+ All Categories
Home > Documents > ONAKU, LINDA O. PG/ M.PHARM/09/51916 Linda.pdf · 1.3.2.1 Mechanism of action of artesunate - - ......

ONAKU, LINDA O. PG/ M.PHARM/09/51916 Linda.pdf · 1.3.2.1 Mechanism of action of artesunate - - ......

Date post: 09-Jul-2018
Category:
Upload: dinhdien
View: 230 times
Download: 0 times
Share this document with a friend
124
i ONAKU, LINDA O. PG/ M.PHARM/09/51916 EVALUATION OF COMBINATIONS OF ARTESUNIC ACID AND AQUEOUS EXTRACT OF Azadirachta indica OR Carica papaya ON THE REDUCTION OF PARASITEMIA IN MICE INFECTED WITH Plasmodium berghei PHARMACEUTICAL SCIENCES A THESIS SUBMITTED TO THE DEPARTMENT OF PHARMCEUTICS, FACULTY OF PHARMACEUTICAL SCIENCES, UNIVERSITY OF NIGERIA, NSUKKA Webmaster Digitally Signed by Webmaster’s Name DN : CN = Webmaster’s name O= University of Nigeria, Nsukka OU = Innovation Centre NOVEMBER, 2010
Transcript

i

ONAKU, LINDA O.

PG/ M.PHARM/09/51916

PG/M. Sc/09/51723

EVALUATION OF COMBINATIONS OF ARTESUNIC ACID

AND AQUEOUS EXTRACT OF Azadirachta indica OR Carica

papaya ON THE REDUCTION OF PARASITEMIA IN MICE

INFECTED WITH Plasmodium berghei

PHARMACEUTICAL SCIENCES

A THESIS SUBMITTED TO THE DEPARTMENT OF PHARMCEUTICS, FACULTY OF

PHARMACEUTICAL SCIENCES, UNIVERSITY OF NIGERIA, NSUKKA

Webmaster

Digitally Signed by Webmaster’s Name

DN : CN = Webmaster’s name O= University of Nigeria, Nsukka

OU = Innovation Centre

NOVEMBER, 2010

ii

EVALUATION OF COMBINATIONS OF ARTESUNIC ACID AND AQUEOUS

EXTRACT OF Azadirachta indica OR Carica papaya ON THE REDUCTION OF

PARASITEMIA IN MICE INFECTED WITH Plasmodium berghei

BY

ONAKU, LINDA O.

PG/ M.PHARM/09/51916

A PROJECT REPORT SUBMITTED TO THE DEPARTMENT OF

PHARMACEUTICS, FACULTY OF PHARMACEUTICAL SCIENCES, IN

PARTIAL FULFILMENT OF THE REQUIREMENTS FOR THE AWARD OF

MASTER OF PHARMACY (M.PHARM) DEGREE OF THE UNIVERSITY

OF NIGERIA NSUKKA.

NOVEMBER, 2010

iii

CERTIFICATION

This is to certify that Onaku Linda Onyeka, a postgraduate student in the

Department of Pharmaceutics, University of Nigeria Nsukka, with registration

number, PG/ M.PHARM/09/51916, has satisfactorily completed the research work for

the award of the Master of Pharmacy degree in pharmaceutics. The work embodied in

this project report is original and has not been submitted in part or full for any

diploma or degree in this or any other university.

Prof. A. A. Attama Date Prof. V. C. Okore Date

(SUPERVISOR) (CO-SUPERVISOR)

………………………………………………

Prof. A. A. Attama Date

(HEAD OF DEPARTMENT)

iv

DEDICATION

This research project is dedicated, first, to God; and then my father, Arch. P. C.

Onaku and my Mum, Lady Ify Onaku.

v

ACKNOWLEDGEMENT

The Lord has been my light and my help, and I am forever grateful to Him for making

this work see the light of the day.

The contributions of my lecturers, siblings, fellow students and friends to my

academic growth have been considerable. But, the guidance, sacrifice, patience, as

well as moral and academic support of my supervisors, Prof. A. A. Attama and Prof.

V. C. Okore, is deeply appreciated. The contributions of Prof. C. O. Esimone,

Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe

University, who spearheaded this project, are deeply appreciated.

I am forever indebted to my parents, Arch. P. C. Onaku and Lady Ify Onaku and my

uncle, Dr. Peter Oforah, whose moral and financial support has seen me through life

and especially through this project.

As far as this project is concerned, I would like to thank Dr. A. Y. Tijani, NIPRD,

Abuja, and Mr. Ngene, Faculty of Veterinary Medicine, UNN, whose assistance in

obtaining the Plasmodium berghei and determining the parasitemia levels

respectively, shall be bountifully rewarded by God.

Special thanks go to the entire staff of the Department of Pharmaceutics, UNN,

especially Mr. Ogboso Kalu (Expert), Mr. Gugu Thaddeus and Mr. Muogbo Chijioke,

and my mentor and friend, Prof. S. I. Ofoefule, for their assistance and advice.

Onaku, Linda .O

November, 2010.

vi

TABLE OF CONTENT

Title page - - - - - - - - - i

Certification - - - - - - - - - ii

Dedication - - - - - - - - - iii

Acknowledgement - - - - - - - - iv

Table of content- - - - - - - - - v

Abstract - - - - - - - - - ix

CHAPTER ONE: INTRODUCTION

1.1 Malaria - - - - - - - 1

1.1.1 Life cycle of malaria - - - - - - 4

1.1.2 Pathophysiology of malaria - - - - - 10

1.1.3 Biochemistry of the malaria parasite - - - - 11

1.1.3.1 Carbohydrate metabolism - - - - - 11

1.1.3.2 Lipid metabolism - - - - - - 12

1.1.3.3 Protein metabolism - - - - - - 12

1.1.3.4 Nucleotides and nucleic acid metabolism - - - 13

1.1.3.5 Vitamins and co-factors metabolism - - - - 13

1.1.3.6 Heme metabolism - - - - - - 14

1.1.4 The malaria parasite genome - - - - - 14

1.1.5 Diagnosis of malaria - - - - - - 15

1.2 Prevention of malaria - - - - - - 18

1.3 Treatment of malaria - - - - - 23

1.3.1 Drugs used in the treatment of malaria - - - 24

vii

1.3.2 Artesunate - - - - - - 27

1.3.2.1 Mechanism of action of artesunate - - - - 29

1.3.2.2 Pharmacokinetics of artesunate - - - - 29

1.3.2.3 Pharmacological actions of artesunate - - - 29

1.3.2.4 Side effects of artesunate - - - - 30

1.3.2.5 Artemisinin-based combination therapy - - - 30

1.4 Pharmacodynamic interaction - - - - 31

1.5 Antimalarial drug resistance - - - - - 33

1.6 WHO guildelines for treatment of uncomplicated malaria - 38

1.7 Plants with antimalarial activity - - - - 43

1.7.1 Antimalarial activity of Carica papaya - - - 45

1.7.2 Antimalarial activity of Azadirachta indica - - - 46

1.8 Methods employed in the evaluation of antimalarial

activity of a substance - - - - - - 49

1.8.1 In vitro methods for screening antimalarial compounds - 49

1.8.2 In vivo methods for screening antimalarial compounds - 50

1.8.2.1 Plasmodium berghei 4 day suppression test - - - 51

1.9 Objective of the study- - - - - - 52

CHAPTER TWO: MATERIALS AND METHODS

2.1 Materials- - - - - - - - 54

2.1.1 Experimental animals - - - - - - 54

2.1.2 The parasites - - - - - - - 54

2.1.3 Plants extract - - - - - - - 54

2.1.4 Chemicals- - - - - - - - 55

viii

2.2 Methods - - - - - - - - 55

2.2.1 Chemical preparation - - - - - - 55

2.2.1.1 Extraction of crude drugs - - - - - 55

2.2.1.2 Preparation of artesunate - - - - - 58

2.2.2 In vivo schizontocidal activity of combination of a fixed

dose of artesunate with varying doses of crude extracts - 58

2.2.3 Determination of ED50 of the crude extracts - - - 59

2.2.4 Determination of the kind of pharmacodynamic interaction

between the pure drug and plant extract - - - 60

2.2.5 Data analysis - - - - - - 60

CHAPTER THREE: RESULTS AND DISCUSSION

3.1 Chemical Preparation - - - - - - 61

3.1.1 Percentage yield of crude drugs - - - - 61

3.1.2 Preparation of stock solution of artesunate - - - 61

3.2 In vivo schizontocidal activity of combination of a fixed

dose of artesunate with varying doses of crude extracts - 62

3.2 Survival time and percentage cure of P. berghei infected

mice after treatment - - - - - - 70

3.4 Determination of ED50 of the crude extracts - - - 74

3.5 Determination of the kind of pharmacodynamic interaction

between the pure drug and plant extracts- - - - 78

CHAPTER FOUR: CONCLUSION - - - - - 82

References - - - - - - - - 84

ix

Appendices - - - - - - - - - 106

x

ABSTRACT

Fresh leaves of Azadirachta indica, and mature fresh leaves of Carica papaya were

separately homogenized in sterile cold distilled water for 24 hrs. During the study

each extract was stored in the fridge for maximum of seven days. The extraction was

done prior to the determination of the in vivo schizontocidal activity of the crude

extracts (NCE and PCE), and artesunic acid, and fresh extracts were made as needed.

The Peter’s 4-day suppressive test was the model used in this study. The ED50 and

ED90 were calculated from the dose-response relationship and the values obtained

were used to categorize the antimalarial activity of the crude extracts and the

Pharmacodynamic interaction of the combinations of the crude extracts and artesunic

acid. The average percentage yield of NCE (8.33 %) was higher than that of PCE

(5.42 %). It was found that a one –dose combination of 1000 mg/kg of NCE and 15

mg/kg of artesunic acid, produced a significant reduction of parasitemia (96.87 %)

when compared to artesunic acid alone at a dose of 20mg/kg (68.14 %). The

combination of 50 mg/kg of PCE and 15 mg/kg of artesunic acid produced a

significant reduction of parasitemia (81.25 %) when compared to 50 mg/kg of PCE

alone (37.70%). The ED50 of NCE and PCE showed that they have moderate and very

good activity respectively. The isobolar equivalent (IE) calculated from the ED90 of

NCE and PCE in combination with artesunic acid showed that the interaction between

artesunic acid and NCE is synergistic, while the interaction between artesunic acid

and PCE is antagonistic. The combinations of NCE and PCE with artesunate

produced a cure, while the given dose of artesunate did not produce cure during the

30-day period of the study. These results have showed that antiplasmodial

combinations of an artemisinin derivative and aqueous extract of neem leaf (NCE) are

possible, providing a potentiated reduction of parasitemia, and increased cure rate.

xi

xii

CHAPTER ONE

INTRODUCTION

1.1 Malaria

Malaria is an acute and chronic mosquito-borne disease of man caused by a

eukaryotic protist of the genus Plasmodium. The disease is characterized by chills and

fever, anemia, splenomegaly and damage to other organs such as the liver and brain

(1). Malaria is one of the world’s leading killers. Half of the world’s population –

about 3.3 billion people- is at risk. About 1.2 billion of these are considered to be in

high risk areas: Africa or South East Asia. Malaria has a greater morbidity and

mortality record than any other infectious disease (2, 3, 4). The incidence of mortality

is greatest in children and pregnant women (4). Reports have shown that in 2006, 86

% of the 247 million cases of malaria occurred in Africa. Over half of these cases

occurred in Nigeria, Democratic Republic of Congo, Ethiopia, United Republic of

Tanzania and Kenya. Out of the 881000 deaths that occurred from the disease, 91 %

were in Africa, and 85 % of these were children below 5 years (2). The disease has

negative impact on the economy of malaria- endemic countries (5, 6). Indeed HIV

also has a negative impact on the economy of a nation. HIV infection and malaria

complement each other. HIV infection increases a person’s susceptibility to malaria

infection, while malaria increases viral load of HIV patients (7). According to Centers

for Disease Control and Prevention, in 2008, an estimated 190 - 311 million cases of

malaria occurred worldwide and 708,000 - 1,003,000 people died, most of them

young children in sub-Saharan Africa (8).

Five species of the plasmodium parasite can infect humans and they are transmitted

naturally by the bite of an infected female anopheles mosquito. The most serious

forms of the disease are caused by Plasmodium falciparum. Malaria caused by

xiii

Plasmodium vivax, Plasmodium ovale and Plasmodium malariae causes milder

disease in humans that is not generally fatal. A fifth species, Plasmodium knowlesi, is

a zoonosis that causes malaria in macaques but can also infect humans (9, 10).

Typically, one species will cause malaria, but mixed infections do occur.

The life-cycle of plasmodia involves a sexual phase in the female mosquito and an

asexual stage in man. The asexual stage in man includes; the liver stage (tissue

schizonts), erythrocytic stage (erythrocytic schizonts); where the erythrocytes will

burst at intervals and bring about the febrile attack of malaria, and the gametocytic

stage, where the gametocytes (male and female gametes) also develop in some

infected erythrocytes, thus, this stage is responsible for the continual transmission of

malaria, as when the female mosquito feeds, they take in the erythrocyte housed

gametes, which then initiate the sexual stage. Therefore, a compound acting on the

gametes is very important.

Symptoms of malaria include; fever, shivering, arthralgia (joint pain), vomiting,

anemia (caused by hemolysis), hemoglobinuria, retinal damage, (11) and convulsions.

The classic symptom of malaria is cyclical occurrence of sudden coldness followed by

rigor and then fever and sweating lasting three to eight hours, occurring every two

days in P. vivax and P. ovale infections, while every three days for P. malariae (1).

P. falciparum can have recurrent fever every 36–48 hours or a less pronounced and

almost continuous fever. For reasons that are poorly understood, but that may be

related to high intracranial pressure, children with malaria frequently exhibit

abnormal posturing, a sign indicating severe brain damage (12). Malaria has been

found to cause cognitive impairments, especially in children. It causes widespread

anemia during a period of rapid brain development and also direct brain damage. This

neurologic damage results from cerebral malaria and children are more vulnerable

xiv

(13, 14). Cerebral malaria, a complication of severe malaria is associated with retinal

whitening, (15) which may be a useful clinical sign in distinguishing malaria from

other causes of fever (11).

Severe malaria is almost exclusively caused by P. falciparum infection, and usually

arises 6–14 days after infection. Consequences of severe malaria include coma and

death if untreated—young children and pregnant women are especially vulnerable.

Splenomegaly (enlarged spleen), severe headache, cerebral ischemia, hepatomegaly

(enlarged liver), hypoglycemia, and hemoglobinuria with renal failure may occur.

Renal failure is a feature of black water fever, where hemoglobin from lysed red

blood cells leaks into the urine. Severe malaria can progress extremely rapidly and

cause death within hours or days (16). In the most severe cases of the disease, fatality

rates can exceed 20%, even with intensive care and treatment (17). In endemic areas,

treatment is often less satisfactory and the overall fatality rate for all cases of malaria

can be as high as one in ten (18). Over the longer term, developmental impairments

have been documented in children who have suffered episodes of severe malaria (19).

Chronic malaria is seen in both P. vivax and P. ovale, but not in P. falciparum

infections. Here, the disease can relapse months or years after exposure, due to the

presence of latent parasites in the liver. Describing a case of malaria as cured by

observing the disappearance of parasites from the bloodstream can, therefore, be

deceptive. The longest incubation period reported for a P. vivax infection is 30 years

(16). Approximately one in five of P. vivax malaria cases in temperate areas involve

overwintering by hypnozoites (i.e., relapses begin the year after the mosquito bite)

(20).

Pregnant women are especially attractive to the mosquitoes, (21) and malaria in

pregnant women is an important cause of stillbirths, infant mortality and low birth

xv

weight, (22) particularly in P. falciparum infection, but also in other species infection,

such as P. vivax (23). Normal immune responses are reduced during pregnancy. In

areas of stable malaria transmission like Nigeria, a pregnant woman would have

acquired a partial immunity enough to protect against serious clinical falciparum

malaria, but heavy parasitic infection of the placenta and often severe anaemia do

occur leading to low birth weights of the baby, who may not even survive. First

pregnancies are at a greater risk. In areas of unstable malaria transmission, pregnant

women have no protective immunity and are at serious risk of developing severe life-

threatening falciparum malaria, especially in the last few months of pregnancy and for

several weeks after delivery. Untreated infection in this case can result in abortion,

still-birth, premature labour or low birth weight. Cerebral malaria, pulmonary oedema

and hypoglycaemia frequently occur. This situation is similar in all pregnancies in this

region (24).

1.1.1 Life cycle of the malaria parasite

Malaria parasites are members of the genus Plasmodium (Phylum Apicomplexa). In

humans, malaria is caused by P. falciparum, P. malariae, P. ovale, P. vivax and P.

knowlesi. (10, 25) P. falciparum is the most common cause of infection and is

responsible for about 80% of all malaria cases and about 90% of the deaths from

malaria (26). Parasitic Plasmodium species also infect birds, reptiles, monkeys,

chimpanzees and rodents (27). There have been documented human infections with

several simian species of malaria, namely P. knowlesi, P. inui, P. cynomolgi, (28) P.

simiovale, P. brazilianum, P. schwetzi and P. simium. However, with the exception of

P. knowlesi, these are mostly of limited public health importance (29). P. knowlesi

resembles P. malariae morphologically, but unlike P. malariae its parasite numbers in

the blood is high and clinical symptoms is more severe (24).

xvi

Malaria in humans develops via two phases: an exoerythrocytic and an erythrocytic

phase. The exoerythrocytic phase involves infection of the hepatic system, or liver,

whereas the erythrocytic phase involves infection of the erythrocytes, or red blood

cells. The exoerythrocytic phase does not produce any symptom: It is the erythrocytic

phase that produces symptoms. Malaria parasites are introduced as sporozoites,

present in the saliva of the infected mosquito, into the human body via the bite of the

female anopheles mosquito as they attempt to suck blood needed for them to lay eggs.

About 68 species of the 400 Anopheles spp. can transmit malaria, but the most

efficient vectors belong to the A. gambiae complex, which is widely distributed in

tropical Africa. The mosquitoes are attracted by increased CO2 concentration, warmth

of skin, moisture (breath), lactic acid, urine, feaces and most importantly, a number of

substances called kairomones. The normal body flora produces these kairomones.

But, malaria transmission can also occur congenitally or via inoculation of infected

blood (e.g. blood transfusion).

Within 30 min the sporozoites pass into the liver, their entry into the liver cell being

facilitated by circumsporozoite protein (CSP). The sporozoite carry out exo-

erythrocytic reproduction, i.e. an asexual reproduction called schizogony. Here, the

parasite grows and undergoes several nuclear divisions without the cytoplasm

dividing until it reaches a diameter of 30-70 nm, and this stage or form of parasite is

called liver schizonts. Division of the cytoplasm of the multicellular liver schizonts

occurs giving rise to thousands of merozoites (offspring) (24).

The infected liver cell burst after sometime to release merozoites into the blood

stream. The minimal time required from infection to the appearance of the first

merozoite, which lasts for 6-15 days, is called the prepatent period (30). Incubation

period is the minimal time from infection to appearance of signs and symptoms of

xvii

malaria, and is somewhat longer than the prepatent period. In the case of P.vivax and

P. ovale, the prepatent period extend to typically 6-12 months, as some liver cells do

not burst and will contain parasites called hypnozoites for a long time and they cause

new attacks of the disease if reactivated (recrudescence). They are also responsible for

delayed exacerbations of the disease post-treatment.

The released merozoites bind to the surface of the erythrocytes via merozoite surface

protein -1 (MSP-1) present on their membranes and then penetrate the red blood cell

and remain in a vacuole there. The MSP-1 is highly variable and the parasite may

change structural features within the course of a single infection. This enables it to

carry out immune evasion. The life-cycle is depicted in the diagram below.

xviii

Figure 1: Lifecycle of Plasmodium spp.- Ref 31

Merozoites undergo differentiation into trophozoites (ring form) that then feed on

heamoglobin. The haemozoin formed can be seen after 12-24 h as malaria pigment.

The vacuole of the trophozoites disappears as the parasite becomes older.

Trophozoites reproduce asexually to form schizont (multinucleated parasite).

xix

Schizonts divide to form merozoites (about 8-24 per schizont) within 48 h (as for P.

vivax, P. ovale and P. falciparium) or 72 h (as for P. malariae). Infected red blood

cells burst after a while to release merozoites which penetrates new erythrocytes in

seconds. The bursting is accompanied by a bout of fever, but the fever will not usually

follow a typical pattern as in every 48 or 72 h as all parasites cannot be at the same

stage of development.

Some merozoites transform into male and female gametocytes after a few days.

Gametocytes do not cause symptoms but are responsible for transmission of the

disease. Female anopheles mosquito may ingest gametocytes as they suck, and the

gametes once ingested divide mitotically three times and develop several flagellae

(exflagellation). Microgametes are formed after 10 min, from one male gametocyte. A

fall in temperature and gametocyte activating factor (xanthurenic acid, present at a

higher concentration in mosquitoes than in the human blood) triggers exflagellation.

The female gametocyte undergoes a slight change to produce macrogametes.

Microgamete and macrogametes will fuse to form a diploid zygote (parasite is

haploid). This zygote then undergoes meiosis to give four haploid parasites, which

later become motile and are called ookinete. The ookinete penetrates a membrane to

pass from the mosquito’s midgut to the intestinal wall, and it migrates through this

wall to the outside of the intestine. Here, the ookinete changes into an immobile form

called oocyst. After a week of repeated mitotic nuclear division in the oocyst,

countless of fusiform parasite called sporozoites are produced, which after rupture of

the oocyst will migrate to the mosquito’s thoracic three-lobed salivary glands.

Sporozoites mature in the salivary gland and are ready to be injected into a human

during a blood meal (24, 32).

xx

The parasite is relatively protected from attack by the body's immune system because

for most of its human life cycle it resides within the liver and blood cells and is

relatively invisible to immune surveillance. However, circulating infected blood cells

are destroyed in the spleen. To avoid this fate, the P. falciparum parasite displays

adhesive proteins on the surface of the infected blood cells, causing the blood cells to

stick to the walls of small blood vessels, thereby sequestering the parasite from

passage through the general circulation and the spleen (33). This "stickiness" is the

main factor giving rise to hemorrhagic complications of malaria. High endothelial

venules (the smallest branches of the circulatory system) can be blocked by the

attachment of masses of these infected red blood cells. The blockage of these vessels

causes symptoms such as in placental and cerebral malaria. In cerebral malaria the

sequestrated red blood cells can breach the blood brain barrier possibly leading to

coma (34).

Although the red blood cell surface adhesive proteins called Plasmodium falciparum

erythrocyte membrane protein 1 (PfEMP1) are exposed to the immune system, they

do not serve as good immune targets, because of their extreme diversity; there are at

least 60 variations of the protein within a single parasite and effectively limitless

versions within parasite populations (33). The parasite switches between a broad

repertoire of PfEMP1 surface proteins, thus staying one step ahead of the pursuing

immune system.

1.1.2 Pathophysiology of malaria

xxi

When the schizonts in the erythrocytes mature and rupture they release merozoites,

cellular debris and pigments which stimulates the secretion of cytokines from

leucocytes and other cells. This then causes the fever characteristic of malaria attack.

The fever is usually severe and irregular or continuous at the early stage of infection,

but subsequent attack may be milder and regular, every 72 h for P. malariae, or every

48 h for P. ovale and P.vivax. Typical fever attack starts with a cold stage (rigor)

where the patient shivers and feels cold, although his or her temperature is rising. This

is followed by a hot stage where the patient’s temperature rises to maximum and he or

she feels severe headache, arthralgia, back pains, and the patient may have diarrhea

and may vomit. The last stage of this fever attack is called the wet stage, whence the

patient perspires, the temperature falls, pains and headache are relieved and the

patient feels exhausted. There is always some degree of splenomegaly and anaemia,

and jaundice may occur but are particularly seen in falciparum malaria. Other

pathological changes occur depending on the species of plasmodium causing the

malaria.

Malaria caused by P. falciparum was formerly called malignant tertian malaria. The

untreated infection causes fever, splenomegaly, gastrointestinal disturbances, edema,

anaemia, postural hypotension, mental confusion, profound weakness etc.

Cytoadherence of parasitized red blood cells do occur, where these red blood cells

adhere to one another and to the walls of the capillaries of the brain, muscle, kidney,

placenta etc., leading to congestion, hypoxia, and blockage and rupturing of small

blood vessels. If left untreated, severe malaria results, where the parasite attains high

level of parasitemia (up to 30-40 %); leading to hepatomegaly, jaundice, renal failure,

cerebral malaria, black water fever, delirium, coma and death. P. falciparum has no

latent liver form, so that relapse is due to recrudescence.

xxii

Malaria caused by P. vivax and P. ovale are called vivax and ovale malaria

respectively. Their infections are rarely life threatening and parasitemia levels rarely

exceed 2 %. Relapse is due to latent liver forms.

Malaria caused by P. malariae is called malariae malaria or quartan malaria. Its

parasitemia level is usually below 1 %, but its exoerythrocytic stage can last as long

as 30-40 days, and a serious complication called nephrotic syndrome do occur mostly

in children. This syndrome is caused by the deposition of antigen-antibody complexes

on the glomerular basement membrane of the kidney. It progresses to renal failure and

produces edema, marked proteinuria and low serum albumin level (1, 24).

1.1.3 Biochemistry of the malaria parasite

Malaria parasite obtains nutrients from its environment for the production of other

molecules and energy (catabolism), these are then used to maintain the homeostasis of

the parasite and for anabolism (growth and reproduction). Their metabolic pathways

differ from, but are intertwined with that of the human host, because of the intimate

relationship between the host and the parasite. These pathways and the enzymes

involved can be exploited in the design of therapeutic agent, viz; many antimalarials

affect the food vacuole of the parasite, a special organelle for the digestion of host

hemoglobin (35, 36). Its metabolic pathways are summarized below.

1.1.3.1 Carbohydrate metabolism

The blood stage of the parasite actively ferments glucose as a primary source of

energy through glycolysis. The process of glycolysis is similar to that of other

organism, with lactate being the end product. The infected erythrocyte utilizes 75

times more glucose than uninfected erythrocytes. Patients with severe malaria will

thus suffer hypoglycemia, and coupled with their lack of appetite will lead to

convulsion and shock. Enzymes of the pentose phosphate pathway have also been

xxiii

identified and provide some of the ribose sugars needed for nucleotide metabolism

and regeneration of reduced NADPH, utilized for biosynthesis or defense against

reactive oxygen intermediates (ROI).

Pyruvate from the glycolytic pathway is employed in an incomplete TCA cycle by the

blood stages of the parasite and a complete TCA cycle by the mosquito-borne stage.

The Hydrogen atoms produced here are employed in oxidative phosphorylation and

the CO2 produced is a by-product. Atovaquone has been shown to inhibit electron

transport and to collapse the mitochondrial membrane potential (37, 38).

1.1.3.2 Lipid metabolism

Malaria parasite needs a huge demand of lipid to grow and its lipid metabolism can be

targeted. The parasite has enzymes that are associated with type II fatty acid synthetic

pathway (also found in plants and prokaryotes) that appear to be located in the

apicoplast. The apicoplast is involved in biosynthesis of fatty acids, isoprenoid

precursors and heme. They also have enzymes involved in lipid synthesis from

glycerides and fatty acids, and in the remodeling of lipid polar head (39).

1.1.3.3 Protein metabolism

The blood stage obtains amino acids for protein synthesis from 3 sources;

I. Degradation of ingested haemoglobin (most abundant source of amino acids).

Up to 60 -80 % of the host total haemoglobin is digested into amino acids. At

20 % parasitemia, 110 g of haemoglobin will be consumed during 48 hrs.

II. De novo synthesis. The parasite can fix CO2 and so synthesize alanine,

aspartate and glutamate.

III. Uptake of free amino acid from the host plasma (or cells) (36, 40).

1.1.3.4 Nucleotides and nucleic acid metabolism

xxiv

Malaria parasites obtain preformed purines by salvage pathways and synthesize

pyrimidines de novo. The primary purine salvaged is hypoxanthine from the host

plasma. For de novo synthesis of pyrimidine, bicarbonates and glutamine are used and

will require co-factors like folates (41).

1.1.3.5 Vitamins and co-factors metabolism

Malaria parasites cannot utilize preformed folate from the host erythrocytes and must

synthesize it from p-aminobenzoic acid, glutamate triphosphate and glutamate. Other

preformed vitamins can be utilized by the parasite. Folic acid and its derivatives are

co-factors in the synthesis of nucleotides and amino acids. A dihydrofolate cycle is

involved in de novo pyrimidine synthesis. Dihydrofolate is reduced to tetrahydrofolate

(this pathway is inhibited by pyrimethamine). The tetrahydrofolate is methylated by

serine hydroxymethyltransferase and the resulting methylene tetrahydrofolate fuctions

as methyl donor. For example, thymidylate synthase catalyses the formation of

deoxythymidylatemonophosphate from deoxyuridinemonophosphate by transferring

the methyl group from methylene tetrahydrofolate and this methylene tetrahydrofolate

is recycled back to dihyrofolate in the process (41).

1.1.3.6 Heme metabolism

Heme, an important component of many enzymes, is synthesized by the parasites de

novo as the heme released from hemoglobin degradation by the parasite cannot be

utilized by it. Free heme is indeed toxic to the parasite as it destabilizes and lyses

parasite membranes, and inhibits the activity of several enzymes. To prevent this, the

heme is detoxified by

xxv

I. Sequestration of the free heme into the hemozoin or the malaria pigment.

II. A degradation reaction facilitated by hydrogen peroxide (an ROI) within the

food vacuole.

III. A glutathione-dependent degradation which occurs in the parasite’s cytoplasm

(42,43, 44).

Chloroquine and other 4-aminoquinolines inhibit hemozoin formation, as well as

other heme degradative processes, thus preventing the detoxification of heme. The

free toxic heme also leads to death of the parasite. Also, the electrons released during

the conversion of iron in the haemoglobin from its ferrous state to its ferric state (in

the heme) promotes the formation of ROIs, which can cause damage to lipids,

proteins and nucleic acids, and thus cellular damage. Superoxide dismutase, catalase

and other free radical scavengers prevent this oxidative damage (45).

1.1.4 The malaria parasite genome

The malaria parasite is haploid throughout the life cycle, except for a brief time after

fertilization. Plasmodium falciparum has 5200 genes located on 14 chromosomes.

DNA is also present in the mitochondria and apicoplast. The apicoplast is transmitted

via the macrogamete. A large number of these genes are responsible for evasion of the

host immunity. The average gene density is approximately 1 gene / 4338 base pairs.

The mapping of this genome sequence provides new avenues for research on possible

vaccines. The genome of P. berghei, a rodent malaria parasite used in the evaluation

of antimalarias and vaccines, is highly similar, both in structure and gene content,

with P. falciparum (32).

1.1.5 Diagnosis of malaria

Diagnosis of malaria is based on clinical criteria (clinical diagnosis) supplemented by

the detection of parasites in the blood (parasitological or confirmatory diagnosis).

xxvi

Clinical diagnosis or symptomatic diagnosis involves evaluation of the clinical

presentation of the patient. The clinical presentation is based on if it is acute

uncomplicated malaria or acute severe malaria or chronic malaria. For acute

uncomplicated malaria, the clinical presentation includes; fever (temperature > 37.5

˚C, splenomegaly or hepatomegaly (especially in children), nail bed pallor, and the

patients history may include; subjective fever, chills and shivers, headache, tiredness,

body pains and joint weakness (46,47). The diagnosis of malaria based on clinical

symptoms alone is not reliable, as it results in unnecessary expenditure and

inappropriate use of antimalarial drugs, and a delay in establishing the correct

diagnosis and treatment objectives for a patient. 46 However, the manifestation of

malaria may range from asymptomatic to mild to severe disease such that the patient

can even present with only fever, due to the development of some level of immunity.

Consequently, it has been suggested that in stable malaria high-transmission areas like

Nigeria, the occurrence of fever (> 37.5 ˚C ) or a history of fever and no other obvious

cause, in children under 5 years and pregnant women also presenting with

unexplained pallor, suggests malaria, and treatment should be administered without

delay (46,47).

The clinical features of severe malaria includes; prostration, impaired consciousness,

respiratory distress, multiple convulsions (>2), shock, pulmonary oedema, abnormal

bleeding, jaundice, haemoglobinuria, renal failure, severe anemia and hypoglycemia.

Complications of severe malaria includes; cerebral malaria- a complication of severe

malaria caused by P. falciparum and it is malaria with coma persisting for greater

than 30 min after a seizure (46). Another complication of severe falciparum malaria is

black water fever, with features like; irregular fever, jaundice, dyspnea, intravascular

xxvii

hemolysis, renal failure, uremia and hemoglobinuria (coke coloured urine) (1, 46, 47).

Severe malaria is a medical emergency.

Laboratory diagnosis involves the use of microscopic examination of blood films and

antigen test or rapid diagnostic tests, RDTs. Since Charles Laveran first visualized the

malaria parasite in blood in 1880, (48) microscopic examination of blood films has

been the mainstay in the diagnosis of malaria. Its obvious advantage being that it is

quantitative nature. It involves the use of thin or thick blood smears that has been

stained with Romanowsky stain, and determination of the percentage parasitemia or

number of parasite per µl of blood respectively (24).

Thin films are similar to usual blood films and allow species identification of the

Plasmodium type, because the parasite's appearance is best preserved in this

preparation. Thick films allow the medical laboratory scientist to screen a larger

volume of blood and are about eleven times more sensitive than the thin film. Indeed

an experienced person can detect parasite levels (or parasitemia) down to as low as

0.0000001% of red blood cells. Therefore, picking up low levels of infection is easier

on the thick film, but the appearance of the parasite is much more distorted and

therefore distinguishing between the different species can be much more difficult.

With the pros and cons of both thick and thin smears taken into consideration, it is

imperative to utilize both smears while attempting to make a definitive diagnosis (49).

In severe malaria hyperparasitemia (i.e. a thin blood smear report of > 5% parasitemia

and a thick blood smear report of > 250,000 per µl of blood) is seen (47).

One important thing to note is that P. malariae and P. knowlesi (which is the most

common cause of malaria in South East Asia) look very similar under the microscope.

However, P. knowlesi parasitemia increases very fast and causes more severe disease

than P. malariae, so it is important to identify and treat infections quickly. Rapid

xxviii

Diagnostic Test and Molecular Methods should be used to distinguish between the

two in this area (50).

Rapid Diagnostic Test (or Antigen-Capture assay or Dip Stick Test) is a number of

immunochromatographic tests that make use finger-stick or venous blood, the

completed test takes a total of 15–20 minutes, and the results are read visually as the

presence or absence of colored stripes on the dipstick, so they are suitable for use in

the field. The threshold of detection by these rapid diagnostic tests is in the range of

100 parasites/µl of blood (commercial kits can range from about 0.002% to 0.1%

parasitemia) compared to 5 by thick film microscopy. Their disadvantage is that they

are qualitative but not quantitative. They also only differentiate falciparium malaria

from non-falciparium malaria; they cannot distinguish between other types of malaria

not caused by P. falciparium (24). The first rapid diagnostic tests were using P.

falciparum glutamate dehydrogenase as antigen (51). PGluDH was soon replaced by

P.falciparum lactate dehydrogenase, a 33 kDa oxidoreductase. It is the last enzyme of

the glycolytic pathway, essential for ATP generation and one of the most abundant

enzymes expressed by P. falciparum. PLDH does not persist in the blood but clears

about the same time as the parasites following successful treatment. The lack of

antigen persistence after treatment makes the pLDH test useful in predicting treatment

failure. In this respect, pLDH is similar to pGluDH. Depending on which monoclonal

antibodies are used, this type of assay can distinguish between all five different

species of human malaria parasites, because of antigenic differences between their

pLDH isoenzymes (50). RDTs are used in areas where microscopy is not available or

the laboratory staff is not experienced at malaria diagnosis (24).

A newer method of diagnosis called the molecular method involves the use of

Polymerase Chain Reaction (PCR) and QT-NASBA (based on the PCR) (52). PCR is

xxix

more accurate than microscopy, but is more expensive and requires specialized

laboratories.

1.2 Prevention of malaria

Malaria can be prevented through prevention of its spread and protection of

individuals in endemic areas. Methods here include both environmental methods and

use of prophylactic treatment and includes; methods employed in mosquito

eradication, prevention of mosquito bites and use of prophylactic drugs. The

continued existence of malaria in an area requires a combination of high human

population density, high mosquito population density, and high rates of transmission

from humans to mosquitoes and from mosquitoes to humans. If any of these is

lowered sufficiently, the parasite will sooner or later disappear from that area, as

happened in North America, Europe and much of Middle East. However, unless the

parasite is eliminated from the whole world, it could become re-established if

conditions revert to a combination that favors the parasite's reproduction. Many

countries are seeing an increasing number of imported malaria cases due to extensive

travel and migration (50).

Brazil, Eritrea, India and Vietnam which are developing countries have successfully

reduced their malaria burden. Common success factors included conducive country

conditions, a targeted technical approach using a package of effective tools, data-

driven decision-making, active leadership at all levels of government, involvement of

communities, decentralized implementation and control of finances, skilled technical

and managerial capacity at national and sub-national levels, hands-on technical and

programmatic support from partner agencies, and sufficient and flexible financing

(53). Indeed, many researchers have argued that prevention of malaria is more cost-

xxx

effective than treatment in the long run (54). Methods employed in mosquito

eradication encompass methods used in vector control and includes;

I. Draining of wetland breeding grounds.

II. Improved sanitation

III. Poisoning of breeding grounds of mosquitoes or aquatic habitats of the larva

stages, by filling or applying oil to stagnant waters.

IV. Researchers are going on to produce genetically modified mosquitoes that will

be malaria-resistant and then introduce them in the wild so that they gradually

replace the existing mosquitoes. This technique is called the sterile insect

technique. However, this approach contains many difficulties and success is a

distant prospect (55).

A futuristic method of vector control based, on the idea of using lasers to kill flying

mosquitoes (56).

Prevention of mosquito bites can be carried out by;

A. Indoor residual spraying (IRS): Here insecticides are sprayed on the interior

walls of homes in malaria affected areas. This is because after feeding many

mosquito species rest on nearby surface while digesting the blood meal, and if

the surface happens to be an insecticide sprayed wall, the mosquito will be

killed before they can take another blood meal (50). DDT was the first

pesticide used for this purpose (54). But, because it was also used in

agriculture and there was thus, emergence of mosquitoes resistant to DDT.

The use of DDT has now been banned or has limited use in agriculture for

some time now. Therefore, DDT may be more effective as a method of

malaria-control now (50). The WHO currently advises the use of 12 different

insecticides in IRS operations. These include DDT and a series of alternative

xxxi

insecticides (such as the pyrethroids, permethrin and deltamethrin), to combat

malaria in areas where mosquitoes are DDT-resistant and to slow the

evolution of resistance (57). This public health use of small amounts of DDT

is permitted under the Stockholm Convention on Persistent Organic Pollutants

(POPs), which prohibits the agricultural use of DDT. However, because of its

legacy, many developed countries discourage DDT use even in small

quantities (58). The problem with IRS is insecticide resistance which is

brought about by evolution. Mosquito species that are affected by IRS are

endophilic species (species that tend to rest and live indoors), and due to the

irritation caused by spraying, their evolutionary descendants are trending

towards becoming exophilic (species that tend to rest and live out of doors),

meaning that they are not as affected—if affected at all—by the IRS,

rendering it somewhat useless as a defense mechanism (59).

B. Use of mosquito nets and bedclothes: This involves the use of insecticide

treated nets (ITNs) that serve as repellants, thus keeping mosquitoes away

from people, and greatly reducing the infection and transmission of malaria.

The insecticide kills the mosquito before it has time to find a way through the

net and get to the individual. Even people sleeping near but not inside the net

are a bit protected. ITNs offer 70% protection compared with no net and are

twice as effective as untreated nets (60). However, less than 2% of children in

urban areas in sub-Saharan Africa are protected by ITNs. Insecticides

impregnated into the nets include; permethrin or deltamethrin. ITNs are the

most cost-effective method of malaria prevention (50). ITNs are to be re-

impregnated with insecticides every six months. Long-lasting Insecticidal nets

(LLINs), for example Olyset TM

, have now been produced and they release

xxxii

insecticides for approximately 5 years, but they cost more (50). Awareness on

the importance and appropriate use of ITNs has to be carried out.

A study carried out among Afghan refugees in Pakistan, has shown that top-sheets

and chadders (Head coverings) treated with permethrin has similar effectiveness as

ITNs, but is much cheaper (61).

Malaria prophylaxis involves the use of drugs to prevent the development of clinical

malaria, and can be in form of either intermittent preventive treatment (in pregnancy

or childhood) or chemoprophylaxis (in advantaged non-immune individuals travelling

to endemic areas). Most drugs used here are also used in the treatment of malaria, but

are taken at a lower dose than would be used for treatment, daily or weekly. Malaria

chemoprophylaxis is restricted to short term visitors and travelers to malaria endemic

regions, because of the adverse effects of long term use and cost of purchasing the

drug (50). Drugs used here includes; quinine, chloroquine, primaquine, quinacrine,

mefloquine, doxycycline, atovaquone + proguanil (malarone®).

There is currently no available effective vaccine. Malaria vaccines are still under

development and include: pre-erythrocytic vaccines (target parasites before it reaches

the blood), vaccines based on circum-sporozoite protein ( make up the largest group

of research for malaria vaccines), vaccines that seek to avoid more severe pathologies

of malaria by preventing adherence of the parasite to blood venules and placenta,

vaccines that seek to induce immunity to the blood stage of the infection, transmission

blocking vaccines ( it blocks the development of the parasite after a mosquito has

taken a blood meal from an infected person) (62). The first vaccine developed that has

undergone field trials, is the SPf66, developed by Manuel Elkin Patarroyo in 1987. It

presents a combination of antigens from the sporozoite (using CS repeats) and

merozoite parasites. During phase I trials a 75% efficacy rate was demonstrated and

xxxiii

the vaccine appeared to be well tolerated by subjects and immunogenic. The phase IIb

and III trials were less promising, with the efficacy falling to between 38.8% and

60.2%. A trial was carried out in Tanzania in 1993 demonstrating the efficacy to be

31% after a year’s follow up, however the most recent (though controversial) study in

The Gambia did not show any effect. Despite the relatively long trial periods and the

number of studies carried out, it is still not known how the SPf66 vaccine confers

immunity; it therefore remains an unlikely solution to malaria. The CSP was the next

vaccine developed that initially appeared promising enough to undergo trials. It is also

based on the circumsporoziote protein, but additionally has the recombinant (Asn-

Ala-Pro15Asn-Val-Asp-Pro)2-Leu-Arg(R32LR) protein covalently bound to a

purified Pseudomonas aeruginosa toxin (A9). However at an early stage a complete

lack of protective immunity was demonstrated in those inoculated. The study group

used in Kenya had an 82% incidence of parasitaemia whilst the control group only

had an 89% incidence. The vaccine intended to cause an increased T-lymphocyte

response in those exposed; this was also not observed (62).

The RTS, S/AS02A vaccine is the candidate furthest along in vaccine trials. It is being

developed by a partnership between the PATH Malaria Vaccine Initiative (a grantee

of the Gates Foundation), the pharmaceutical company, GlaxoSmithKline, and the

Walter Reed Army Institute of Research (63). In the vaccine, a portion of CSP has

been fused to the immunogenic "S antigen" of the hepatitis B virus; this recombinant

protein is injected alongside the potent AS02A adjuvant (62). In October 2004, the

RTS, S/AS02A researchers announced results of a Phase II b trial, indicating the

vaccine reduced infection risk by approximately 30% and severity of infection by

over 50%. The study looked at over 2,000 Mozambican children (64). More recent

testing of the RTS, S/AS02A vaccine has focused on the safety and efficacy of

xxxiv

administering it earlier in infancy: In October 2007, the researchers announced results

of a phase I/IIb trial conducted on 214 Mozambican infants between the ages of 10

and 18 months in which the full three-dose course of the vaccine led to a 62%

reduction of infection with no serious side-effects save some pain at the point of

injection (65).

Other methods of prevention of malaria includes; mass drug administration,

intermittent preventive therapy, education of the populace on preventive methods, and

providing accessible malaria diagnostic facilities and affordable, accessible, effective

drugs. More so, screening of windows and doors with mosquito netting, and use of

mosquito repellant cream can protect against malaria infection (66, 24).

1.3 Treatment of malaria

Historically, the first effective treatment for malaria came from the bark of cinchona

tree, which contains quinine. This tree grows on the slopes of the Andes, mainly in

Peru. The indigenous peoples of Peru made a tincture of cinchona to control malaria.

The Jesuits noted the efficacy of the practice and introduced the treatment to Europe

during the 1640s, where it was rapidly accepted (67). It was not until 1820 that the

active ingredient, quinine, was extracted from the bark, isolated and named by the

French chemists Pierre Joseph Pelletier and Joseph Bienaimé Caventou (68). Due to

the toxicity of quinine, a less toxic analoque, chloroquine was developed and was

designated the drug of choice in 1946 (69). Other more effective and less toxic

antimalarials have been synthesized since then.

The therapy for malaria differs based on if the malaria is uncomplicated or severe.

Uncomplicated malaria can be treated at home while severe malaria is an emergency

and is treated in the hospital. Whether uncomplicated or not, supportive

measures/treatment must be added in addition to the specific antimalarial drug used.

xxxv

The antimalarial drug can be inform of approved herbal remedies, as is most preferred

in the rural areas of developing countries due to the high cost of orthodox antimalarial

drugs (50). The WHO guidelines for the treatment of malaria is presented in section

1.6 below and it provides a protocol on how the drugs discussed here are to be used in

the treatment of uncomplicated malaria.

1.3.1 Drugs used in the treatment of malaria

Chemotherapy is the prevention or treatment of a disease through the use of chemical

substances (70). From the chemotherapeutic standpoint, antimalarial agents are

classified into four main groups;

i. Tissue schizontocides- Antimalarial agents that are tissue schizontocides act

on the developing or dormant forms of the parasite in the hepatocytes (71),

and so will prevent relapse (fresh infection) from P. vivax and P. ovale,

Examples of tissue schizontocides are proguanil and pyrimethamine (1). They

are also used in chemoprophylaxis.

ii. Blood schizontocides- Antimalarial agents belonging to this group act on the

erythrocytic asexual forms of the parasite. They thus prevent the manifestation

of clinical disease, thus producing a cure. Majority or all of effective

antimalarial drugs kill the erythrocytic form of the parasite, and thus reduce

the number of blood forms before they grow in sufficiently large quantities to

cause clinical disease (71).

iii. Gametocytocide- Antimalarial agents in this group act on the sexual forms

(gametocytes) of the parasite in the blood, which is responsible for the

continual transmission of malaria by mosquitoes. Some gametocytocides act

on the early forms of the gametocytes, while others act on all stages of

maturation. Different gametocytocides are known to demonstrate varying

xxxvi

effects on the gametocytes of the different species of Plasmodium. Only

primaquine is known to act on the gametocytes of all species (1).

iv. Sporontocides- The antimalarial agents in this group render the gametocytes

infertile, instead of killing them, by inhibiting the sporogonic cycle in the

mosquito that has sucked the infected blood from a person treated with

sporontocides. An example of sporontocides is pyrimethamine (1).

Based on their function or the way they are used, antimalarial agents can be classified

as;

i. Antimalarial agents used for causal prophylaxis: Causal prophylaxis is the

process by which the infection is controlled before symptoms of the disease

begin to manifest. Therefore an agent used for causal prophylaxis must act on

the hepatic forms of the parasite, thus eradicating the parasite before they

reach the blood stream. P. falciparium is the most susceptible to antimalaria

prophylactic treatment. Examples of antimalarial agents used for causal

prophylaxis are proguanil and primaquine, which is more active than the

former (1).

ii. Antimalarial agents used for suppressive treatment: the drugs in this group act

on the erythrocytic forms of the parasite, thus suppressing the symptoms of

malaria. Symptoms of the malaria may return if these agents are taken for a

short period due to the hepatic stage of the parasite still in the liver, which may

emerge and enter into the blood. But if the drug is taken for a longer period a

cure may be achieved. An example of such drugs is pyrimethamine (1).

iii. Antimalarial agents used for clinical cure: drugs in this group are active

against the asexual erythrocytic stages, thus preventing the development of

schizonts. Examples of drugs here are; chloroquine, amodiaquine, quinacrine,

xxxvii

quinine, mefloquine, halofantrine, artemisinin derivatives, atovaquone,

sulfedoxine +pyrimethamine etc (1).

iv. Antimalarial agents used for radical cure: drugs in group can eradicate both

the exoerythrocytic and the erythrocytic stages of the parasite, but no single

drug can reliably produce radical cure (71), the drugs have to be used in

combination. Examples include; primaquine + chloroquine, primaquine +

quinine etc

v. Miscellaneous Antimalarial agents: this includes antibiotics that have been

shown to have antimalaria activity either in vitro or in vivo. The most widely

used antibiotics are the tetracyclines, doxycycline and minocyclin, because of

their activity on both the exoerythrocytic schizonts and the asexual forms in

the blood (1). They are not used alone in the treatment of malaria; they are

combined with drugs used for clinical cure.

Combinations potentially offer a number of important advantages over

monotherapies. First, they should provide improved efficacy. Appropriately chosen

combinations must be at least additive in potency, and may provide synergistic

activity. However, combination regimens that rely on synergy may not offer as much

protection against the selection of resistance as expected, as resistance to either

component of the combination could lead to a marked loss of efficacy. Indeed, the

widely used synergistic combination SP acts almost as a single agent in this regard,

with rapid selection of resistance (72), and similar concerns apply to the new

atovaquone/proguanil (Malarone; GlaxoSmithKline) combination (73). Other

desirable properties of antimalaria combination are that; resistance should not develop

to the two drugs at a time and the combination should reduce the selection of

antimalaria drug resistance. It was also recently shown that SP selected for resistance-

xxxviii

conferring mutations and subsequent treatment failure, but that SP combined with

artesunate prevented the selection of SP-resistant parasites in subsequent infections

(72). Combinations might offer additional advantages if the separate agents are active

against different parasite stages and if they provide the opportunity to decrease

dosages of individual agents, thereby reducing cost and/or toxicity. Ideally,

combination regimens will incorporate two agents that are both new (so that parasites

resistant to either agent are not already circulating), offer potent efficacy and

preferably have similar pharmacokinetic profiles (to limit the exposure of single

agents to resistance pressures). Unfortunately, these are challenging requirements that

are not met by any combination available at present. One current, widely advocated

strategy is to combine artemisinins — which have no resistance problem but suffer as

monotherapy from late recrudescences due to their short half-lives (71) with longer-

acting agents. The hope is that the potent action of artemisinins will prevent

significant selection of parasites resistant to the longer-acting component like

amodiaquine, mefloquine and lumefantrin etc (74).

1.3.2 Artesunate

Artesunate is a semisynthetic derivative of artemisinin (a potent antimalaria drug

isolated from Artemisia annua) that was developed so as to address the problems

encountered during the formulation of artemisinin into oral dosage forms, via

improving the solubility of the parent drug (71). Other analogues include artemether,

dihydroartemisinin etc. Artemisinin and its derivatives have superior antimalarial

efficacy (fever clearance time is shortened to 32 h as compared with 2-3 days with

older drugs (75)), are effective in the treatment of severe malaria (comparable to that

of quinine), and is effective in cases of multi-drug resistant falciparium malaria

(1,71). They are even effective against quinine resistant strains.

xxxix

Although their efficacy is limited by their short half-life, such that recrudescence rates

are unacceptably high after a short course or even after a seven day therapy, efficacy

can be maintained by combining them with other antimalarial like mefloquine,

lumefantrine, fansider etc. These combinations also curb resistance. They are also

safer than quinine and have lesser adverse effects compared to quinine (1,71). It is

soluble in water but has poor stability in aqueous solutions at neutral or acid pH. In

the injectable form, artesunic acid is drawn up in sodium bicarbonate to form sodium

artesunate immediately before injection.

Figure 2: Artesunic acid

1.3.2.1 Mechanism of action of artesunate

Artesunate is converted rapidly in the body into mainly dihydroartemisinin (DHA),

the active metabolite of artesunate, by the plasma esterases. Dihydroartemisinine

(DHA) like artesunate has an endoperoxide bridge, as shown in Figure 2. This

endoperoxide bridge is split by heme or molecular iron within the infected

xl

erythrocyte, generating singlet oxygen or free radicals and electrophilic (alkylating)

intermediates. Parasite proteins, particularly in membraneous structures, are thus

akylated, leading to parasite death. They are active against a broad spectrum of the

life cycle of the parasite, from the relatively inactive ring stage to the late schizonts in

the blood cells. The schizontocidal and gametocytocidal activities of artesunate

administered orally have been demonstrated in vivo on chloroquine sensitive stains of

P. berghei in mice and P. knowlesi in monkey) and chloroquine resistant strains of P.

berghei in mice (76).

1.3.2.2 Pharmacokinetics of artesunate

Artesunate is given orally, intraveneously, intramuscularly and rectally. It is rapidly

absorbed after oral administration and then rapidly converted by plasma esterases to

mainly DHA, with peak plasma concentration at 1.5 hrs post administration, and a

half-life of 45 min. DHA drug levels appear to decrease after a number of days of

therapy. DHA is highly protein bound. DHA is then metabolized in the liver via

glucuronidation prior to excretion (46, 71).

1.3.2.3 Pharmacological actions of artesunate

After administration artesunate acts very rapidly against blood schizontocides of all

species of human plasmodium, more rapidly than even oral chloroquine or

intravenous quinine, but has no effect on the hepatic stage. Artemisinins have

gametocytocidal effects (71). Artesunate is indeed the fastest acting artemisinin used

clinically (77).

1.3.2.4 Side effects of artesunate

Artemisinins and its derivatives are better tolerated than most antimalarias. Side

effects are few and includes; nausea, vomiting, diarrhea, rarely; transient

xli

reticulocytopenia, neurological abnormalities and some liver enzyme elevation. At

doses higher than those used to treat malaria irreversible neurotoxicity occur (76).

1.3.2.5 Artemisinin-based combination therapy

As in HIV management, the World Health Organization (WHO) guidelines now

recommend antimalarial combination therapy to forestall the development of further

resistance (46). Combination therapy of antimalarial drug refers to the simultaneous

use of two or more blood schizontocidal drugs with independent mode of action and

different biochemical targets in the parasites. The concept of combination therapy is

based on the synergistic or additive potential of two or more drugs to improve

therapeutic efficacy and also to delay the development of resistance to the individual

components of the combination (78). Combination

therapy should include a

gametocidal agent to decrease transmission

of both sensitive and drug-resistant

parasites (79). The effect of combination therapy is enhanced by the inclusion of an

artemisinin derivative, as they decrease the parasite density more rapidly than other

antimalaria drug (80). When used alone, the short half-life of the artemisinin

derivative minimizes the period of parasite exposure to subtherapeutic blood levels. In

combination with another drug with a longer half-life, the short half-life and rapid

parasite clearance time of artemisinin derivatives mean that fewer parasites are

exposed to the companion drug alone after elimination of the artemisinin component.

Furthermore, exposure occurs when blood levels of the drug close to the maximum

are still present (81).

Another benefit of artemisinin combination is the 90% reduction in gametocyte levels

in treated patients (82). These characteristics minimizes the probability that a resistant

mutant will survive therapy and may also reduce the overall malaria transmission

rates, especially via pre-existing resistant strains. Artemisinin-combination therapies

xlii

(ACTs) are WHO-recommended treatment policy for uncomplicated malaria (83) in

countries where standard antimalarials are ineffective due to drug resistance (79). The

use of this drug alone results in recrudescence because of its short half-life; therefore,

it must be used in combination. In Africa, artemeter-lumefantrine is recommended as

the standard combination therapy, but other combination therapy have been studied

and are used (84,85). Polymorphisms within the pfatp6 gene have been associated

with the resistant phenotype is some studies. It may be of concern that pfmdr1

mediates resistance to a number of unrelated classes of agents that are now being used

in combination (86). Continued monitoring of genotypic, phenotypic, and in vivo

outcomes will be necessary to monitor efficacy of these new combinations.

Current

combinations

recommended by the WHO include artemether-lumefantrine,

artesunate-amodiaquine, artesunate-mefloquine, and artesunate-SP, for second line

treatment; alternative ACT or quinine in combination with either tetracycline or

doxycycline or clindamycin (46).

1.4 Pharmacodynamic interaction

Pharmacodynamic interactions include the concurrent administration of drugs having

the same (or opposing) pharmacologic actions and alteration of the sensitivity or the

responsiveness of the tissues to one drug by another. Many of these interactions can

be predicted from knowledge of the pharmacology of each drug (87). When

discussing drug interactions, the drug affected by the interaction is called the “object

drug,” and the drug causing the interaction is called the “precipitant drug.”(88). It

involves competition at receptor sites or activity of the interacting drugs on the same

physiological system.

Types of pharmacodynamic drug interaction include;

xliii

A. Additive/ synergistic pharmacodynamic drug interaction: When two or more

drugs with similar pharmacological actions are given, the combined effect

may be additive or synergistic, i.e., an effect that is more than the singular

effect of the individual drugs. This can result in excessive response and

toxicity. Examples include combinations of alcohol and hypnosedatives,

which may result in excessive drowsiness; potassium supplements and

potassium-sparing diuretics, which will cause marked hyperkalaemia.

B. Antagonistic pharmacodynamic drug interaction: Here drugs with opposing

pharmacological actions acting on the same receptor. The response to one or

both drugs may be reduced. For example, drugs that tend to increase blood

pressure (such as nonsteroidal anti-inflammatory drugs) may inhibit the

antihypertensive effect of drugs such as ACE inhibitors.

C. Pharmacodynamic interaction due to fluid/electrolyte imbalance:

E.g. diuretics that cause hypokalaemia can increase the toxicity of digoxin.

D. Pharmacodynamic interaction due to changes in drug transport mechanisms:

chlorpromazine, when given together with guanethidin, an antihypertensive,

will inhibit the uptake of guanethidine into the adrenergic neurons, thus

inhibiting its antihypertensive effect (87).

1.5 Antimalarial drug resistance

Drug resistance is reduced susceptibility of the causal agent of a disease to a drug.

According to WHO, antimalarial drug resistance is the ability of a parasite strain to

survive and/or multiply despite the administration and absorption of a medicine given

in equal to or higher than those usually recommended but within the tolerance of the

xliv

subject, with certainty that the form of the drug active against the parasite will gain

access to the parasite or the infected red blood cell for the duration of time necessary

for its normal action in the body. Resistance arises due to selection of parasites with

genetic mutations or gene amplifications that confer reduced susceptibility (46).

Efforts to eradicate or control the disease, by eradicating the female mosquito via the

use of insecticides or by therapy with antimalarials have proved abortive, especially in

developing nations due to the emergence of insecticide resistant mosquito and drug-

resistant malaria parasites (78). The high prevalence of infection, constant drug

pressure, sexual reproduction, and large parasite biomass

contribute to the

development of a resistant parasite. These factors present a unique challenge in

optimizing drug treatment strategies. This scenario is similar to HIV, in which

continuous drug pressure and high viral burden have resulted in drug-resistant

strains.

As in HIV management, the World Health Organization (WHO) guidelines now

recommend antimalarial combination therapy to forestall the development of further

resistance (46). The United Nations International Children’s’ Education Fund

(UNICEF) notes that the greatest challenge in malaria control is that the cheapest

antimalarial drug chloroquine is rapidly losing its effectiveness, via the development

of resistance (1). Gametocytes are often resistant to standard antimalarials used to

clear the asexual-stage parasite. This may be due to the

unique biology of

gametocytes, which shuts down a subset of

metabolic pathways, rendering the

antimalarials ineffective (89). The post treatment gametocyte carriage rate varies by

antimalarial treatment, and in vitro evidence suggests that chloroquine may

actually

induce gametocytogenesis (90,91,92). The artemisinin compounds

are a notable

exception and can clear gametocyte stages.

xlv

The history of antimalarial drug resistance dates back to 1963. This resistance did not

appear when pure single compounds were used, most probably because the herbal

remedies used to treat malaria were already in form of a complex mixture, with

numerous antimalarial agents acting at the same time thus making it difficult for the

parasite to develop resistance against a single agent or even all the agents at the same

time. Indeed, combination therapy existed in herbal remedies from time immemorial.

Chloroquine was designated the drug of choice in 1946 (69). In Malaysia, chloroquine

resistant case was first reported in 1963 (93). Subsequently, several Chloroquine-

resistant cases have been reported in Sabah, west Malaysia (94). In Africa,

chloroquine resistant Plasmodium falciparum was first found in 1978 in nonimmune

travellers from Kenya and Tanzania (95, 96). This was followed 2 to 3 years later by

reports from Madagascar (97). Resistance spread from the African coastal areas inland

and by 1983 had been observed in Sudan, Uganda, Zambia and Malawi (98, 99, 100,

101). The Sulphonamides-Pyrimethamine (SP) combination was not left out

(102,103,104,105,106). Recently, there has been wide spread resistance of

falciparium malaria both to chloroquine and SP in endemic areas of Peninsular

Malaysia (107). In Thailand, SP replaced chloroquine in 1973 due to resistance, and

then mefloquine-sulphadoxine-pyrimethamine (MSP) was introduced in 1984 in the

Thai-cambodian border refugee camp and later extended to the whole of Thailand in

1985. In 1989, there was a reduction in the sensitivity (95% to 50%) to mefloquine

(MQ) at 15mg/kg, in specific areas of Thailand (108). MQ at 25mg/kg was even still

unable to prolong higher efficacy substantially, so that the use of MQ plus 3 days of

artesunate (an ACT) was adopted in such areas from 1994-1996, and this restored the

cure rate to 90-95% (109). In, Africa, efficacy trials have also shown the superiority of

artemisinin combination therapy when compared with monotherapy in areas of drug-

xlvi

resistant malaria

(110,111). Moreover, the therapeutic failure of artesunate

monotherapy in non-immune individuals from Central Africa were associated with

reduced in vitro susceptibility (112,113), but, there is no confirmed in vivo evidence

of resistance of P. falciparum to artemisinin and its derivatives.

Methods for assessing and thereby classifying antimalarial drug resistance include;

A. In vivo Testing: This involves the assessment of clinical and parasitological

outcomes of treatment over a certain period (≥ 28 days) following the start of

treatment, to check for the reappearance of parasites in the blood.

Reappearance indicates reduced parasite sensitivity to the treatment drug. The

parasitological cure rates should also be assessed. Blood or plasma of the

antimalaria drug has to be assessed also so as to distinguish treatment failures

due to pharmacokinetic reasons (46).

B. Molecular genotyping: this involves the use of molecular markers for drug

resistant malaria. These markers are then detected by using PCR, where small

amounts of parasite DNA material in finger-prick blood dried on filter paper

are used. These markers are based on genetic changes that confer parasite

resistance to drugs used to treat and prevent malaria. Polymorphisms in the

Plasmodium falciparum chloroquine resistance transporter (PfCRT) confer

resistance to chloroquine (114,115), and mutations in the P-glycoprotein

homologue (Pgh1) encoded by pfmdr1 modulate this resistance (116).

Polymorphisms in pfmdr1 and amplifications of this gene also affect

susceptibility to structurally unrelated antimalarial drugs, including

mefloquine, artesunate, lumefantrine and quinine (117,118,119).

Polymorphisms in P. falciparum dihydrofolate reductase (DHFR) cause

xlvii

resistance to the antifolate drugs including pyrimethamine and other DHFR

inhibitors, and polymorphisms in dihydropteroate synthase (DHPS) cause

resistance to sulphadoxine and other sulphas and sulphones (120,121). For

ACT, molecular markers are to be used to monitor its partner drug. Each ACT

partner drug is likely to select for resistance, potentially leading to loss of

treatment efficacy as well as failure to protect the artemisinins against the

development of resistance. At present the most important partner drugs used

with artemisinins in ACTs are amodiaquine, lumefantrine, and piperaquine.

The molecular mediators of resistance are not as well defined for these drugs

as they are for chloroquine and SP, but recent data show hints of mechanisms

of resistance. For amodiaquine, polymorphisms in both PfCRT and Pgh1

appear to predict resistance and to be selected for by treatment with

amodiaquine (122) or artesunate-amodiaquine (123). Artemether-lumefantrine

treatment selects for polymorphisms in pfmdr1 associated with diminished

sensitivity to the related drug halofantrine (124,125). Markers for piperaquine

resistance have not been identified, but this aminoquinoline may well act

similarly to chloroquine and amodiaquine in its selection of resistance-

mediating mutations. To avoid unacceptably long delays in identifying,

validating and deploying molecular markers of ACT resistance, and the

malaria research and control community must be prepared to investigate

aggressively early reports of resistance, confirm resistance with careful in

vitro assays, and bring genetic and genomic tools to bear to elucidate

mechanisms and identify candidate molecular markers. The sequencing of the

P. falciparum genome has led to genome-wide approaches that may help to

xlviii

identify genetic markers of drug resistance far more quickly than was

previously possible (126,127,128).

There is a network database being created, that will be used to monitor and

deter resistance, and to guide malaria treatment and prevention policies called

World Antimalaria Resistance Network (WARN), which uses a global

database for molecular markers of drug resistant malaria and links it to

databases for malaria drug efficacy trials, in vitro drug resistance, and

pharmacokinetics (129). Molecular genotyping using PCR technology should

be used to distinguish recrudescent parasites from newly acquired infections.

C. In vitro testing: It involves the collection of parasitized blood from patients

and the testing of parasite susceptibility to drugs in culture. Here the most

commonly used methods are the in vitro micro-test Mark III the isotopic test

and drug sensitivity assay based on the measurement of HRP2/or pLDH/ in an

enzyme-linked immunosorbent assay (ELISA). To support evidence of a

failing antimalarial, in vitro tests can be used to provide a more accurate

measure of drug sensitivity under controlled experimental conditions.

Parasites obtained from finger-prick blood are placed in microtitre wells,

exposed to precisely known concentrations of a particular drug and examined

for the inhibition of maturation into schizont parasite stages (130). This test

overcomes some of the many confounding factors influencing the results of in

vivo tests, such as subtherapeutic drug concentrations and the influence of host

factors on parasite growth (e.g. factors related to acquired immunity), and

therefore provide a more accurate picture of the “true” level of resistance to

the drug. Multiple tests can be performed on parasite isolates, using several

xlix

drugs and drug combinations. But only P. falciparium and P. vivax can be

tested using this method. In vitro testing is more demanding in terms of

technology and resources, and is not ideal for routine drug efficacy evaluation

under field conditions. It should therefore primarily be used to provide

additional information to support clinical efficacy data at selected resistance-

monitoring sites.

1.6 WHO Guildlines for treatment of uncomplicated malaria

For several decades, the gold standard for the treatment of malaria was chloroquine, a

4-aminoquinoline (it was efficacious, had low toxicity and was affordable (74). It now

recommended that the first line treatment for malaria in any region be changed if the

total failure proportion exceeds 10%. However, it is acknowledged that a decision to

change may be influenced by a number of additional factors, including the prevalence

and geographical distribution of reported treatment failures, health service provider

and/or patient dissatisfaction with the treatment, the political and economical context,

and the availability of affordable alternatives to the commonly used treatment. To

overcome the threat of resistance of P. falciparum to monotherapies, and to improve

treatment outcome, combinations of antimalarials are now recommended for the

treatment of uncomplicated falciparum malaria. Drug combinations such as

sulfadoxine–pyrimethamine, sulfalene–pyrimethamine, proguanil-dapsone,

chlorproguanil-dapsone and atovaquone-proguanil rely on synergy between the two

components. The drug targets in the malaria parasite are linked. These combinations

are operationally considered as single products and treatment with them is not

considered to be antimalarial combination therapy. Multiple-drug therapies that

include a non-antimalarial medicine to enhance the antimalarial effect of a blood

l

schizontocidal drug (e.g. chloroquine and chlorpheniramine) are also not antimalarial

combination therapy.

The rationale for combining antimalarials with different modes of action is twofold:

(1) the combination is often more effective; and (2) in the rare event that a mutant

parasite that is resistant to one of the drugs arises de novo during the course of the

infection, the parasite will be killed by the other drug. This mutual protection is

thought to prevent or delay the emergence of resistance.

To realize the two advantages, the partner drugs in a combination must be

independently effective. The possible disadvantages of combination treatments are the

potential for increased risk of adverse effects and the increased cost.

Artemisinin and its derivatives (artesunate, artemether, artemotil, dihydroartemisinin)

produce rapid clearance of parasitaemia and rapid resolution of symptoms. They

reduce parasite numbers by a factor of approximately 10000 in each asexual cycle,

which is more than other current antimalarials (which reduce parasite numbers 100- to

1000-fold per cycle). Artemisinin and its derivatives are eliminated rapidly. When

given in combination with rapidly eliminated compounds (tetracyclines, clindamycin),

a 7-day course of treatment with an artemisinin compound is required; but when given

in combination with slowly eliminated antimalarials, shorter courses of treatment (3

days) are effective. In 3-day ACT regimens, the artemisinin component is present in

the body during only two asexual parasite life-cycles (each lasting 2 days, except for

P. malariae infections). This exposure to 3 days of artemisinin treatment reduces the

number of parasites in the body by a factor of approximately one hundred million (104

× 104 =10

8). However, complete clearance of parasites is dependent on the partner

medicine being effective and persisting at parasiticidal concentrations until all the

infecting parasites have been killed. Thus the partner compounds need to be relatively

li

slowly eliminated. As a result of this the artemisinin component is “protected” from

resistance by the partner medicine provided it is efficacious and the partner medicine

is partly protected by the artemisinin derivative. Courses of ACTs of 1–2 days are not

recommended; they are less efficacious, and provide less protection of the slowly

eliminated partner antimalarial. The artemisinin compounds are active against all four

species of malaria parasites that infect humans and are generally well tolerated. The

only significant adverse effect to emerge from extensive clinical trials has been rare

(with an occurrence of 1:3000) and is type 1 hypersensitivity reactions (manifested

initially by urticaria). The artemisinins also have the advantage of reducing

gametocyte carriage and thus the transmissibility of malaria. This contributes to

malaria control in areas of low endemicity.

Non-artemisinin based combinations (non-ACTs) include sulfadoxine–pyrimethamine

with chloroquine (SP+CQ) or amodiaquine (SP+AQ). However, the prevailing high

levels of resistance have compromised the efficacy of these combinations. There is no

convincing evidence that SP+CQ provides any additional benefit over SP, so this

combination is not recommended; SP+AQ can be more effective than either drug

alone, but needs to be considered in the light of comparison with ACTs.

The following ACTs are currently recommended (alphabetical order);

artemether-lumefantrine,

artesunate + amodiaquine,

artesunate + mefloquine,

artesunate + sulfadoxine–pyrimethamine.

Amodiaquine plus sulfadoxine–pyrimethamine may be considered as an interim

option where ACTs cannot be made available, provided that efficacy of both is high.

lii

The following drugs have not yet been recommended as an antimalarial-combination

therapy;

Chlorproguanil-dapsone has not yet been evaluated as an ACT partner drug, so

there is insufficient evidence of both efficacy and safety to recommend it as a

combination partner.

Atovaquone-proguanil has been shown to be safe and effective as a

combination partner in one large study, but is not included in these

recommendations for deployment in endemic areas because of its very high

cost.

Halofantrine has not yet been evaluated as an ACT partner medicine and is not

included in these recommendations because of safety concerns.

Dihydroartemisinin (artenimol)-piperaquine has been shown to be safe and

effective in large trials in Asia, but is not included in these recommendations

as it is not yet available as a formulation manufactured under good

manufacturing practices, and has not yet been evaluated sufficiently in Africa

and South America.

Treatment failure within 14 days of receiving an ACT is very unusual. Treatment

failures within 14 days should be treated with a second-line antimalarial;

alternative ACT known to be effective in the region,

artesunate + tetracycline or doxycycline or clindamycin,

quinine + tetracycline or doxycycline or clindamycin.

Recurrence of fever and parasitaemia more than 2 weeks after treatment, which could

result either from recrudescence or new infection, can be retreated with the first-line

ACT. Parasitological confirmation is desirable but not a precondition. If it is a

recrudescence, then the first-line treatment should still be effective in most cases. This

liii

simplifies operational management and drug deployment. However, reuse of

mefloquine within 28 days of first treatment is associated with an increased risk of

neuropsychiatric sequelae and, in this particular case; second-line treatment should be

given. If there is a further recurrence, then malaria should be confirmed

parasitologically and second-line treatment given.

In pregnancy, the antimalarials considered safe in the first trimester of pregnancy are

quinine, chloroquine, proguanil, pyrimethamine and sulfadoxine–pyrimethamine. Of

these, quinine remains the most effective and can be used in all trimesters of

pregnancy. In reality women often do not declare their pregnancies in the first

trimester and so, early pregnancies will often be exposed inadvertently to the

available first-line treatment. Inadvertent exposure to antimalarials is not an indication

for termination of the pregnancy. There is increasing experience with artemisinin

derivatives in the second and third trimesters (over 1000 documented pregnancies).

There have been no adverse effects on the mother or fetus. The current assessment of

benefits compared with potential risks suggests that the artemisinin derivatives should

be used to treat uncomplicated falciparum malaria in the second and third trimesters

of pregnancy, but should not be used in the first trimester until more information

becomes available. The choice of combination partner is difficult. Mefloquine has

been associated with an increased risk of stillbirth in large observational studies in

Thailand, but not in Malawi. Amodiaquine, chlorproguanil-dapsone, halofantrine,

lumefantrine and piperaquine have not been evaluated sufficiently to permit positive

recommendations. Sulfadoxine–pyrimethamine is safe but may be ineffective in many

areas because of increasing resistance. Clindamycin is also safe, but both medicines

(clindamycin and the artemisinin partner) must be given for 7 days. Primaquine and

liv

tetracyclines should not be used in pregnancy. Dapsone and tetracycline are not to be

used by lactating mothers.

P. malariae and P. ovale are still very sensitive to chloroquine, and are also sensitive

to amodiaquine, mefloquine and the artemisinin derivatives. P. vivax is still very

sensitive to chloroquine but resistance is prevalent and increasing in some areas,

notably Oceania, Indonesia and Peru. It is also sensitive to all other antimalarias

except SP, proguanil and chlorproguanil. In contrast to P. falciparum, asexual stages

of P.vivax are susceptible to primaquine. Thus the combination of chloroquine and

primaquine can be considered a combination treatment. The only drugs with

significant activity against the hypnozoites are the 8-aminoquinolines (bulaquine,

primaquine, tafenoquine). The recommended treatment for the relapsing malaria

caused by P. ovale is the same as that given to achieve radical cure in vivax malaria,

i.e. with chloroquine and primaquine. Mixed infections are common and ACTs are

effective against all malaria species, and so is the treatment of choice. Radical

treatment with primaquine should be given to patients with confirmed P. vivax and P.

ovale infections except in high transmission settings where the risk of re-infection is

high (46).

1.7 Plants with antimalarial activity

Plants remain an important source of medicines for both traditional and orthodox

health care practices. For example, artemisinin, which was isolated from Artemisia

annua L. (Qinghaosu), the sweet wormwood, and is a sesquiterpene lactone

endoperoxide (1). Traditionally, plants are used in form of herbal drugs or

phytomedicines. Over the past century, chemical and pharmacologic science

established the compositions, biological activities and health giving benefits of

numerous plant extracts. But often when individual components were separated from

lv

the whole there was loss of activity—the natural ingredient synergy became lost.

Standardization was developed to solve this problem. But even with standardization,

poor bioavailability often limited their clinical utility (131). In 2001, researchers

identified 122 compounds used in mainstream medicine which were derived from

ethnomedical plant sources; 80% of these compounds were used in the same or

related manner as the traditional ethnomedical use. Major pharmaceutical companies

are currently conducting extensive research on plant materials gathered from the rain

forests and other places for possible new pharmaceuticals (132). Three quarters of

plants that provide active ingredients for prescription drugs came to the attention of

researchers because of their use in traditional medicine. Among the 120 active

compounds currently isolated from the higher plants and widely used in modern

medicine today, 75 percent show a positive correlation between their modern

therapeutic use and the traditional use of the plants from which they are derived (132).

More than two thirds of the world's plant species - at least 35,000 of which are

estimated to have medicinal value - come from the developing countries. At least

7,000 medical compounds in the modern pharmacopoeia are derived from plants

(133).

In Indonesia’s malaria endemic regions, medicinal plants such as Carica papaya

leaves, Eurycoma longifolia, Alstonia scolaris, Phyllanthus niuriri and Azadirachta

indica are often used to treat malaria. However, scientific information on the

antimalarial activity of these plants is very limited. Artemisia annua and Azadirachta

indica are considered as reference medicinal plants by numerious author due to their

wide use traditionally, in the treatment of malaria (134). Key medicinal plants used by

ethnomedicinal practitioners in the treatment of malaria in Nigeria are; Venonia

amygdalina, Ageratum conyzoides and Azadirachta indica (135). Most commonly

lvi

used plant for the treatment of malaria in Southwest, South south and Middle Belt are;

A. indica, Cymbopogon citrates and Carica papaya (136). The following herbs have

also shown some antimalarial activity in vitro or in animals: Artemisia vulgaris,

Cochlospermum planchonii, Cochlospermum tinctorium, Jatropha curcas, Gossypium

hirsutum, Physalis angulata, Delonix regia, Khaya grandifolia, Cryptolepsis

sanguinolenta, Tabebuia impetiginosa, Carica papaya, Swertia chirayita,

Azadirachta indica, Cajanus cajan, Euphorbia lateriflora, Mangifera indicia, Senna

alata, Cymbopogon giganteus, Nauclea latifolia, Newbouldia leavis, Adansonia

digitata, Cassia occidentalis, Tamarindus indica, Tridax procumbens, Vernonia

amygdalina, Psidium guajava, Morinda lucida and Uvaria chamae etc.

Phytomedicines initially made it possible to reliably treat malaria (via the use of

quinine from Cinchona spp.) and continue to provide exciting new antimalarial drugs

(the discovery of the artemisinins from A. annua).

1.7.1 Antimalaria activity of Carica papaya

The male and female parts of Carica papaya Linn. (Caricaceae; Common Name:

pawpaw) exist in different trees. The fruits, leaves, and latex are used medicinally.

The taxonomy of Carica papaya is shown below;

Kingdom: Plantae

Division: Magnoliophyta

Class: Magnoliopsida

Order: Violales

Family: Caricaceae

Genus: Carica L.

Species: Carica papaya L.

Its mature leaves are widely used to treat malaria and splenomegaly while the fruit is

used against anaemia, which can also be caused by malaria (137). The anti-plasmodial

activity of Carica papaya is weak as it has an IC50 of 60 mg/mL (138). However, a

lvii

recent study has shown it to reduce parasitemia at an activity second to that of SP.

Aqueous extract of its leaves have been shown to reduce parasite count from 9.20 ±

0.06 to 2.60 ± 0.06 % in P. berghei infected mice (139). In Ghana, pawpaw leaves,

neem leaves/bitter leaves, boiled together in water and cooled is taken at a dosage of

30 ml taken 3 x daily x 7 days. In Nigeria, a weak decoction of the leaves is taken for

the treatment of malaria (140).

Its antimalarial activity has been attributed to its ability to increase total antioxidant

status in patients and thus, inhibiting the development of anemia in malaria (141).

1.7.2 Antimalaria activity of Azadirachta indica

Azadirachta indica juss. (Common Name: neem) is an evergreen tree, but in severe

drought it may shed most or nearly all of its leaves. Its (white and fragrant) flowers

are arranged axillary, normally in more-or-less drooping panicles. It also bears fruits.

The neem tree is noted for its drought resistance. Neem can grow in many different

types of soil, but it thrives best on well drained deep and sandy soils. It is a typical

tropical to subtropical tree and exists at annual mean temperatures between 21-32 °C.

It can tolerate high to very high temperatures and does not tolerate temperature below

4 °C (142).

The taxonomy of Azadirachta indica is shown below;

Kingdom: Plantae

Division: Magnoliophyta

Order: Rutales

Family: Meliaceae (mahogany family)

Genus: Azadirachta

Species: A. indica

lviii

Numerous pharmacological activities have been ascribed to the various part of the

large evergreen tree, Azadiracta indica. Its leaf extract has been prescribed for oral

use for the treatment of malaria by Indian ayurvedic practitioners from time

immemorial (143). Dried neem leaves in the form of tea are used by the people of

Nigeria and Haiti to treat this disease (144). It is a Nigerian naturalized medicinal

plant known locally as ‘Dogoyaro’, which has been found to have antimalarial

properties in vivo and in vitro (145,146). The antimalarial activity of its leaf extract

has not been found to be great (147). The mechanism of action of neem is believed to

be probably due to redox perturbation in the form of the imposition of substantial

oxidant stress during therapy. The aqueous leaf extract inhibits NADPH-cytochrome c

(P-450) reductase activity in rats with significant increase in microsomal protein

(140). Neem extract have a schizontocidal and gamatocytocidal effect (148,149).

Neem seed and leaf extract are effective against malaria parasites (150,151). The leaf

extract contains; sterols, limonoids, flavonoids and their glycosides and coumarins. Of

interest are gedunin (150, 152, 153), nimbolide (153,154), nimbinin (153), 11-β-

acetoxy gedunin (153), dihydrogedunin (153), meldenin (154), isomeldenin (154),

nimocinol (154), nimbandiol (154), maldenin, azadirachtin and quercetin; which are

responsible for this property. Meldenin was found to be the most active of four

limonoids isolated from its leaves, against the chloroquine- resistant K1 strain of P.

falciparum(155). Azadiractin and three of its semisynthetic derivatives have been

found to inhibit the formation of motile male gametes in vitro (148, 155). This has

raised the possibility of developing an azadiractin-based compound as antimalarial

with transmission-blocking potential (156).

Both alcohol and water extracts of neem leaf have been confirmed as effective. It

blocks the development of the gamete in an infected person. It greatly increases the

lix

state of oxidation in red blood cells, which prevents the normal development of the

parasite. Some studies show that even chloroquine-resistant strains of malaria are

sensitive to neem, particularly a component called irodin A. 100 % of the malaria

gamete are dead within 72 hrs with a 1:20,000 ration of active ingredients (157).

Gedunine (a limonoid) and quercetin (a flavonoid) compounds found in the leaves are

also effective against malaria. In vitro, gedunin possesses activity about three times

higher than chloroquine, but twenty-times lower than quinine (153). Gedunin has also

been shown to exhibit an additive effect when combined with chloroquine (158,159).

However, despite the promising in vitro activity of gedunin, it has not been found to

inhibit Plasmodium berghei in mice (153). This is probably due to poor

bioavailability, and loss of natural synergy or ability of other constituent to protect it

from digestion and/or enhance its solubility.

Leaves of neem, especially its gedunin content have been shown to be active against

chloroquine resistant strains in vitro and this activity can be used to standardize A.

indica when it is to be used as antimalarial (160). The aqueous extract of neem leaves

has an IC50 of 2µg/ml, with complete inhibition of the schizonts maturation exhibited

at 7.8 µg/ml (161), and shows significant activity at 125-500 mg/kg against P. berghei

(162), and 41.2% parasite suppression was found at this dose range (163). Some

scientists believe that stimulation of the immune system is a major factor in neem’s

effectiveness against malaria. The plant also lowers fever and increase appetite,

enabling a stronger body to fight the parasite and recover more quickly.

The safety evaluation of neem leaf extract reveals that acute and sub-acute effect of

the aqueous leaf extracts are mostly beneficial (164). Intravenously administered

aqueous leaf extract at a dose greater than 40 mg/kg body weight produces toxic

manifestation leading to death in guinea pigs (165). Successive doses of 5–200 mg/kg

lx

reduce heart rate and increased the arterial pulse rate in guinea pigs (166). Aqueous

leaf extract also shows antifertility effect in mice when given through the oral route

(165,167). Crude neem leaf extracts causes genotoxicity in male mice germ cell at a

dose of 0.5–2 g/kg body weight for 6 weeks. Some structural change in meiotic

chromosomes along with chromosome strand breakage or spindle disturbances and

abnormal regulation of genes controlling sperm shape were observed (168). Neem

leaf extract when administered for 48 days in albino rats causes decrease in sperm

count, sperm motility and forward velocity, probably due to androgen deficiency

(169). Oral administration of 20–60 mg dry leaf powder for 24 days in rats causes

decrease in the weight of seminal vesicle and ventral prostrate and regressive changes

of the histological parameters through its antiandrogenic property (170).

1.8 Methods employed in the evaluation of antimalaria activity of a substance

The antimalarial activity of a substance can be evaluated via the use of the following

models (171);

1.8.1 In vitro methods for screening antimalarial compounds

Different in vitro methods have been developed, viz;

I. 3H-Hypoxanthine uptake method

II. Giemsa stained slide method (MIC method)

III. Use of flow cytometry

IV. Measurement of LDH activity of Plasmodium falciparum

V. Isobologram analysis for combination therapy

1.8.2 In vivo methods for screening antimalarial compounds

Plasmodium species that cause human disease are essentially unable to infect non

primate animal models. So, in vivo evaluation of antimalarial compounds begins with

lxi

the use of rodent malaria parasite. Plasmodium berghei, P. yoelii, P. chabaudi, P.

vinckei have been used extensively in drug discovery and early development. Choice

of rodent malaria species and mouse strains need to be considered during

experimental design and interpretation. P. chabaudi and P. vinckei generate a high

parasitemia and produce synchronous infections (propogation of specific stage),

enabling studies on parasite stage specificity. P. chabaudi and P. vinckei are more

sensitive than P. berghei to iron chelators and lipid biosynthesis inhibitors. In vivo

models include;

I. Rodent models: Here activity is represented as EC (effective concentration).

Such models include;

a. Plasmodium berghei 4 day suppression test

b. Hill’s test for causal prophylaxis and residual activity

c. Sporontocidal activity testing

d. Rane or curative test

e. Use of immunocompromised mice and P. falciparum: Immunocompromised

mice can support Plasmodium falciparum infection as it lacks T and LAK (

Lymphokine activated killer cell ) cells.

II. Avian models, which is no longer popular due to the introduction of rodent

models

III. Primate models: e.g Plasmodium cynomolgi rhesus model

1.8.2.1 Plasmodium berghei 4 day suppression test

This is a preliminary test method when evaluating the antimalaria activity of a

compound. Here, the efficacy of a compound is assessed by comparison of blood

lxii

parasitemia and mouse survival time in treated and untreated mice groups. Naval

Medical Research Institute (NMRI) mice free from Eperythrozoon coccoides and

Haemobartonella muris are the standard of mice to be used and they should be

maintained at 22 ˚C at 50-70 % humidity, fed with diet containing p-aminobenzoic

acid 45 mg/kg and water ad libitum. Mice contaminated with Eperythrozoon

coccoides survive infection with P. berghei longer than clean mice whereas the

presence of Haemobartonella muris tends to accelerate the malaria infection. On day

0, mice are injected with 0.2 ml of aliquot (2X108 parasitized erythrocytes)

intravenously or intraperitoneally. The animals are then grouped into groups of five

mice each. Vehicle treated mice (control group) is compared with the test drug treated

group(s). A positive control group given chloroquine or any other reference drug is

included in the study. The drugs are prepared at required concentration, as a solution

or suspension containing 7 % Tween 80/3 % ethanol and administered 2-4 hr post

infection by appropriate routes. On day 1 to 3, the experimental groups are treated

again (with the same dose and same route) as on day 0. On day 4, 24 h after the last

dose (i.e. 96 h post-infection); thin blood smears from all animals are prepared with

Giemsa stain. Parasitemia is determined microscopically by counting 4 fields of

approximately 100 erythrocytes per field. For low parasitemias (< 1%), up to 4000

erythrocytes have to be counted. The difference between the mean value of the control

group (taken as 100%) and those of the experimental groups is calculated and

expressed as percent reduction or activity using the following equation:

……………………Eqn. 1

For slow acting drugs, additional smears should be taken on days 5 and 6, to

determine parasitemia from which the activity is calculated accordingly. Untreated

lxiii

control mice typically die approximately one week after infection. For treated mice

the survival-time (in days) is recorded and the mean survival time is calculated in

comparison with the untreated and standard drug treated groups. Mice without

parasitemia on day 30 of post-infection are considered cured (171).

1.9 Objective of the study

New antimalarial drugs must meet the requirements of rapid efficacy, minimal

toxicity, and low cost. Immediate prospects for drugs to replace chloroquine and SP

include amodiaquine and chlorproguanil-dapsone (another antimalarial acting like

SP). But they already suffer some cross resistance with chloroquine and SP. The

artemisinins are next in line but they have very short half-life necessitating their use in

combination with a long acting drug. This long acting drug should probably be one to

which no known resistance has been developed yet. Moreover, antimalarial

combinations apart from improving efficacy should also forestall loss of antimalarial

activity in the face of resistance to one of the agents in the combination and reduce the

selection of antimalarial drug resistance. Such combinations may also provide for the

reduction of dosages, cost and toxicity. Additional advantages of a combination may

also arise if the individual drugs are active against different stages of the parasite.

Neem is known to be active against asexual forms in the blood and the development

of gametocyte and is also gametocytocidal. Artesunate has gametocytocidal effects.

Thus the combination of two gamecytocidal agent, ie artesunate and neem will further

decrease transmission of malaria plasmodia, even of resistant plasmodia, in high

transmission areas like Nigeria. Pawpaw leaf extract also have antimalarial activity

together with the special effect of preventing the development of anemia. Therefore

its combination with artesunate will improve treatment outcomes and enable patients

to recover quickly.

lxiv

This study therefore aims to;

Determine the pharmacodynamic interaction between the combinations of

artesunate and neem or Carica papaya aqueous crude leaf extract, based

on their antimalarial activity.

Provide the basis for the development of an artemisinin-based combination

therapy with crude drugs, so as to provide cost effective ACTs, and thus

bring orthodox and herbal medicines closer together.

Provide the basis for the possible isolation of a particular antimalarial

constituent of the neem or Carica papaya leaf extract, which can be

combined with artesunate, thus making a new and effective orthodox

combination therapy.

CHAPTER TWO

MATERIALS AND METHODS

2.1 MATERIALS

2.1.1 Experimental animals

The mice used in this study were 8-14 weeks old, non-pregnant Swiss albino females

of 25 ± 2 g, obtained from the Faculty of Veterinary Medicine and Faculty of

lxv

Pharmaceutical Sciences, UNN. The mice were left to acclimatize in the experimental

animal house unit of the Department of Biochemistry, UNN, for 5 days, during the

time of which their fed was supplemented with 45 mg/kg p-aminobenzoic acid and

water ad libitum. They were kept in a cool room. The animals were handled according

to the guidelines for laboratory animal use of the University of Nigeria, Nsukka.

2.1.2 The parasite

The parasite used in this study is chloroquine-sensitive Plasmodium berghei, NK 65

strain, obtained from the National Institute of Pharmaceutical Research and

Development (NIPRD), Abuja, and constitute the blood of mice already infected with

P. berghei, serving as the donor mice for further use in this experiment.

2.1.3 Plant extracts

The plants in this study were Azadirachta indica and Carica papaya, which were

identified by Mr. Ozioko of Bioresources Development Centre, BDC, and a voucher

specimen with No. Intercedd (International Centre for ethnomedicine and drug

development) 917 and Intercedd 918 respectively was deposited there. The plant

extract used were aqueous crude extract of the fresh leaves of Azadirachta indica and

mature fresh leaves of Carica papaya.

2.1.4 Chemicals

The pure artesunate powder used in this study was a generous gift from Emzor

Pharmaceutical Industries, Lagos. Other chemicals include; Giemsa stain (Kiran Light

Lab., Mumbai, India), Hayem’s solution, sodium chloride (M&B, England), sodium

dihydrogen phosphate (Kermel, India), Tween 80 (BDH, England), and absolute

methanol and ethanol (Sigma-Aldrich, USA).

lxvi

Other materials include; mice cages, 1 ml syringes, heparinized capillary tubes, glass

slides, dissecting set, light microscope, universal indicator paper, distilled water and

improved Neubauer Counting Chamber.

2.2 METHODS

2.2.1 Chemical preparation

2.2.1.1 Extraction of Crude drugs

Fresh leaves of Azadirachta indica (60 g) and Carica papaya (80 g), were separately

cleaned by rinsing in clean water twice, and then were homogenized with 200 ml of

sterile cold distilled water for 24 h. They were then sieved with muslin cloth and the

filtrate stored in the refrigerator. The concentration of the filtrate was then determined

by evaporating a given volume of the supernatant of the filtrate to dryness, and the

concentration in weight/ml was determined. This extraction was done 48 h prior to the

day 0 of the in-vivo schizontocidal activity testing. The extracts were discarded after

storage in the fridge for 7 days, and fresh ones were made as and when needed. The

percentage yield of each crude drug was calculated from;

Yield (%) =

The stock extract of neem was diluted with a mixture of Tween 80 and ethanol in

sterile distilled water, so as to enable the administration of doses of 100, 500 and 1000

mg/kg of the extract, and the concentration of Tween 80 and ethanol was not beyond

7% and 3% respectively. The stock extract of pawpaw was diluted with the same

solvent as for neem, to give doses of 50, 100 and 200 mg/kg. The control and test

groups were given equal volumes of the doses above. The selection of these doses

was based on the effect of neem or pawpaw leaf extract on Plasmodium in infected

mice (162, 139).

lxvii

Carica papaya

lxviii

Azadirachta indica

Figure 3: Pictures of Carica papaya and Azadirachta indica

2.2.1.2 Preparation of artesunic acid

The artesunic acid powder was dissolved in 7 % Tween 80 and 3 % ethanol in cold

sterile distilled water to give concentrations of 6, 15 and 20 mg/kg. The doses used

were based on the ED50 of artesunate on P. berghei infected mice (172).

2.2.2 In vivo schizontocidal activity of combination of a fixed dose of artesunic

acid with varying doses of crude extracts

The effect of combination therapy on early infection was checked. This was carried

out according to standard protocol following the classical Peter’s 4-day suppressive

test (171).

On day 0 of this test, the percentage parasitemia and red blood cell count of the donor

mice were determined by using a Giemsa-stained thin blood smear of the donor mice

and improved Neubauer Counting Chamber, respectively. The blood of the donor

mice was collected by cardiac puncture and from the retro-orbital plexus vein, and

diluted with physiological saline (normal saline) to give a concentration of 108

parasitized erythrocytes per ml. Parasitized erythrocytes (2 x 107, i.e., 0.2 ml of 10

8

parasitized erythrocytes/ml) was injected intraperitoneally into each of the

experimental mice. The mice was then shared randomly into 10 groups of five mice; a

negative control group (given 7 % Tween 80 and 3 % ethanol in sterile distilled

water), and the test group given the different doses of artesunate and the two crude

extracts. The drug, extracts and placebo were administered orally at 4, 24, 48 and 72 h

post infection. Day 4 (24 h) and day 7 (96 h) after the last treatment, thin blood

smears from the tail vein of all animals were fixed with methanol and stained with 10

% Giemsa solution for 10 min.

lxix

Percentage parasitemia was determined microscopically by counting 4 fields of

approximately 100 erythrocytes per field. The antimalarial activity was calculated by

converting the fractional reduction in parasitemia to percentage as shown in the

equation:

……………Eqn. 1

The mice were left till day 30 post infection during the time of which the mean

survival time of each group was noted, calculated and compared. The mouse that was

still alive on day 30 was checked for parasitemia and those with none were considered

cured. The dose of artesunate which gave a minimally significant reduction in

parasitemia, i.e. 15 mg/kg, was chosen as the fixed dose of artesunate to be combined

with different doses of the crude extract in the next phase of this test.

In the next phase, as explained previously when the test was carried out using drugs

and extracts singly, all groups of animals were given equal volumes of the

combinations of drug and extracts, and placebo. Similarly 24 h and 96 h after

administering the last treatment, blood smears were taken and the percentage

parasitemia determined and used to calculate the antimalarial activity by using the

equation above.

The mice were also left till day 30 post infection during the time of which the mean

survival time of each group was noted, calculated and compared. Each mouse still

alive on day 30 was checked for parasitemia and those with none were considered

cured.

2.2.3 Determination of ED50 of the crude extracts

The dose-response relationship of the crude drugs and artesunic acid was obtained by

plotting the percentage reduction in parasitemia, i.e., antimalarial activity of the drugs

lxx

against the logarithm of their respective doses. The linear equation of the graph was

then used to calculate the ED50 of the drugs in P. berghei infected mice.

2.2.4 Determination of the kind of pharmacodynamic interaction between the

pure drug and plant extracts

Here a dose-response relationship of the crude drugs in combination with artesunate

was obtained by plotting the percentage reduction in parasitemia, i.e., antimalarial

activity of the combination therapy against the logarithm of their respective doses.

The linear equation of the graph was then used to calculate the ED90 of the crude

drugs in combination with artesunic acid in P. berghei infected mice. The ED90 of the

crude drugs alone were also calculated from the equation of their dose-response

relationship already drawn in the previous section. The ED90 was then used to

calculate the isobolar equivalent (IE) of the crude drugs by using the equation (172);

………………………………………….Eqn. 2

However, the IE was not used to draw an isobologram as the Checkerboard technique

was not what was used here.

2.2.5 Data analysis

The significance of treatment effect was evaluated by Games-Howell’s Post Hoc

Multiple Comparism Test instead of Tukey HSD or LSD or Scheffe because the

Levene’s Test for homogeneity of variance showed that the variance of the groups is

significantly different, at p< 0.05.

The significance of survival time was evaluated by Tukey HSD as the Levene’s Test

for homogeneity of variance showed that the variance of the groups are homogeneous

at p<0.05.

CHAPTER THREE

RESULTS AND DISCUSSION

lxxi

3.1 Chemical preparation

3.1.1 Percentage yield of crude drugs

The concentration of the extracts varied; 20, 25 and 30 mg/ml for neem and 10, 20

and 35 mg/ml for pawpaw. The percentage yield ranged from 6.67 to 10 % for neem,

and 2.5 to 8.75 % for pawpaw. Therefore, on the average the percentage yield of

neem aqueous crude leaf extract (NCE) was higher (8.33 ± 1.67 %) than that of

pawpaw aqueous crude leaf extract (PCE) (5.42 ± 3.16 %).

3.1.2 Preparation of stock solution of artesunate acid

Artesunate comes in pure powder form, for injection as artesunic acid (173). It is then

dissolved in 5% NaHCO3 and diluted with dextrose water. But, the powder used did

not dissolve in 5 % NaHCO3 but in 5% NaOH. The vehicle (7 % Tween 80 and 3 %

ethanol) used to dissolve artesunic acid (ARTA) here, is specified for antimalarial

work in the literature (171).

3.2 In vivo schizontocidal activity of combination of a fixed dose of artesunate

with varying doses of crude extract.

Appendix II show the pictures of the thin blood smear made during this test, while,

Fig. 4 and 5 show the graphical representation of the mean percentage parasitemia

lxxii

values (calculated from the parasitemia values shown in appendix III) of the various

dose levels of each drug and combination, i.e. each treatment, on day 4 and 7.

lxxiii

lxxiv

lxxv

The parasitemia level of the untreated group is expected to increase as seen in Fig. 5

for the combination treatment, but that of the single treatment, Fig. 4, did not increase

considerably. This is most probably due to the plasmodiastatic effect that was brought

about by contamination of the donor mice blood by Eperythrozoon coccoides, during

serial passage to maintain the parasite, during passage into test group, or was

transmitted by blood-feeding arthropod vectors like; lice (Polypax spinulosa and

Polypax serrata) (174). Eperythrozoon coccoides is an epierythrocytic organism that

causes mild haemolytic anaemia in laboratory and wild mice and is currently thought

to be a rickettsia. This parasite inhibits the growth of the plasmodium, and enables the

mice to survive longer as seen in Table 3, below even though they are infected with P.

berghei. The parasite can be detected on the erythrocytes by the use of Romanowsky

or acridine orange dyes and a fluoresence microscope, prevented by hygiene and

treated by the use of antibiotics like; organic arsenicals and neoarsphenamine.

Infections produce an intense parasitemia that peaks on day 2 to 5 (acute infection

stage) and subsequently declines rapidly (latent infection stage), so that by day 6 or 7,

the number of organisms in the peripheral blood is very low, and the organism may go

undetected in blood smears unless they are specifically searched for. Death from

infections with Eperythrozoon coccoides is very rare (174).

This may have produced an artificial enhancement in the antimalarial activity, i.e.

ability to suppress parasite growth, of neem 100 mg/kg (66.37%), as shown in Table

1, on day 4. For the single therapy phase, it is assumed that the parasite started

growing from day 6 or 7, when the effect of Eperythrozoon coccoides must have

subsided, thus allowing the P. berghei to multiply. According to a study, when albino

mice harboring a latent infection of Eperythrozoon coccoides are infected with P.

berghei, the former infection remains latent and exerts no influence on the course of

lxxvi

malaria infection, but if the two infections are introduced concurrently the course of

malaria will be affected (175). From literature, doses of aqueous neem leaf extract

found to be effective are 125 to 500 mg/kg. Here, the percentage reduction is 36.25%

and is not significant at p< 0.05.

When the parasite eventually started growing, on day 7, the parasitemia level

decreased for neem and artesunate in a dose dependent manner, while that of pawpaw

increased in a dose dependent manner, as it has weak activity, Fig.4. Neem and

Pawpaw may also be slow acting. The activity of artesunate decreased in a dose

dependent manner, with artesunate 6 mg/kg losing its activity on day 7, probably due

to recrudescence.

The reduction in the level of parasitemia, compared to the control (5.38%) was

significant for dose level of 1000 mg/kg of neem, 50 mg/kg of pawpaw, and dose

levels of 15 and 20 mg/kg of artesunate(1.56, 1.45,1.6 and 1.5% respectively) on day

7.

The combination treatment depicts clearly the behaviour of P. berghei in both treated

and untreated groups, with the parasitemia levels increasing from day 4 to day 7 for

the untreated and treated groups, Fig.5. The parasitemia of the treated group is

significantly lower for all treated groups on day 4 as compared to the control (4.80

%). On day 7, the parasitemia of the treated group are also significantly lower for all

treated groups as compared to the control (12.25%).

Table 1 shows the antimalarial activity, i.e., the mean percentage reduction in

parasitemia (calculated from the individual reduction in parasitemia shown in

appendix IV) of the drugs alone and in combination, compared to the control.

lxxvii

Table 1: Effect of artesunic acid and/or NCE or PCE on the growth of P. berghei in

mice, day 4 of checking parasitemia

Treatment Percentage of parasitemia reduction (%)

Arta 6 mg/kg 63.72 ± 16.96*

Arta 15 mg/kg 62.83 ± 17.31*

Arta 20 mg/kg 68.14 ± 8.51*

NCE 100 mg/kg 66.37 ± 11.54*

NCE 500 mg/kg 36.25 ± 32.07

NCE 1000 mg/kg 40.71 ± 23.54

PCE 50 mg/kg 37.70 ± 11.42

PCE 100 mg/kg 34.73 ± 24.34

PCE 200 mg/kg 59.29 ± 20.37*

Arta 15 mg/kg + NCE 100 mg/kg 79.17 ± 20 .50*

Arta 15 mg/kg + NCE 500 mg/kg 83.33 ± 11.88**

Arta 15 mg/kg + NCE 1000 mg/kg 96.87 ± 2.85***

Arta 15 mg/kg + PCE 50 mg/kg 81.25 ± 5.94****

Arta 15 mg/kg + PCE 100 mg/kg 76.04 ± 17.89*

Arta 15 mg/kg + PCE 200 mg/kg 58.04 ± 12.05*

*- Significantly greater than the control, **- significantly greater than the control &

pawpaw 50 mg,***-significantly greater than the control, pawpaw 50 mg, art 20 mg

and arta 15 mg + pawpaw 50 mg/ pawpaw 200 mg, ****- significantly greater than

pawpaw 50mg all at p< 0.05

Table 2 : Effect of artesunic acid and/or NCE or PCE on the growth of P. berghei in

mice, day 7 of checking parasitemia

lxxviii

Treatment Percentage reduction in parasitemia (%)

Arta 6 mg/kg 37.27± 18.78

Arta 15 mg/kg 70.26± 11.66*

Arta 20 mg/kg 72.12± 13.55*

NCE 100 mg/kg 37.27± 21.63

NCE 500 mg/kg 52.60± 21.40

NCE 1000 mg/kg 72.12± 13.15*

PCE 50 mg/kg 73.05± 10.07*

PCE 100 mg/kg 47.03± 19.33

PCE 200 mg/kg 48.96± 12.73

Arta 15 mg/kg + NCE 100 mg/kg 77.30± 14.70*

Arta 15 mg/kg + NCE 500 mg/kg 84.08± 5.67*

Arta 15 mg/kg + NCE 1000 mg/kg 89.80± 4.02*

Arta 15 mg/kg + PCE 50 mg/kg 57.59± 16.69*

Arta 15 mg/kg + PCE 100 mg/kg 72.57± 17.11*

Arta 15 mg/kg + PCE 200 mg/kg 81.40± 6.66*

*- Significantly greater than the control at p< 0.05

Table 1, shows that the treatment of P. berghei infected mice with 6 mg/kg of

artesunate, produced a significant reduction in parasitemia (63.72%) compared to the

control on day 4. Higher doses of artesunate 15 and 20 mg/kg equally significantly

lxxix

suppressed the parasitemia of the infected mice to give activities of 62.83 and 68.14

% respectively.

P. berghei infected mice treated with 100 mg/kg of neem and 200 mg/kg of pawpaw

experienced a significant reduction in their parasitemia levels (66.37 and 59.29 %

respectively), while those treated with 500 and 1000 mg/kg of neem, and 50 and 100

mg/kg of pawpaw experienced a mild reduction in their parasitemia levels compared

to the untreated group on day 4.

On day 7, the reduction in parasitemia of the single treatment group decreased for the

treatment groups that previously showed a significant reduction in parasitemia,

(artesunic acid 6mg/kg, pawpaw 200 mg/kg and neem 100 mg/kg), probably due to

recrudescence as a result of Eperythrozoon coccoides plasmodiastatic effect. But,

artesunic acid 15 and 20 mg/kg still retained their activity.

Paradoxically, pawpaw 50 mg/kg, significantly suppressed the parasitemia of the

infected mice on day 7 (69.09%), compared to the control. This may be due to its

slow activity. Increasing the dose of pawpaw produced a mild reduction in

parasitemia on day 7, probably due to ingredients of the complex mixture of the

pawpaw crude extract antagonizing the activity of one another at higher dose levels.

The combination treatment yielded a more significant reduction in parasitemia

compared to the control on day 4. The combination of artesunic acid 15 mg/kg and

pawpaw 50 mg/kg, produced a significant reduction in parasitemia compared to

pawpaw50mg/kg, alone, Table 1. The artesunate thus, enhanced the antimalarial

activity of pawpaw via a pharmacodynamic interaction.

lxxx

The combination of artesunate 15 mg/kg and neem 1000 mg/kg, produced a

significant reduction in parasitemia than artesunate 20 mg/kg, alone, the combination

of artesunic acid 15 mg/kg and pawpaw 50 mg/kg, pawpaw 50 mg/kg and the

combination of artesunic acid 15 mg/kg and pawpaw 200 mg/kg, Table 1. Even on

day 7, activity of this combination still remained significantly high (89.80 %), Table

2, although, it was not significantly higher than artesunic acid, 20mg/kg, alone. Neem

thus, enhances the antimalarial activity of artesunate via a pharmacodynamic

interaction and a pharmacokinetic interaction, as neem has been shown to increase the

serum concentrations of artesunate in our laboratory (176).

The combination of artesunic acid 15 mg/kg and neem 500 mg/kg produced a

significant reduction in parasitemia compared to pawpaw 50 mg/kg alone.

On day 7, all the combinations of artesunic acid 15 mg/kg and the crude extract still

retained their antimalarial activities.

3.3 Survival time and percentage cure of P. beghei infected mice after treatment

Table 3 and 4, show the survival time based on a 30 days observation period,

percentage survival at the end of this period and percentage of the infected mice that

are cured, i.e. do not have any parasite in their blood at day 30.

Table 3: Effect of single treatment (PCE or NCE or artesunic acid) to P. berghei

infected mice survival and percentage cure

Experimental

condition

Mean survival time

(days)

Percentage survival

(%)

Percentage cure

(%)

lxxxi

Control Group 19.80 ± 4.44 0.00 0.00

Arta 6 mg/kg 21.00 ± 3.74 0.00 0.00

Arta 15 mg/kg 21.80 ± 3.34* 0.00 0.00

Arta 20 mg/kg 22.80 ± 2.28* 0.00 0.00

NCE 100 mg/kg 11.20 ± 8.29 0.00 0.00

NCE 500 mg/kg 23.80 ± 4.76* 20.00 0.00

NCE 1000 mg/kg 18.20 ± 4.32 0.00 0.00

PCE 50 mg/kg 21.00 ± 3.74 0.00 0.00

PCE 100 mg/kg 17.40 ± 4.93 0.00 0.00

PCE 200 mg/kg 17.60 ± 5.27 0.00 0.00

* - Significant when compared to NCE 100 mg/kg at p< 0.05

Table 4: Effect of combination treatment (PCE or NCE and artesunic acid ) to P.

berghei infected mice survival and percentage cure

Experimental

condition

Mean survival time

(days)

Percentage

survival (%)

Percentage cure

(%)

Control group 11.20±4.55 0.00 0.00

Arta 15 mg + NCE 100 28.20±4.02* 80.00 40.00

lxxxii

mg/kg

Arta 15 mg + NCE 500

mg/kg

28.20±4.02* 80.00 60.00

Arta 15 mg + NCE

1000 mg/kg

28.20±4.02* 80.00 40.00

Arta 15 mg + PCE 50

mg/kg

23.00±6.60* 40.00 20.00

Arta 15 mg + PCE 100

mg/kg

22.60±8.62* 40.00 20.00

Arta 15 mg + PCE 200

mg/kg

19.60±5.94 20.00 20.00

* - Significant when compared to control at p< 0.05

The group treated with neem 100 mg/kg survived for an average of 11.20 days, its

survival time being lower than that of control group (19.8 days), and significantly

lower than that of the groups given neem 500 mg/kg, artesunate 15 and 20 mg/kg. Its

percentage survival and cure rate are 0%.

The survival times of the following groups are equally lower than that of the control

group, but not significantly lower; neem 1000 mg/kg, pawpaw 100 and 200 mg/kg.

This may be due to their toxicity at this dose level. This also, contributes to the

paradoxical effect of pawpaw 50 mg/kg, as it would have been expected to have a

lxxxiii

lower survival time as higher doses of the crude extract did. Their percentage survival

and cure rate are equally 0%. It is only one mouse in the group that received 500

mg/kg of neem that survived up till day 30, even so the mouse was still infected, thus

making their percentage survival to be 20% and their cure rate to be 0%.

For the combination treatment, all the treatment groups survived significantly

compared to the control (11.20 days, 0%), except for the combination of artesunate 15

mg/kg and papaw 200 mg/kg as shown in table 4. This further justifies its least

percentage in reduction in parasitemia, Table 2.

At least one of the mice in all the combination treatment group were cured, with the

highest cure rate being 60 % (for the combination of artesunate 15 mg and neem 500

mg) and the lowest cure rate being 20 % for all the combinations of artesunate and

pawpaw.

The percentage survival is highest for all the combinations of artesunate and neem

(80%), and lowest for the combination of artesunate 15 mg/kg and pawpaw 200

mg/kg (20 %). Possibly, some constituents of this high dose of the crude extract of

pawpaw are high enough to antagonize to an extent the antimalarial activity of some

other constituent of pawpaw leaf or artesunate itself. Pawpaw is known to have

antioxidant effect, it being a free radical scavenger, and helping with splenomegaly,

while artesunate is known to act by being converted to a free radical. Thus, a

pharmacological antagonism may be occurring at some minute level, thus inhibiting

to an extent the activity of artesunate. The toxicity of pawpaw at this dose may also

play a role in the reduction of the survival time, percentage survival and cure rate of

this combination.

lxxxiv

The percentage survival and cure rates are higher for all combinations than the extract

or pure drug given alone, except for the combination of artesunate 15 mg/kg and

pawpaw 200 mg/kg, whose percentage survival is equal to that of neem 500 mg/kg

group. Its survival time is equal to that of the control in the single treatment group.

The survival time for the combination of artesunate 15 mg/kg and pawpaw 50 mg/kg

is equal to that of neem 500 mg/kg.

3.4 Determination of ED50 of the crude extracts

Due to the plasmodiastatic effect brought about by Eperthrozoon coccoides, the ED50

of artesunic acid, and aqueous extract of neem and pawpaw leaf was calculated based

on day 7 not day 4.The ED50 of the drugs was calculated by using the linear equation

of the dose-response relationship of the drugs, shown in Fig. 6,7 and 8.

lxxxv

lxxxvi

lxxxvii

The ED50 of artesunate, neem and pawpaw aqueous leaf extract were; 8.814, 277.95

lxxxviii

and 143.53 mg/kg respectively. The ED50 of artesunic acid agrees with the ED50 of

artesunate against P. berghei, ANKA strain from literature, and is 8mg/kg (177).

The in vivo anti-plasmodial activity of the crude extract can be classified as moderate,

good and very good depending on if the extract displays a percentage growth

inhibition equal to or greater than 50% at a dose of 500, 250 and 100 mg/kg

respectively (178). Based on this, NCE has a moderate activity, while PCE has a very

good activity.

3.5 Determination of the kind of pharmacodynamic interaction between the pure

drug and plant extracts.

The ED90 of the drug alone and in combination was calculated from the linear

equation of the dose-response relationship of the drugs alone (on day 7) and in

combination (on day 7). (Fig. 6-10)

lxxxix

xc

The ED90 was then used to calculate the isobolar equivalent (IE) of NCE and PCE.

The IEs of NCE and PCE were 0.26 and 22.29 respectively. The IE of artesunic acid

xci

could not be calculated as its dose was fixed in the combination. Based on the IE, the

kind of pharmacodynamic interaction between the crude extracts and artesunic acid

was determined from the criteria; synergistic effect (IE < 1), additive effect (IE = 1),

antagonistic effect (IE > 1) (173).

Based on this, combinations of artesunate and NCE are synergistic for neem, while

the combinations of artesunate and PCE are antagonistic for pawpaw.

CHAPTER FOUR

CONCLUSIONS AND RECOMMENDATION

xcii

Most antimalarial drugs that are now in use were not developed on the basis of

rationally identified targets, but following serendipitous identification of the

antimalarial activity of natural products, for example; quinine and artemisinin,

compounds chemically related to natural products, for example chloroquine and

artesunate, or compounds active against other infectious agents, for example; the

antifolates and tetracycline. With the notable exception of the artemisinin family of

drugs, almost all antimalarial drugs developed till date are active against asexual stage

of the parasite and therefore do not prevent transmission of malaria. Transmission

blocking activity is a desirable property for a new antimalarial drug.

This study combines two antimalarial drugs with transmission blocking activity; neem

and artesunate and then pawpaw and artesunate. Neem enhanced the antimalarial

activity of artesunate, while artesunate enhanced the antimalarial activity of pawpaw.

The combinations of artesunate and neem also prolonged the survival of the treated

infected mice, compared to artesunate alone. This combinations even produced a cure

(at least 40%), while the doses of artesunate used in this study did not produce any

cure at the end of the 30 day period of this study. The combinations of artesunate and

pawpaw also prolonged the survival time of the infected mice and increased the cure

rate, compared to artesunate alone, but the percentage survival and cure rate was

lower than that of the combinations of artesunate and neem aqueous crude extract.

However, the combinations of artesunate and pawpaw are antagonistic for pawpaw,

despite the fact that artesunate enhances its activity, because its IE is 22.29, and even

its percentage survival is lowest for the combination of its highest dose with

artesunate. Combinations of artesunate and pawpaw show little promise.

xciii

The combinations of neem and artesunate are synergistic for neem. Therefore, this

combination is therefore a promising candidate for a new antimalarial combination

therapy development, although the dose level of the neem extract is high. The

findings in this study show that it is possible to have a neem constituent- based

combination therapy, and this is supported by an earlier finding that Azadirachtin-

based compounds as antimalarial agents(with transmission blocking potential) can be

developed as Azadirachtin of A. indica was able to block the development of motile

malaria gametes in vitro.

Other techniques can be used to evaluate the drug interaction between artesunate and

neem leaf extract, so as to determine if such combinations are synergistic, additive or

antagonistic for artesunate.

Isolation of a particular antimalarial constituent of the neem leaf extract, which can be

combined with artesunate, can be done, thus making a new and effective orthodox

combination therapy.

The neem aqueous leaf extract may be toxic to infants and pregnant women at the

dose levels used in the combinations, and so the combinations may also be toxic to

them.

REFERENCES

xciv

1. Ukwe, C.V. (2004). Malaria: In Therapeutic basis of Clinical Pharmacy in the

Tropics, Aguwa C. N.(Ed.) 3rd

Edn., SNAAP Press Ltd., Enugu, pp 250-252.

2. World Malarial Report (2008). Information available at

http://www.who.int/malaria/docs/TreatmentGuidelines2008.pdf. (accessed on

11/03/10﴿

3. Smith AL (1978). Text on Microbiology and Pathology, 11th edition, CV

Mosby Com. USA.

4. WHO (2000). Press release on African Summit on Roll Back Malaria.

5. African Summit on Roll Back Malaria (2000). Summit on Roll Back Malaria

Abuja, Nigeria. Abuja Malsum Int. brief 2: 02 March.

6. Abuja Malaria Summit (2000). Available on: J. /Div. Data/Abuja Malaria

Summit/Abjbint2. doc.

7. Abu-Raddad L, Patnaik P, Kublin J (2006).Dual infection with HIV and

malaria fuels the spread of both diseases in sub-Saharan Africa. Science 314

(5805): 1603–6.

8. Malaria. Centres for Disease Control and Prevention. 1600mClifton Rd.

Atlanta, GA 30333, USA. Available at [email protected]. (accessed on

11/04/10﴿

9. Fong YL, Cadigan FC, Coatney GR (1971). A presumptive case of naturally

occurring Plasmodium knowlesi malaria in man in Malaysia. Trans. R. Soc.

Trop. Med. Hyg. 65 (6): 839–40.

10. Singh B, Kim Sung L, Matusop A, Radhakrishnan A, Shamsul SS, Cox-Singh

J, Thomas A and Conway DJ (2004). A large focus of naturally acquired

Plasmodium knowlesi infections in human beings. Lancet, 363(9414):1017-

1024.

xcv

11. Beare, N.A, Taylor, T.E, Harding, S.P, Lewallen, S, and Molyneux, M.E

(2006). Malarial retinopathy: a newly established diagnostic sign in severe

malaria. Am. J. Trop. Med. Hyg. 75 (5): 790–7.

12. Idro, R, Otieno, G., White, S., Kahindi, A., Fegan, G., Ogutu, B., Mithwani, S,

Maitland, K, Neville, BG and Newton, CR. Decorticate, decerebrate and

opisthotonic posturing and seizures in Kenyan children with cerebral malaria.

Malaria Journal 4 (57): 57

13. Boivin, MJ. (2002). Effects of early cerebral malaria on cognitive ability in

Senegalese children. J Dev Behav. Pediatr. 23 (5): 353–64.

14. Holding, PA, Snow, RW. (2001). Impact of Plasmodium falciparum malaria

on performance and learning: review of the evidence. Am. J. Trop. Med. Hyg.

64 (1-2 Suppl): 68–75.

15. Maude, RJ, Hassan, MU and Beare, NAV. (2009). Severe retinal whitening in

an adult with cerebral malaria. Am J Trop Med Hyg. 80 (6): 881.

16. Trampuz, A, Jereb, M, Muzlovic, I and Prabhu, R. (2003). Clinical review:

Severe malaria. Crit Care 7 (4): 315–23.

17. Kain, K, Harrington, M, Tennyson, S and Keystone, J. (1998). Imported

malaria: prospective analysis of problems in diagnosis and management. Clin

Infect Dis 27 (1): 142–9.

18. Mockenhaupt, F, Ehrhardt, S, Burkhardt, J, Bosomtwe, S, Laryea, S,

Anemana, S, Otchwemah, R, Cramer, J, Dietz, E, Gellert ,S and Bienzle, U.

(2004). Manifestation and outcome of severe malaria in children in northern

Ghana. Am J Trop Med Hyg 71 (2): 167–72.

xcvi

19. Carter, JA, Ross, AJ, Neville, BG, Obiero, E, Katana, K, Mung'ala-Odera, V,

Lees, JA and Newton, CR. (2005). Developmental impairments following

severe falciparum malaria in children. Trop Med Int Health 10 (1): 3–10.

20. Adak, T, Sharma, V and Orlov, V (1998). Studies on the Plasmodium vivax

relapse pattern in Delhi, India. Am. J. Trop. Med. Hyg. 59 (1): 175–9.

21. Lindsay, S, Ansell, J, Selman, C, Cox, V, Hamilton, K and Walraven G.

(2000). Effect of pregnancy on exposure to malaria mosquitoes. Lancet 355

(9219): 1972.

22. Van Geertruyden, J, Thomas, F, Erhart, A and D'Alessandro, U. (2004). The

contribution of malaria in pregnancy to perinatal mortality. Am. J. Trop. Med.

Hyg. 71 (2 Suppl): 35–40.

23. Rodriguez-Morales, AJ, Sanchez, E, Vargas, M, Piccolo, C, Colina, R, Arria,

M and Franco-Paredes C. (2006). Pregnancy outcomes associated with

Plasmodium vivax malaria in northeastern Venezuela. Am J Trop Med Hyg 74

(5): 755–757.

24. Monica Cheesbrough (2005). District Laboratory Practice in Tropical

Countries. Cambridge University press, Edinbourgh, UK. pp 239-258.

25. Mueller, I, Zimmerman, PA and Reeder, JC (2007). Plasmodium malariae and

Plasmodium ovale - the "bashful" malaria parasites. Trends Parasitol. 23 (6):

278–83.

26. Mendis, K, Sina, B, Marchesini, P and Carter R. (2001). The neglected burden

of Plasmodium vivax malaria. Am. J. Trop. Med. Hyg. 64 (1-2 Suppl): 97–106.

27. Escalante, A and Ayala, F (1994). Phylogeny of the malarial genus

Plasmodium, derived from rRNA gene sequences. Proc. Natl. Acad. Sci. USA

91 (24): 11373–7.

xcvii

28. Garnham, PCC. (1966). Malaria parasites and other haemosporidia. Oxford:

Blackwell Scientific Publications. ISBN 186983500X.

29. Collins, WE and Barnwell, JW. (2009). Plasmodium knowlesi: Finally being

recognized. J Infect. Dis. 199 (8): 1107–1108

30. Bledsoe, GH (2005). Malaria primer for clinicians in the United States. South.

Med. J. 98 (12): 1197–204.

31. Icke, G, Davis, R and McConnel, W (2005). Teaching health workers malaria

diagnosis. PLoS Med 2: e11. doi: 10.1371/journal.pmed.0020011.

32. http://en.wikipedia.org/wiki/Plasmodium

33. Chen, Q, Schlichtherle, M and Wahlgren, M (2000). Molecular aspects of

severe malaria. Clin. Microbiol. Rev. 13 (3): 439–50.

34. Adams, S, Brown H and Turner G. (2002). Breaking down the blood-brain

barrier: signaling a path to cerebral malaria? Trends Parasitol 18 (8): 360–6.

35. Lang-Unnasch, N. and Murphy, A.D. (1998). Metabolic changes of the

malaria parasite during the transition from the human to the mosquito host.

Annu. Rev. Microbiol. 52: 561-590.

36. Ginsburg, H. (1994). Transport pathways in the malaria-infected erythrocyte -

their characterization and their use as potential targets for chemotherapy.

Biochem Pharmacol 48: 1847-1856.

37. Atamna, H. and Ginsburg, H. (1997). The malaria parasite supplies

glutathione to its host cell-Investigation of glutathione transport and

metabolism in human erythrocytes infected with Plasmodium falciparum. Eur.

J. Biochem. 250: 670-679.

xcviii

38. Uyemura, S.A. , Luo, S. , Moreno, S.N.J and Docampo, R. (2000). Oxidative

phosphorylation, Ca2+

transport, and fatty acid-induced uncoupling in malaria

parasites mitochondria. J. Biol. Chem. 275: 9709-9715.

39. Mitamura, T. and Palacpac, N.M.Q. (2003). Lipid metabolism in Plasmodium

falciparum-infected erythrocytes: possible new targets for chemotherapy.

Microbes and Infection 5, 545-552.

40. Krugliak, M., Zhang, J. and Ginsburg, H. (2002). Intraerythrocytic

Plasmodium falciparum utilizes only a fraction of the amino acids derived

from the digestion of host cell cytosol for the biosynthesis of its proteins. Mol.

Biochem. Parasitol. 119: 249-256.

41. Gutteridge, W.E., Dave, D. and Richards, W.H. (1979). Conversion of

dihydroorotate to orotate in parasitic protozoa. Biochem. Biophys. Acta 582:

390-401.

42. Goldberg DE (2005). Hemoglobin degradation. Curr Top Microbiol Immunol

295: 275-291.

43. Goldberg, DE, Slater, AFG, Beavis, RC, Chait, B, Cerami, A, and Henderson,

GB (1991) Hemoglobin degradation in the human malaria pathogen

Plasmodium falciparum: a catablolic pathway initiated by a specific aspartic

protease. J Exp Med 173: 961-969.

44. Egan, TJ (2008) Haemozoin formation. Mol Biochem Parasitol 157: 127-136

45. Ginsburg H, Ward SA, and Bray PG (1999). An integrated model of

chloroquine action. Parasitol. Today 15: 357.

46. World Health Organization. Guidelines for the Treatment of Malaria. Geneva,

Switzerland: World Health Organization; 2006. Available at:

http://www.who.int/malaria/docs/TreatmentGuidelines2006.pdf

xcix

47. National Antimalaria Treatment Guildlines.(2005). Federal Ministry of Health,

National Malaria and Vector Control Division Abuja-Nigeria.

48. Sutherland, CJ and Hallett, R. (2009). Detecting malaria parasites outside the

blood. J Infect Dis 199 (11): 1561–1563.

49. Warhurst DC and Williams JE (1996). Laboratory diagnosis of malaria. J Clin

Pathol 49 (7): 533–38.

50. http://en.wikipedia.org/wiki/Malaria

51. Ling IT., Cooksley S., Bates PA., Hempelmann E. and Wilson, RJM. (1986).

Antibodies to the glutamate dehydrogenase of Plasmodium falciparum.

Parasitology 92: 313–24.

52. Mens, PF, Schoone, GJ, Kager, PA, and Schallig, HD. (2006). Detection and

identification of human Plasmodium species with real-time quantitative

nucleic acid sequence-based amplification. Malaria Journal 5 (80): 80.

53. Barat, L (2006). Four malaria success stories: how malaria burden was

successfully reduced in Brazil, Eritrea, India, and Vietnam. Am J Trop Med

Hyg 74 (1): 12–6.

54. http://www.cdc.gov/malaria/history/eradication_us.htm Centers for Disease

Control. Eradication of Malaria in the United States (1947-1951) 2004.

(accessed on 23/02/10)

55. Ito J, Ghosh A, Moreira L. A., Wimmer E. A. and Jacobs-Lorena M (2002).

Transgenic anopheline mosquitoes impaired in transmission of a malaria

parasite. Nature 417 (6887): 387–8.

56. Robert, G. (2009). Rocket Scientists Shoot Down Mosquitoes With Lasers.

WSJ.com.

c

57. World Health Organization; 2006.Indoor Residual Spraying: Use of Indoor

Residual Spraying for Scaling Up Global Malaria Control and Elimination.

58. Rosenberg, T (2007). What the world needs now is DDT. New York Times.

59. Pates, H and Curtis, C. (2005). Mosquito behaviour and vector control. Annual

Review on Entomology 50: 53-70.

60. Bachou, H, Tylleskär T, Kaddu-Mulindwa, DH and Tumwine, JK. (2006).

Bacteraemia among severely malnourished children infected and uninfected

with the human immunodeficiency virus-1 in Kampala, Uganda. BMC Infect.

Dis. 6: 160.

61. Rowland M, Durrani N, Hewitt S, Mohammed N, Bouma M, Carneiro I,

Rozendaal J and Schapira, A. (1999). Permethrin-treated chaddars and top-

sheets: appropriate technology for protection against malaria in Afghanistan

and other complex emergencies. Trans R Soc Trop Med Hyg 93 (5): 465–72.

62. Matuschewski, K. (2006). Vaccine development against malaria. Curr Opin

Immunol 18 (4): 449–57.

63. Heppner, DG, Jr, Kester KE, Ockenhouse CF, Tornieporth N, Ofori O, Lyon

JA, et al. (2005). Towards an RTS,S-based, multi-stage, multi-antigen vaccine

against falciparum malaria: progress at the Walter Reed Army Institute of

Research. Vaccine 23 (17-18): 2243–50.

64. Alonso, P, Sacarlal, J., Aponte, J., Leach, A., Macete, E. Milman, J.,

Mandomando, I, Spiessens, B., Guinovart, C., Espasa, M. (2004). Efficacy of

the RTS,S/AS02A vaccine against Plasmodium falciparum infection and

disease in young African children: randomized controlled trial. Lancet 364

(9443): 1411–20.

65. Africa: Malaria - Vaccine Expected in 2011. Accra Mail. 2007.

ci

66. Corby-Harris, V., Drexler, A., Watkins de Jong, L., Antonova, Y., Pakpour,

N., Ziegler, R., Ramberg, F., Lewis, E., Brown, J., Luckhart, S., and Riehle,

M. (2010). Activation of Akt Signaling Reduces the Prevalence and Intensity

of Malaria Parasite Infection and Lifespan in Anopheles stephensi Mosquitoes.

PLoS Pathog 6 (7): e1001003.

67. Kaufman, T and Rúveda, E (2005). The quest for quinine: those who won the

battles and those who won the war. Angew Chem Int Ed Engl 44 (6): 854–85.

68. Kyle, R and Shampe, M (1974). Discoverers of quinine. JAMA 229 (4): e320.

69. Coatney GR. (1963) Pitfalls in a discovery: the chronicle of chloroquine. Am.

J. Trop. Med. and Hyg. 12:121-128.

70. Oxford Concise Medical Dictionary. 2002. 6th

Edn., p 124.

71. Rosenthal, R.J. and Goldsmith, R.S.(2002). Antiprotozoal drugs, In: Basic and

Clinical Pharmacology ( Katzung, B. G. ed.), 8th

Edn., Lange Medical Books/

MCGraw-Hill. Chapter 53, Pp 892-893.

72. Dorsey, G., Vlahos, J., Kamya, M. R., Staedke, S. G. and Rosenthal, P. J.

(2003). Prevention of increasing rates of treatment failure by combining

sulfadoxine-pyrimethamine with artesunate or amodiaquine for the sequential

treatment of malaria. J. Infect. Dis. 188, 1231–1238.

73. Looareesuwan, S., Chulay, J. D., Canfield, C. J. and Hutchinson, D. B. (1999).

Malarone (atovaquone and proguanil hydrochloride): a review of its clinical

development for treatment of malaria. Malarone Clinical Trials Study Group.

Am. J. Trop. Med. Hyg. 60, 533–541.

74. Fidock , D. A., Rosenthal , P. J., Croft , S.L., Brun , R. and Nwaka , S., (2004).

Antimalarial drug discovery: efficacy models for compound screening. Nature

reviews. drug discovery .3:509-520.

cii

75. Aweeka, F.T and German, P. I. (2008). Clinical Pharmacokinetics: Clinical

Pharmacology of Artemisinin-Based Combination Therapies. Review Article.

47(2):91-102.

76. Meshnick, SR, Taylor, TE, and Kamchonwongpaisan, S. (1996). Artemisinin

and the Antimalarial Endoperoxides: from Remedy to Targeted

Chemotherapy. Microbiological reviews 60(2)301–315.

77. Eckstein-Ludwig, U, Webb RJ, Van Goethem, ID, East, JM, Lee, AG,

Kimura, M and O'Neill, PM. (2003). Artemisinins target the SERCA of

Plasmodium falciparum. Nature. 424: 957-961.

78. World health organization (2000). Severe falciparum malaria. Transaction of

the royal Society of Tropical Medicine and Hygiene 94: 36-37.

79. Hallett, RL, Sutherland, CJ, Alexander, N, Ord R, Jawara, M, Drakeley, CJ,

Pinder, M, Walraven, G, Targett, GA and Alloueche. (2004). A Combination

therapy counteracts the enhanced transmission of drug-resistant malaria

parasites to mosquitoes. Antimicrob Agents Chemother. 48: 3940-3943.

80. White,N.J.,(1997). Assessment of the Pharmacodynamic properties of

Antimalaria drugs in vivo. Antimicrobial agents and chemotherapy. 41: 1413-

1422.

81. White, N.J., (1998) Preventing antimalarial Drug Resistance through

combination. Drug-Resistance updates, 1:3-9.

82. Price, RN, Nosten, F, Luxemburger, C, ter Kuile, FO, Paiphun, L,

Chongsuphajaisiddhi, T and White, NJ. (1996). Effects of artemisinin

derivatives on malaria transmissibility. Lancet 347 (9016): 1654–8.

ciii

83. Mutabingwa, TK. (2005). Artemisinin-based combination therapies (ACTs):

best hope for malaria treatment but inaccessible to the needy! Acta Trop

95:305–15.

84. World Health Organization. (2001). Antimalarial Drug Combination Therapy:

Report of a WHO Technical Consultation. Geneva, Switzerland.

85. Omari AA, Preston C, Garner P. Artemether-lumefantrine for treating

uncomplicated falciparum malaria. Cochrane Database Syst Rev. 2002;

CD003125

86. Duraisingh, MT and Cowman, AF. (2005). Contribution of the pfmdr1 gene

to antimalarial drug-resistance. Acta Trop. 94: 181-190.

87. Ojewole, J.A.O., (2004). Drug interactions. In: Therapeutic basis of Clinical

Pharmacy in the Tropics, 3rd

Edn.(Aguwa, C. N.(ed.), Published by SNAAP

Press Ltd., Enugu, Nig. pp. 23-48

88. Hansten, P. and Horn, J. (2003). Drug-drug interaction mechanism. H&H

Publications.

89. Young, J. A., Fivelman, Q. L., Blair ,P. L., de la Vega, P. and Le Roch ,K. G.,

(2005). The Plasmodium falciparum sexual development transcriptome: a

microarray analysis using ontology-based pattern identification. Mol Biochem

Parasitol.143: 67-79.

90. Strickland GT, Fox E, Sarwar M, Khaliq AA and Macdonald M.(1986).

Effects of chloroquine, amodiaquine and pyrimethamine-sulfadoxine on

Plasmodium falciparum gametocytemia. Am J Trop Med Hyg. 35: 259-262.

91. Sokhna, CS, Trape, JF and Robert, V. (2001). Gametocytaemia in Senegalese

children with uncomplicated falciparum malaria treated with chloroquine,

amodiaquine or sulfadoxine + pyrimethamine. Parasite. 8: 243-250.

civ

92. Buckling, A, Ranford-Cartwright, LC, Miles, A, and Read, AF. (1999).

Chloroquine increases Plasmodium falciparum gametocytogenesis in vitro.

Parasitology. 118(pt 4): 339-346.

93. Montgomery, R. and Eyles, D.E. (1963). Chloroquine resistant falciparum

malaria in Malaya. Transactions of the Royal society of Tropical Medicine and

Hygiene 57: 409-416.

94. Clyde, D.F., Han, C.M. and Haung, Y.S. (1973).Resistance to chloroquine of

Plasmodium falciparum from Sabah. Transactions of the Royal Society of

Tropical Medicine and Hygiene 67:146.

95. Campbell, CC, Chin, W, Collins, WE, Teutsch, SM and Moss, DM (1979).

Chloroquine-resistant Plasmodium falciparum from East-Africa: cultivation

and drug sensitivity of the Tanzanian I/CDC strain from an American tourist.

Lancet, ii: 1151-1154.

96. Fogh, S, Jepsen, S, and Effersoe, P (1979). Chloroquine-resistant Plasmodium

falciparum malaria in Kenya. Transactions of the Royal Society of Tropical

Medicine and Hygiene. 73:228-229.

97. Aronson, B., Bengsston, E., Bjorkman, A., Pehrson, PO., Rombo, L. and

Wahlgren, M. (1981). Chloroquine-resistant falciparum malaria in Madasgar

and Kenya. Annals Trop. Med. Parasitol. 75:367-373.

98. Onori, E (1984). The problem of Plasmodium falciparum drug resistance in

Africa south of the Sahara. Bull. World Health Organization. 62(Suppl.):55-

62.

99. Ekue , JM, Ulrich ,AM, Njelesani EK (1983). Plasmodium Malaria resistant

to chloroquine in a Zambian living in Zambia. British Medical Journal

(Clinical Research Ed.). 286:1315-1316.

cv

100. Fogh, S, Jepsen, S, and Mataya, RH (1984). R-III Chloroquine-resistant

Plasmodium falciparum malaria from Northern Malawi. Transactions of the

Royal Society of Tropical Medicine and Hygiene 78:282

101. Slatter MJ, Pettengell JK, and Taylor RK (1983). Chloroquine-resistant

malaria. South African Medical Journal. 63:838.

102. Dondero, T.J.Jr., Parsons, R.E. and Ponnampalam, J.T. (1976). Studies on

the resistance of malaria to chloroquine and to a combination of chloroquine

and pyrimethamine in Peninsular Malaysia. Transactions of the Royal Society

of Tropical Medicine and Hygiene 70: 145-148

103. Hurwitz, E.S., Johnson, D. and Campbell, C.C. (1981). Resistance of

Plasmodium falciparum malaria to sulfadoxinepyrimethamine (‘Fansidar’) in

a refugee camp in Thailand. Lancet 1: 1068-1070.

104. Onori, E. (1988). Malaria control constraints.In: Malaria Principles and

Practice of Malariology (Editors, W.H. Wernsdorfer & I.A. McGregor)

Churchill Livingstone. Edinburgh .pp. 1721-1740.

105. Black, F., Bygbjerg, I., Effersoe, P., Gomme,G., Jepsen S. and Jensen, G.A.

(1982).Fansidar resistant falciparum malaria acquired in South East Asia.

Transactions of the Royal Society of Tropical Medicine and Hygiene 75: 715-

716.

106. Ponnampalam, J.T. (1982). Falciparum malaria resistant to Fansidar

(sulphadoxine-pyrimethamine) occurring in three children of the same family.

Singapore Medical Journal 23:37-38.

107. Lokman H.S., Sharifah Roohi, S.W., Zurkurnai, Y., Noor Rain, A., Mansor,

S.M., Palmer, K., Navaratnam, V. and Mak, J.W. (1996). Plasmodium

falciparum: Increased proportion of severe resistance (RII and RIII) to

cvi

chloroquine and high rate of resistance to sulfadoxine-pyrimethamine in

Peninsular Malaysia after two decades. Transactions of the Royal Society of

Tropical Medicine and Hygiene 90: 294-297.

108. Nosten, F et al., (2000). Effects of artesunate–mefloquine combination on

incidence of Plasmodium falciparum malaria and mefloquine resistance in

western Thailand: a prospective study, Lancet 356: 297–302

109. WHO bulletin (2000) vol.78, No. 12:1378-1389

110. Adjuik, M, Agnamey,P, Babiker, A, Borrmann, S., Brasseur, P., Cisse, M,

Cobelens, F and Diallo, S. (2002). Amodiaquine-artesunate versus

amodiaquine for uncomplicated Plasmodium falciparum malaria in African

children: a randomised, multicentre trial. Lancet 359:1365–72.

111. Mutabingwa TK, Anthony D, Heller A, Hallett R, Ahmed J, Drakeley C, and

Greenwood BM, (2005). Amodiaquine alone, amodiaquine + sulfadoxine-

pyrimethamine, amodiaquine + artesunate, and artemether-lumefantrine for

outpatient treatment of malaria in Tanzanian children: a four-arm randomised

effectiveness trial. Lancet 365:1474–80.

112. Menard, D., Matsika-Claquin,M. D., Djalle,D, Yapou,F., Manirakiza, A,

Dolmazon,V, Sarda,J and Talarmin, A. (2005). Association of failures of

seven-day courses of artesunate in a non-immune population in Bangui,

Central African Republic with decreased sensitivity of Plasmodium

falciparum. Am J Trop Med Hyg 73:616–21.

113. Krishna S, Woodrow CJ, Staines HM, Haynes RK and Mercereau-Puijalon

O. (2006) Re-evaluation of how artemisinins work in light of emerging

evidence of in vitro resistance. Trends Mol Med 12:200–5.

cvii

114. Fidock DA, Nomura T, Talley AK, Cooper RA, Dzekunov SM, Ferdig MT,

Ursos LM, Sidhu ABS, Naude B, Deitsch K, Su X-Z, Wootton JC, Roepe PD,

Wellems TE. (2000). Mutations in the P. falciparum digestive vacuole

transmembrane protein PfCRT and evidence for their role in chloroquine

resistance. Mol Cell. 6:861-871.

115. Sidhu AB, Verdier-Pinard D and Fidock DA (2000). Chloroquine resistance

in Plasmodium falciparum malaria parasites conferred by pfcrt mutations.

Science. 298:210-213.

116. Reed MB, Saliba KJ, Caruana SR, Kirk K and Cowman AF (2000). Pgh1

modulates sensitivity and resistance to multiple antimalarials in Plasmodium

falciparum. Nature. 403:906-909.

117. Price RN, Cassar C, Brockman A, Duraisingh M, van Vugt M, White NJ,

Nosten F and Krishna S (1999). The pfmdr1 gene is associated with a

multidrug-resistant phenotype in Plasmodium falciparum from the western

border of Thailand. Antimicrob Agents Chemother . 43:2943-2949.

118. Price RN, Uhlemann AC, Brockman A , McGready R, Ashley E, Phaipun L,

Patel R, Laing K, Looareesuwan S, White NJ, Nosten F, and Krishna S.

(2004). Mefloquine resistance in Plasmodium falciparum and increased

pfmdr1 gene copy number. Lancet. 364:438-447.

119. Duraisingh MT, Jones P, Sambou I, von Seidlein L, Pinder M and Warhurst

DC (2000). The tyrosine-86 allele of the pfmdr1 gene of Plasmodium

falciparum is associated with increased sensitivity to the anti-malarials

mefloquine and artemisinin. Mol Biochem Parasitol . 108:13-23.

cviii

120. Peterson DS, Walliker D and Wellems TE (1988). Evidence that a point

mutation in dihydrofolate reductase-thymidylate synthase confers resistance to

pyrimethamine in falciparum malaria. Proc Natl Acad Sci USA. 85:9114-

9118.

121. Brooks DR, Wang P, Read M, Watkins WM, Sims PF, and Hyde JE (1994).

Sequence variation of the hydroxymethyldihydropterin pyrophosphokinase:

dihydropteroate synthase gene in lines of the human malaria parasite,

Plasmodium falciparum, with differing resistance to sulfadoxine. Eur J

Biochem. 224:397-405.

122. Dokomajilar C, Lankoande ZM, Dorsey G, Zongo I, Ouedraogo JB and

Rosenthal PJ (2006). Roles of specific Plasmodium falciparum mutations in

resistance to amodiaquine and sulfadoxine-pyrimethamine in Burkina Faso.

Am J Trop Med Hyg . 75:162-165.

123. Humphreys GS, Merinopoulos I, Ahmed J, Whitty CJ, Mutabingwa TK,

Sutherland CJ, and Hallett RL. (2006). Amodiaquine and artemether-

lumefantrine select distinct alleles of the P. falciparum pfmdr1 gene in

Tanzanian children treated for uncomplicated malaria. Antimicrob Agents

Chemother. 51:991-997.

124. Sisowath, C., J. Stromberg, A. Martensson, M. Msellem, C. Obondo, A.

Bjorkman, and J. P. Gil. (2005). In vivo selection of Plasmodium falciparum

pfmdr1 86N coding alleles by artemether-lumefantrine (Coartem). J Infect

Dis. 191:1014-1017.

125. Dokomajilar C, Nsobya SL, Greenhouse B, Rosenthal PJ and Dorsey G

(2006). Selection of Plasmodium falciparum pfmdr1 alleles following therapy

cix

with artemether-lumefantrine in an area of Uganda where malaria is highly

endemic. Antimicrob Agents Chemother . 50:1893-1895.

126. Volkman SK, Sabeti PC, Decaprio D, Neafsey DE, Schaffner SF, Milner

DA Jr, et al. (2007). A genome-wide map of diversity in Plasmodium

falciparum. Nat Genet, 39:113-119.

127. Ferdig MT, Cooper RA, Mu J, Deng B, Joy DA, Su XZ, et al.(2004).

Dissecting the loci of low-level quinine resistance in malaria parasites. Mol

Microbiol 52:985-997.

128. Mu J, Awadalla P, Duan J, McGee KM, Keebler J, Seydel K, et al (2007).

Genome-wide variation and identification of vaccine targets in the

Plasmodium falciparum genome. Nat Genet. 39:126-130.

129. Plowe1, CV, Roper , C, Barnwell , JW, Happi , CT, Joshi , H.H, Mbacham ,

W, Meshnick , S, Mugittu , K, Naidoo , I, Price , RN, Shafer, RW, et al.

(2007). World Antimalarial Resistance Network (WARN) III: Molecular

markers for drug resistant malaria. Malaria Journal. 6:121.

130. Rieckmann K.H. et al. (1978). Drug sensitivity of Plasmodium falciparum.

An in-vitro microtechnique. Lancet, 1:22–23.

131. Sanjib Bhattacharya (2009). Phytosomes: The New Technology for

Enhancement of Bioavailability of Botanicals and Nutraceuticals.

International Journal of Health Research, 2(3): 225-232 .

132. Fabricant DS and Farnsworth NR (2001). The value of plants used in

traditional medicine for drug discovery. Environ. Health Perspect. 109 (1):

69–75.

cx

133. Interactive European Network for Industrial Crops and their Applications

(2000-2005). Summary Report for the European Union.

http://ec.europa.eu/research/quality-of-life/ka5/en/00111.html.

134. Benoit-Vical, F, Valentin, A, Pélissier, Y et al. (1996). In vitro antimalaria

activity of vegetable extracts used in West African Traditional Medicine. Am J

Trop Med Hyg 54:67-71.

135. Ehiagbonare, J.E. (2007). Vegetable propagation of some key malaria

medicinal plants in Nigeria. Scientific Research & Essay 2(2): 037-039.

136. Ajaiyeoba, E.O. (2009). Overview and new directions for research in drug

discovery for malaria from the biodiversity of Nigeria. Overview. Presented at

Research Continuum in Natural Products Drug Discovery, Transcorp Hilton

Abuja, May 12 – 14.

137. Adjanohoun, J.E.; Aboubakar, N.; Dramane, K.; Ebot, M.E.; Ekpere, J.A.;

Enoworock, E.G.; Focho, D.; Gbile, Z.O.; Kamanyi, A.; Kamsu, K. J.; Keita,

A.; Mbenkum, T.; Mbi, C.N.; Mbiele, A.C.; Mbome, J.C.; Muberu, N.K.;

Nancy, W.L.; Kongmeneck, B.; Satabie, B.; Sowora, A.; Tamze, V. and

Wirmum, C.K. (1996). Traditional Medicine and Pharmacopoeia:

Contribution to ethno botanical and floristic studies in Cameroon. Ed.

Organization of African Unity; Scientific, Technical and Research

Commission.

138. Ngemenya, M.N., Tane, P., Berzins, K. and Titanji, V.P.K. (2004).

Antiplasmodial Activity of some Medicinal Plants used in Cameroon:-

preliminary toxicity studies of highly active extracts . XIth Annual Conference

of the Cameroon Bioscience Society 16- 18 December, 2004.

cxi

139. Uhegbu, F.O., Elekwa,I. and Ukoha,C.(2008). Comparative efficacy of

crude aqueous extract of Magnifera indica, Carica papaya and sulphadoxine

pyrimethamine on mice infested with malaria parasite in vivo. CABI

Abstracts.

140. Iwu MM, Obidoa O, and Anazodo M. (1986). Biochemical mechanism of

the antimalarial activity of Azadirachta indica leaf extract, Pharmacological

Research Communications, 18(1):81-91

141. Ayoola, G.A., Coker, H.A.B., Adesegun, S.A., Adepoju-Bello, A.A.,

Obaweya, K., Ezennia, E.C. and Atangbayila,T.O. (2008). Phytochemical

screening and antioxidant activities of some selected medicinal plants used for

malaria therapy in southwestern Nigeria. Tropical Journal of Pharmaceutical

Research, 7(3): 1019-1024.

142. http://en.wikipedia.org/wiki/Neem

143. Kausik B, Ishita C, Ranajitk B, Uday B. (2002). Biological activities and

medicinal properties of neem. Current Science, 82:1336-1345.

144. Report, Board on science and Technology for International development,

National Research council, National Academy Press, Washington DC, 1992,

pp. 60-113.

145. Ekanem, T. (1978) Dongoyaro .Does it work? Nigerian Med J. 8(1): 8 – 10

146. Aladesanmi, A. J., Awe, S.O., Adesanya, S.A. and Bray, D.H. (1988).

Antimalarial activity of some Nigerian Medicinal Plants. In: Drug Production

from Natural Products. Proceedings of the Seventh International Symposium

(ed. Adesina, S.K.). Medex Publications Ltd., Lagos, Nigeria; 32-36.

cxii

147. Mohammed Aslam (1996). Asian Medicine and its practice in Britain, In:

Trease and Evans Pharmacognosy, 14th

edn., WB Saunders Company Ltd.

148. Dhar, R, Zhang, K, Talwar, GP, Garg, S and Kumar, N. (1998). Inhibition of

the growth and development of asexual and sexual stages of drug-sensitive

and resistant strains of the human malaria parasite Plasmodium falciparum by

Neem (Azadirachta indica) fractions. J. Ethnopharm.,61(1), 31-39.

149. Udeinya, J. I., Shu, E. N., Quakyi, I. and Ajayi, F. O. (2008) An

Antimalarial Neem Leaf Extract has Both Schizonticidal and Gametocytocidal

Activities. American Journal of Therapeutics, 15:108-110.

150. Khalid, S. A., Duddect, H. and Gonzalez-Sierra, M. (1989). Isolation and

characterization of an antimalarial agent of the neem tree Azadiractha indica.

J. Natural Products. 52: 922-927.

151. Khalid, S. A., Farouk, A., Geary, T. G. and Jensen, J. B. (1986), Isolation

and characterization of an antimalarial agent of the neem tree Azadiractha

indica. J. Natural Products.15: 201–209.

152. Kraus, W. (1995), The Neem Tree: Source of Unique Natural products for

Integrated pest Management, Medicine, Industry and other purposes (ed.

Schmutterer, H.), pp35-88.

153. Bray, D.H., Warhurst, D.C., Connolly, J.D., O’Neil, M.J. and Phillipson,

J.D. (1990). Plants as Source of Antimalaria drug. Part 7. Activity of some

Species of Meliaceae Plants and their constituent limonoids. Phytotherapy

Res, 4: 29-35.

cxiii

154. Rochanakij, S., Thebtaranonth, Y., Yeenjal, C.H. and Yuthavong, Y. (1985).

Nimbolide a constituent of A. indica inhibits Plasmodium falciparium in

culture. Southeast Asian J. Med. Public Health, 16: 66-72.

155. Joshi, S. P., Rojatkar, S.R. and Nagasampagi, B.A. (1998). Antimalaria

activity of Neem (A.indica), J. Med. Arom.Plant Sci. 20, 1000-1002.

156. Jones, I. W., Denholm, A. A., Ley, S. V., Lovell, H., Wood, A. and Sinden,

R. E. (1994). Sexual development of malaria parasites is inhibited in vitro by

the neem extract azadirachtin, and its semi-synthetic analoques. FEMS

Microbiol Lett. 120(3):267-273

157. Abatan, M. O., and Makinde, M. J. (1986). Screening Azadirachta indica in

vitro and in vivo, J. Ethnopharmacol. 17, 85–93.

158. Bickii, J., Nijifutie, N., Foyere, J. A., Basco, L. K. and Ringwald, P. (2000)

In vitro antimalarial activity of limonoids from Khaya grandifoliola C.D.C

(Meliaceae). J. Ethnopharmacol., 69, 27–33.

159. Weenen, H., Nkunya, M. H. H., Bray, D. H., Mwasumbi, L. B., Kinabo, L.

S., Kilimali, V. A. E. B. and Wijnberg, J. B. P. A. (1990). Antimalarial

compounds containing a, β-unsaturated carbonyl moiety from Tanzanian

medicinal plants. Planta Med, 56, 371–373.

160. MacKinnon S, Durst T, Arnason JT, Angerhofer C, Pezzuto J, Sanchez-

Vindas PE, Poveda LJ, Gbeassor M. (1997). Antimalarial activity of tropical

Meliaceae extracts and gedunin derivatives. J. Nat. Prod. 60(4): 336-41.

161. Mohammed A. Alshawsh, Ramzi A. Mothana, Hassan A. Al-shamahy, Salah

F. Alsllami and Ulrike Lindequist. (2007). Assessment of antimalarial activity

against Plasmodium falciparum and phytochemical screening of some Yemeni

cxiv

medicinal plants. . eCAM Advance Access published online eCAM,

doi:10.1093/ecam/nem148 . Oxford Journals.

162. Abatan, M. O., and Makinde, M. J. (1986). Screening Azadirachta indica

and Pisum sativum for possible antimalarial activities. J. Ethnopharmacol. 17,

85–93.

163. Obih, P.O. and Makinde, J.M. (1985). Effect of Azadirachta indica on

Plasmodium berghei berghei in mice. Afr. J. Med. Sci. 14: 51 – 54.

164. Boeke , SJ, Boersma , M.G, Alink , GM, vanLoon , JJA, vanHuis , A, Dicke,

M and Rietjens , IMCM (2004). Safety evaluation of neem (Azadirachta

indica) derived pesticides. Journal of Ethnopharmacology 94:25–41

165. Kanungo, D., (1996). In Neem (eds Randhawa and Parmar, B. S.). 2nd edn,

pp. 77–110.

166. Jacobson, M., (1995). In The Neem Tree: Source of Unique Natural

Products for Integrated Pest Management, Medicine, Industry and Other

Purposes (ed. Schmutterer, H.), pp. 484–495.

167. Despande, V. Y., Mendulkar, K. N. and Sadre, N. L., (1980). J. Postgrad.

Med. (Bombay), 26: 167–170.

168. Awasthy, K.S. (2001). Genotoxicity of a crude leaf extract of neem in male

germ cells of mice. Cytobios 106, 151–164.

169. Aladakatti, R. H., Nazeer Ahmed, R., Ahmed, M. and Ghosewami, M. G.

(2001). J. Basic Clin. Physiol. Pharmacol. 12, 69–76.

170. Kasutri, M., Ahmed, R. N., Pathan, K. M., Shaikh, P. D. and Manivannan,

B., (1997). Indian J. Physiol. Pharmacol. 41, 234–240.

cxv

171. Kalra BS, Chawla S, Gupta P and Valecha, N. (2006). Screening of

antimalarial drugs: An overview. Indian Journal of Pharmacology, 38(1): 5-

12.

172. Vivas L, Rattray L, Stewart LB, Robinson BL, Fugmann B, Haynes RK et

al. (2007). Antimalarial efficacy and drug interactions of the novel semi-

synthetic endoperoxide artemisone in vitro and in vivo. J Antimicrob

Chemotherap. 59 (4): 658-665.

173. http://www.malaria-ipca.com/artesunate.html

174. Neimark, H, Peters, W, Robinson, BL and Stewart, LB (2005). Phylogenetic

analysis and description of Eperythrozoon coccoides, proposal to transfer to

the genus Mycoplasma as Mycoplasma coccoides. Int J Syst Evol Microbiol

55: 1385-1391.

175. Peters, W. (1965). Competitive relationship between Eperythrozoon

coccoides and P. berghei in the mouse. Exp. Parasitol. 16(2): 158-166.

176. Attama, A. A., Nnamani, P.O. and Ezeora, N. D. (2001). Interaction between

artesunate and neem extract: A pharmacokinetic determination. Done in our

laboratory but unpublished.

177. Malik, A., Kumar, L, Kaushik, N, Sushant, KG and Kumar, P (2008). A New

Approach to Artemisinin and Trioxane Antimalarials. Latest Reviews. 6(5).

178. Munoz V, Souvain M, Bourdy G, et al. (2000). The search for natural

bioactive compounds through a multidisciplinary approach in Bolivia: Part II.

Antimalarial activity of some plants used by Mosetena Indians. J

Ethnopharmacol. pp 139-55.

cxvi

APPENDICES

Appendix I: Percentage yield obtained after extraction.

Plant Extract Yield 1(%) Yield 2(%) Yield 3(%)

Neem aqueous crude leaf 6.67 8.33 10

Pawpaw aqueous crude leaf 2.50 5.00 8.75

cxvii

Appendix II: Pictures of thin blood smear made for determination of the in vivo

schizontocidal activity of the drugs alone and in combination.

Day 0 Day 30 not cured

Day 4 Day 30 cured

Ring form

of P.

berghei

cxviii

Appendix III: Parasitemia level of each mouse used in the evaluation of the in vivo

schizontocidal activity of drugs alone and in combination.

Experimental condition

Parasitemia/mice (%)

1 2 3 4 5

Day 4 of drugs alone

Control 6.75 4.75 4.75 5.00 7.00

Neem 100 mg/kg 1.00 1.75 2.75 1.75 2.25

Neem 500 mg/kg NA 5.00 4.25 NA 1.56

Neem 1000 mg/kg 3.75 4.00 5.00 2.25 1.75

Pawpaw 50 mg/kg 3.00 3.75 NA 4.33 3.00

Pawpaw 100mg/kg 5.75 3.00 3.00 3.00 NA

Pawpaw 200 mg/kg 1.75 4.25 2.00 2.25 1.25

Artesunate 6 mg/kg 2.00 2.75 3.25 1.00 1.25

Artesunate 15 mg/kg 1.5 1.75 3.25 1.00 3.00

Artesunate 20 mg/kg 1.25 2.00 1.75 1.50 2.50

Day 7 drugs alone

Control 6.50 4.25 6.50 4.25 NA

Neem 100 mg/kg 4.50 4.25 2.50 2.25 NA

Neem 500 mg/kg 4.00 1.25 2.00 2.00 3.50

Neem 1000 mg/kg 1.25 1.25 1.00 2.75 1.25

Pawpaw 50 mg/kg 2.25 1.00 1.00 1.75 1.25

Pawpaw 100 mg/kg 3.00 2.25 3.25 1.50 4.25

cxix

Pawpaw 200 mg/kg 3.25 3.00 3.00 1.75 NA

Artesunate 6 mg/kg 2.75 4.75 2.50 3.50 NA

Artesunate 15 mg/kg 1.00 1.00 1.75 2.50 1.75

Artesunate 20 mg/kg 1.50 1.25 2.75 1.00 1.00

Day 4 drugs in combination

Control 3.75 5.25 1.25 6.00 7.75

Art 15 mg/kg + Neem 100 mg/kg 0.50 0.50 0.50 2.75 0.75

Art 15 mg/kg + Neem 500 mg/kg 1.75 0.25 0.75 0.50 0.75

Art 15 mg/kg + Neem 1000 mg/kg 0.00 0.25 0.00 0.25 0.25

Art 15 mg/kg + Pawpaw 50 mg/kg 1.00 1.00 1.25 0.75 0.50

Art 15 mg/kg + Pawpaw 100 mg/kg 2.50 0.50 0.50 1.50 0.75

Art 15 mg/kg + Pawpaw 200 mg/kg 2.75 1.71 2.36 1.25 NA

Day 7 drugs in combination

Control 7.00 7.00 22.75 NA NA

Art 15 mg/kg + Neem 100 mg/kg 2.00 2.03 2.00 1.88 6.00

Art 15 mg/kg + Neem 500 mg/kg 2.25 2.25 1.50 1.00 2.75

Art 15 mg/kg + Neem 1000 mg/kg 1.25 0.50 1.88 1.38 1.25

Art 15 mg/kg + Pawpaw 50 mg/kg 8.00 4.00 6.00 5.38 2.60

Art 15 mg/kg + Pawpaw 100 mg/kg 2.00 3.03 7.00 2.87 1.90

Art 15 mg/kg + Pawpaw 200 mg/kg 3.08 1.13 2.81 2.63 1.75

Appendix IV: Percentage reduction in parasitemia of drugs alone and in combination

Experimental condition Reduction in parasitemia (%)

cxx

1 2 3 4 5

Day 4 of drugs alone

Control 0.00 0.00 0.00 0.00 0.00

Neem 100 mg/kg 82.30 69.03 51.33 69.03 60.08

Neem 500 mg/kg NA 11.50 24.78 NA 72.48

Neem 1000 mg/kg 33.63 29.20 11.50 60.18 69.03

Pawpaw 50 mg/kg 46.90 33.63 NA 23.36 46.90

Pawpaw 100mg/kg -1.77 46.90 46.90 46.90 NA

Pawpaw 200 mg/kg 69.03 24.78 64.60 60.18 77.88

Artesunate 6 mg/kg 64.60 51.33 42.48 82.30 77.88

Artesunate 15 mg/kg 73.45 69.03 42.48 82.30 46.90

Artesunate 20 mg/kg 77.88 64.60 69.03 73.45 55.75

Day 7 drugs alone

Control 0.00 0.00 0.00 0.00 0.00

Neem 100 mg/kg 16.36 21.00 53.53 58.18 NA

Neem 500 mg/kg 25.65 76.77 62.83 62.83 34.14

Neem 1000 mg/kg 76.77 76.77 81.41 48.88 76.77

Pawpaw 50 mg/kg 58.18 81.41 81.41 67.47 76.77

Pawpaw 100 mg/kg 44.24 58.18 39.59 72.12 21.00

Pawpaw 200 mg/kg 39.59 44.24 44.24 67.47 NA

Artesunate 6 mg/kg 48.88 11.71 53.53 34.94 NA

Artesunate 15 mg/kg 81.41 81.41 67.47 53.53 67.47

Artesunate 20 mg/kg 72.12 76.77 48.88 81.41 81.41

Day 4 drugs in combination

cxxi

Control 0.00 0.00 0.00 0.00 0.00

Art 15 mg/kg + Neem 100 mg/kg 89.58 89.58 89.58 42.71 84.38

Art 15 mg/kg + Neem 500 mg/kg 63.54 94.79 84.38 89.58 84.38

Art 15 mg/kg + Neem 1000 mg/kg 100.00 94.79 100.00 94.79 94.79

Art 15 mg/kg + Pawpaw 50 mg/kg 79.17 79.17 73.96 84.38 89.58

Art 15 mg/kg + Pawpaw 100 mg/kg 47.92 89.58 89.58 68.75 84.38

Art 15 mg/kg + Pawpaw 200 mg/kg 42.71 64.38 50.83 73.96 NA

Day 7 drugs in combination

Control 0.00 0.00 0.00 NA NA

Art 15 mg/kg + Neem 100 mg/kg 83.67 83.45 83.67 84.69 51.02

Art 15 mg/kg + Neem 500 mg/kg 81.63 81.63 87.76 91.84 77.55

Art 15 mg/kg + Neem 1000 mg/kg 89.80 95.92 84.69 88.78 89.80

Art 15 mg/kg + Pawpaw 50 mg/kg 34.69 67.35 51.02 56.12 78.78

Art 15 mg/kg + Pawpaw 100 mg/kg 83.67 75.26 42.86 76.57 84.49

Art 15 mg/kg + Pawpaw 200 mg/kg 74.86 90.82 77.06 78.53 85.71

Appendix 5: Survival Time in days for each group

Experimental condition

Survival time (days)

1 2 3 4 5

Drugs alone

Control 14.00 17.00 20.00 23.00 25.00

Neem 100 mg/kg 6.00 6.00 9.00 12.00 25.00

cxxii

Neem 500 mg/kg 19.00 19.00 25.00 26.00 30.00

Neem 1000 mg/kg 12.00 16.00 19.00 21.00 23.00

Pawpaw 50 mg/kg 17.00 17.00 23.00 23.00 25.00

Pawpaw 100mg/kg 9.00 17.00 20.00 20.00 21.00

Pawpaw 200 mg/kg 9.00 17.00 19.00 20.00 23.00

Artesunate 6 mg/kg 17.00 19.00 19.00 25.00 25.00

Artesunate 15 mg/kg 19.00 19.00 21.00 23.00 27.00

Artesunate 20 mg/kg 20.00 21.00 23.00 25.00 25.00

Drugs in combination

Control 6.00 9.00 9.00 16.00 16.00

Art 15 mg/kg + Neem 100 mg/kg 21.00 30.00 30.00 30.00 30.00

Art 15 mg/kg + Neem 500 mg/kg 21.00 30.00 30.00 30.00 30.00

Art 15 mg/kg + Neem 1000 mg/kg 21.00 30.00 30.00 30.00 30.00

Art 15 mg/kg + Pawpaw 50 mg/kg 17.00 17.00 21.00 30.00 30.00

Art 15 mg/kg + Pawpaw 100 mg/kg 9.00 21.00 23.00 30.00 30.00

Art 15 mg/kg + Pawpaw 200 mg/kg 16.00 16.00 17.00 19.00 30.00

cxxiii

cxxiv


Recommended