinbami, F.O. (1A989N). Pharmacokinetics of quinine in African children suffering from kwDashiorkor. British Journal of Clinical Pharmacology, 28, 197-201 Salako, L.A., Ajayi, F.O., Sowunmi, AB., AI Walker, O. (1990). Malaria in Nigeria: revisit. Annals of Tropical Me dicine and Parasitology, 84, 435-445. Salako, L.A. and Sowunmi, A. (F1992). Disposition of quinine in plasma, red blood cells and saliva afte r Ooral and intravenous administration to healthy adult Africans. EuropeYan Journal of Clinical Pharmacology; 42: 171-174. Sanchez, C.P., WuInTsch, S., and Lanzer, M. (1997). Identification of chloroquine importer Sin P. falciparum. Journal of Biological Chemistry. 272: 2652-2658. Schapira, AR., Bygbjerg, I.C., Jepsen, S., (1986). The susceptibility of Plasmodium faElciparum to sulphadoxine and pyrimethamine : correlation of in-vivo and in Vvitro results. American Journal of Tropical Medicine and Hygiene, 35 :239-I 245. SNchellenberg, D., Abdulla, S., Roper, C. (2006). Current issues for anti-malarial U drugs to control P. falciparum malaria. Current Molecular Medicine, 6(2):253-60. Serghides, L. and Kain, K.C. (2001). Peroxisome proliferator-activated receptor gamma-retinoid X receptor agonist increase of CD36- dependent phargocytosis of Plasmodium falciparum-parasitised erythrocytes and decrease malaria-induced TNF-alpha secretion by monocytes/macrophages. Journal of Immunology 166 (11): 6742-8. 171 Shanks, G.D., Watt, G., Edstein, M.D., Webster, H.K., Loesuttiviboon, L. and Wechgritaya, S. (1992). Halofantrine given with food for falciparum malaria. Transaction of Royal Society of Tropical Medicine and Hygiene. 86: 233-234. Shaper, A.G. and Lewis, P. (1971). Genetic neutropenia in people of African origin. Lancet, ii, 1021-1023. Sibley, C.H., Hyde, H.E., Sims, P.F.G., Plowe, C.V., Kublin, J.G., Mberu, E.K., Cowman, A.F., Winstanley, P.A., Watkins, W.M., and Nzila, A.M. (2001). Pyrimethamine-sulphadoxine in Plasmodium falciparum: What next?. Tre nds in Parasitology. 17(12) 582-588. Simpson, J.A., Hughes, D., Manyando, C. (2006). Population pharmacokRineYtic and pharmacodynamic modeling of the antimalarial chemotherapy chlorproguanil/dapsone. British Journal of Clinical PharmacoAlogy, 61:289- 300. R Slater, A.F.G. (1993). Chloroquine mechanism of drug actioInB and resistance in P. falciparum. Pharmacological Therapeutics. 57:203 -L235. Snow, R. W., Guerra, C. A., Noor, A. M., Myint, H.N Y. and Hay, S. I. (2005). The global distribution of clinical episodes of Plasmodium falciparum malaria. Nature, 434, 214–217 A Soentjens, P., Delanote, M. and Van GomDpel, A.(2006). Mefloquine-induced pneumonitis. Journal of Travel MedAicine, 13(3):172-4 Sowunmi, A., Fateye, B.A., Adedeji, AI.BA., Gbotosho, G.O., Happi, T.C., Bamgboye, A.E., Bolaji, O.M. & OdFuola, A.M.J. (2006). Predictors of the failure of treatment with pyrimOethamine-sulfadoxine in children with uncomplicated, falciparum malaria. Acta Tropica, 98, 6-14. Sowunmi, A., FateyeT, BY.A, Adedeji, A.A., Fehintola, F.A., Gbotosho, G.O., Happi, T.C., TamboI, E. and A Oduola, M.J. (2005). Predictors of the failure of treatmentS with chloroquine in children with acute, uncomplicated, PlasmRodium falciparum malaria, in an area with high and increasing incidences of chloroquine resistance. Annals of Tropical Medicine and PaErasitology, 99(6): 535-544. SowIuVnmi, A., Fateye, B.A., Adedeji, A.A., Fehintola, F.A., Happi, T.C. (2004). Risk N factors for gametocyte carriage in uncomplicated falciparum malaria in U children. Parasitology, 129, 255-262. Sowunmi, A., Oduola, A.M.J., Salako, L.A., Ogundahunsi, O.A.T., Laoye, O.J., and Walker, O., (1992). The relationship between the response of P. Falciparum malaria to mefloquine in African Children and its sensitivity in-vitro. Transaction Royal Society Tropical Medicine and Hygiene,86: 368-371.. Sowunmi, A. and Salako, L.A. (1992). Evaluation of the relative efficacy of various antimalarial drugs in Nigerian children under five years of age suffering from acute uncomplicated falciparum malaria. Transaction Royal Society Tropical Medicine and Hygiene, 89: 303 - 05. 172 Sowunmi, A., Salako, L.A., Walker, O. and Ogundahunsi, O.A.T. (1990). Clinical efficacy of mefloquine in children suffering from resistant falciparum malaria in Nigeria. Transaction Royal Society Tropical Medicine and Hygiene, 84:761-764. Soyinka, J.O., Onyeji, C.O., Omoruyi, S,I., Owolabi, A.R., Sarma, P.V. and Cook, J.M. (2009). Effects of concurrent administration of nevirapine on the disposition of quinine in healthy volunteers. Journal of Pharmacy and Pharmacology, 61(4):439-43. Stepniewska, K., Taylor, W., Sirima, S.B., Ouedraogo, E.B., Ouedraogo, A., Gansané A., Simpson, J. A., Morgan, C.C., White, N.J. and KiecheYl, J .N. (2009). Population pharmacokinetics of artesunate and amodiRaquine in African children. Malaria Journal, 20th August. A Steveson, M. M. and Zavala, F. (2005). Immunology of MRalaria Infections: Implications for the Design and Development of MalaIriBa Vaccines was held on 8–11 February 2005, in Baltimore, MD, USAL, at the Johns Hopkins Bloomberg School of Public Health. Talisuna, A.O., Bloland, P., D’alessandro, U. (2004). History, dynamics, and public health importance of malaria parasite resisAtanNce. Clinical Microbiology, 17, 235-254. D Ter Kuile, F.O., Nosten, F., LuxemburgerA, C., and White, N.J. (1993). Mefloquine prophylaxis. Lancet, 342: 551. IB Touze, J.E., Fourcade, L., Peyron , F., Heno, P. and Deharo, J.C., (1997). Is halofanthrine still advOisabFle in malaria attacks? Annals of Tropical Medicine and Parasitology. 91 : 867-873. Tracy, J.W. and WeTbsteYr, L.T., (1996). Drugs used in the chemotherapy of Protozoa infections: MIalaria. Goodman & Gilman, The pharmacological Basis of thTherapeuStics 9 Ed. McGraw Hill Companies, New York. Trenqne, T., Simon, N., Villena, I., Chemia, C., Quereux, C., Leroux, B., Jaussaud, RE., RRemy, G., Dupouy, D., Millart, H., Pinon, J.M., Urien, S. (2004). VPopulation pharmacokinetics of Pyrimethamine and sulphadoxine in children I with congenital toxoplamosis. British Journal of Clinical Pharamcology N ,57:735-741. UTriglia, T., and A. F. Cowman. 1994. Primary structure and expression of the dihydropteroate synthetase gene of Plasmodium falciparum. Procedure of National Accademy of Science, USA 91:7149–7153. Triglia, T., J. G. T. Menting, C. Wilson, and A. F. Cowman. 1997. Mutations in dihydropteroate synthase are responsible for sulfone and sulfonamide resistance in Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 94:13944 – 13949. Tuboku-Metzeger, A.F. (1964). Chemotherapy of malaria. British Medical Journal. 1: 1378. 173 Ursing, J., Kofoed, P.E., Rodrigues, A., Bergqvist, Y., Rombo, L. (2009). Chloroquine is grossly overdosed and overused but well tolerated in Guinea- bissau. Antimicrobial Agents and Chemotherapy, 53(1):180-5. Van-Daillen, J., Custers, M. and Wensink A. (1999). A comparison of amodiaquine and sulphadoxine-pyrimethamine as first-line treatment of falciparum malaria in Kenya. Transaction of Royal Society of Tropical Medicine and Hygiene. 93(2):185-188. Van Vugt, M., Ezzet F., Phaipun, L., (1998). The relationship between capillary and venous concentration of the antimalarial drug lumefantrine (benflumetol). Transaction of Society of Tropical Medicine and Hygiene, 92:564-565. Y Vallely, A., Vallely, L., Changalucha, J., Greenwood, B., Chandramohan, RD. (2007). Intermittent preventive treatment for malaria in pregnancy in AAfrica: what's new, what's needed? Malaria Journal, Feb 16;6:16. R Walker, O. and Ademowo, O.G. (1996). A rapid IcBost-effective liquid chromatography method for the determination of chloroquine and desethylchloroquine in biological fluids. NJou r Lnal Therapeutics Drug Monitoring, 18(1) 92-96. Walker, O., Dawodu, A.A., Adeyokunnu, A.AD., SAalako, L.A. and Alvan G. (1983). Plasma chloroquine and desethyl Achloroquine concentrations in children during and after chloroquine treatment for malaria. British Journal of Clinical Pharmacology, 16: 701-705 IB Wang, P., Read, P.M., Sims, P.FF., a nd Hyde, J.E. (1997). Sulfadoxine resistance in the human malaria Oparasite Plasmodium falciparum is determined by mutation in dyhydro pteroate synthase and additional factors associated with folate utilization. YJournal of Molecular Microbiology, 23:979-986. Watkins, W.M., OIloTo, J.A., Lury, J.D., Mosoba, M., Kariuki, D., Mjomba, M., Koech, DS.K., and Gilles, H.M. (1988). Efficacy of multiple dose of halofanthrine in treatment of chloroquine-resistant falciparum malaria in childrRen in Kenya. Lancet ii :247-250. WeidVekaEmm, E., Polzza-Nottebrock, H., Forgo, I. and Dubah, U.C. (1982). Plasma I concentration of pyrimethamine and sulfadoxine and evaluation of N pharmacokinetics data by computerized curve fitting. Bulletin of the world U Health Organisation, 60, 115-112. Weidekamm, E., Schwartz, E., Dubach, U.C. and Weber, B. (1987). Single-dose investigation of possible interactions between the components of the antimalarial combinaion Fansimef, Chemotherapy, 33, 259-265. Wernsdorfer, W. H. and Payne, D. (1991) The dynamics of drug resistance in Plasmodium falciparum. Journal of Pharmacology and Therapeutics, 50:95– 121. 174 White, N.J., Stepniewska, K., Barnes, K., Price, R.N., and Simpson, J. (2008). Simplified antimalarial therapeutic monitoring: using day-7 drug level?. Trend in Parasitology. 24(4):159-163. White, N.J. (2007). Cardio toxicity of antimalarial drugs. Lancet Infectious Diseases; 7:549–58. White, N.J. (2005). Intermittent presumptive treatment for malaria. A better understanding of pharmacodynamics will guide more rational policymaking. PloS Medicine. 2:29-33 White, N.J. (2004). Antimalarial drug resistance. The Journal of Clini cal Investigation 113(8)1084-1092. RY White, N.J. (1997). Assessment of the pharmacodynamic properties oAf antimalarial drugs in vivo. Antimicrobial Agents and Chemotherapy, 41, R1413-1422. White, N.J. (1994). Antimalarial drug resistance and combinBation chemotherapy. Journal of Clinical Investigation, 113 (8): 1084-10 92L. I White, N.J. (1992). Antimalarial drug resistance: the pace quickens. Journal of Antimicrobial Agent and Therapeutics. (5):5A71-N85. White, N.J. and Olliaro PL. (1996). Strategies Dfor the prevention of antimalarial drug resistance: rationale for combinationA chemotherapy for malaria. Parasitology Today, 10 :399-401. B White, N.J., Miller, K.D., ChurchFill, FI.C., Berry, C.D., Brown, J., William, S.B. and Greenwood, B.M. (1988). Chloroquine treatment of severe malaria in children: pharmacokinetics, toxicity and new dosage recommendation. New England Journal Med iOcine, 319: 1493-1509. White, N.J. (1987b)I. TThYe pharmacokinetics of quinine and quinidine in malaria. Acta LeidensiaS 55, 65-76. White, N.J.,R Looareesuwan, S., Edwards, G., Phillips, R.E., Karbwang, J., Nicholl, DE.D., Bunch, C. and Warrell DA (1987a) Pharmacokinetics of intravenous Vamodiaquine. British Journal of Clinical Pharmacology. 23:127–135. WNhiItty, C.J., Edmonds, S., Mutabingwa, T.K. (2005). Malaria in pregnancy, British Journal of Gynecology. 112 (9):1189-95. UWHO (2000). The African summit on Roll Back Malaria, Abuja, Nigeria 25 April 200. Geneva, Http://rbm.who.int/docsabuja_declaration.pdf. WHO (2003). Assessing and monitoring of antimalarial drug efficacy for the treatment of uncomplicated falciparum malaria. Annex 9. Classification of treatment outcomes. WHO (2006). Guidelines for the treatment of malaria. ISBN 978 92 4 154694 2 WHO/HTM/MAL/2006.1108. 175 WHO (2007). Malaria in pregnancy guidelines for measuring key monitoring and evaluation indicator. WHO (2009). Access to treatment and parasite resistance lead agendas.TDR News No. 84; P 22-23 Williams, A. R. F. (1966). Malaria in children. British Medical Journal. ii:1531. Wilairatana, P., Krudsood, S., Trrprasertsuk, S., Silachamroom, U., Looareesuwan S. (2002). Multidrug-resistance Malaria: Research on Antimalarials and where do we go from here?. International Medical Journal of Thaila nd, 18;(3),27.6-280 Y Wilson, T.J. (1993) Clinical correlates of drugs in saliva. Annals of RNew York Acaddemics of Science. 694: 48-61 A Winstanley, P. A., Watkins, W. M., Newton, C. R., Nevill, C., MbReru, E., Warn, P. A., Waruiru, C. M., Mwangi, I. N., Warrell, D. A. and IMBarsh. K. (1992). The disposition of oral and intramuscular pyrimethamine/sulphadoxine in Kenyan children with high parasitaemia but clinically non- sLevere falciparum malaria. British Journal of Clinical Pharmacology. 33:1N43–148. Winstanley, P.A., Edwards, G., Orme, M.L., BreAckenridge, A.M. (1987). Effect of dose size on amodiaquine pharmacDokinetics after oral administration. European Journal of Clinical pharmAacology, 33(3):331–333. WMR (2010). World B Malaria Report, Nigeria; http://www.who.int/malariFa/p ubIlications/country-profiles/profile_nga_en.pdf Yartey, J.E. (2006). Mala riOa in pregnancy: access to effective interventions in Africa. InternatioYnal Journal of Gynecology and Obstetrics, 94(3):364-73. T RS I E NI V U 176 RY A LIB R Appendices N AD A F I B O TY SI VE R I UN 177 . Table 4.a: Standard curve table for extrapolation of unknown concentratiYon o f chloroquine in whole blood on filter paper. AR R LI B Concentration CQ peak Papaverine (PPV) NRatio of CQ/PPV Height (mV) peak Height (mV) peak Height (ng/mL) DA 0 0 6.1 A 0 100 2.8 6.4 IB 0.4375 200 6.5 OF6.7 0.9701 400 12.4 Y 6.8 1.8235 800 21I.2T 6.2 3.4193 1000 RS26.1 5.8 4.5000 2000 E 48.3 5.3 9.1320 2 r = 0.V99158 UN I 178 Table 4.b: Standard curve table for extrapolation of unknown concentration of sulphadoxine in whole blood on filter paper Concentration Sulphadoxine Sulisoxazole Ratio of (IS)Peak Height SDX/SXZ (µg/mL) Peak Height (mV) (mV) Peak Heights 0 0 110.9 0 AR Y 10 45.9 107.0 0.B428R9 I 20 123.6 171.7 N L0.7198 40 146.8 145.3 A 1.0103 50 220.0 1A47.D0 1.4959 60 219.9 IB106.9 2.0579 2 r = 0.99158 F Y O T RS I VE UN I 179 Table 4.c: Standard curve table for extrapolation of unknown concentration of desethyl amodiaquine in plasma. Concentration IS (QND) DAQ Peak heights (ng/ml) peak height peak height DEAQ/QND ratio 0 31.6 - 0.00 Y 100 36.7 3.2 0.08 R 200 38.7 4.9 0.12 RA 400 41.7 13.0 0.31 IB 600 36.0 18.3 0 .5L0 800 37.1 22.5 N0.60 2 r = 0.9907 DA IB A F Y O T RS I E NI V U 180 Table 4.d: Standard curve table for extrapolation of unknown concentration of desethyl amodiaquine in saliva. Concentration IS (QND) peak DEAQ peak Peak height (ng/ml) height height DEAQ/QND ratio 0 30.9 - 0.00 100 32.4 13.6 0.41 Y 200 24.4 14.6 0.59 AR 400 28.8 29.4 1.02 R 600 27.9 42.8 1.5L3 IB 800 28.7 60.0 N2 .09 2 r = 0.9838 A D BA I O F Y T SI ER IV UN 181 RY BR A LI DA N BA F I Y O IT ER S V UN I 182