+ All Categories
Home > Documents > BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte...

BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte...

Date post: 24-Apr-2020
Category:
Upload: others
View: 4 times
Download: 0 times
Share this document with a friend
63
KAUNAS UNIVERSITY OF MEDICINE Faculty of Pharmacy UNIVERSITY OF MONTPELLIER 1 Faculty of Pharmacy Toma KEŽUTYTĖ BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE DETERMINATION OF TWO ANTIMALARIAL THIAZOLIUM COMPOUNDS AND ITS APPLICATION TO A PHARMACOKINETIC STUDY IN RAT Master’s thesis CONFIDENTIAL Supervisors: Prof. Dr. Vitalis BRIEDIS Prof. Dr. Francoise BRESSOLLE Kaunas Montpellier 2007
Transcript
Page 1: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

KAUNAS UNIVERSITY OF MEDICINE Faculty of Pharmacy

UNIVERSITY OF MONTPELLIER 1 Faculty of Pharmacy

Toma KEŽUTYTĖ

BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE DETERMINATION OF TWO

ANTIMALARIAL THIAZOLIUM COMPOUNDS AND ITS APPLICATION TO A PHARMACOKINETIC STUDY IN RAT

Master’s thesis

CONFIDENTIAL

Supervisors: Prof. Dr. Vitalis BRIEDIS

Prof. Dr. Francoise BRESSOLLE

Kaunas Montpellier

2007

Page 2: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

TABLE OF CONTENTS Introduction .......................................................................................................................................... 4 Part 1 – Literature review ................................................................................................................... 5 I. The Problem of Malaria in the World................................................................................................. 5 II. Brief Presentation of the Life Cycle of Plasmodium ........................................................................ 7 2.1. Asexual phase of development ......................................................................................... 7 2.1.1. Exoerythrocytic cycle ............................................................................................... 7 2.1.2. Intraerythrocytic cycle .............................................................................................. 8 2.2. Sexual phase of development ........................................................................................... 8 III. Currently Available Antimalarial Drugs.......................................................................................... 9 3.1. Quinolines......................................................................................................................... 9 3.1.1. Chemical structures................................................................................................... 9 3.1.2. Effects on the stages of parasite’s life cycle and mode of action.............................. 9 3.1.3. Mechanism of resistance ........................................................................................... 10 3.2. Antifolates......................................................................................................................... 11 3.2.1. Chemical structures................................................................................................... 11 3.2.2. Effects on the stages of parasite’s life cycle and mode of action.............................. 11 3.2.3. Mechanism of resistance ........................................................................................... 11 3.3. Atovaquone....................................................................................................................... 12 3.3.1. Chemical structure .................................................................................................... 12 3.3.2. Effects on the stages of parasite’s life cycle and mode of action.............................. 12 3.3.3. Mechanism of resistance ........................................................................................... 12 3.4. Artemisinins...................................................................................................................... 12 3.4.1. Chemical structures................................................................................................... 13 3.4.2. Effects on the stages of parasite’s life cycle and mode of action.............................. 13 3.4.3. Mechanism of resistance ........................................................................................... 14 3.5. Antibiotics......................................................................................................................... 14 3.5.1. Chemical structures................................................................................................... 14 3.5.2. Effects on the stages of parasite’s life cycle and mode of action.............................. 14 IV. Combinations of Antimalarial Drugs............................................................................................... 15 4.1. Artemisinin-based combination therapies (ACTs) ........................................................... 15 4.2. Non-artemisinin based combination therapies (non-ACTs) ............................................. 16 4.3. Synergistically acting combinations of antimalarial drugs............................................... 16 V. New Targets in Plasmodium for the Development of New Antimalarial Drugs ............................. 17 VI. Potential Inhibitors of Phospholipid Metabolism in Plamodium..................................................... 22 6.1. De novo synthesis of phosphatidylcholine in Plasmodium .............................................. 22 6.2. Potential inhibitors of PL metabolism .............................................................................. 23 6.3. Quaternary ammonium compounds exhibit dual mode of action against Plasmodium ... 24 6.4. Structure-activity relationship of quaternary ammonium compounds ............................. 25 6.5. Monothiazolium salts T1 and T2 ...................................................................................... 25 Part 2 – Experimental Part ................................................................................................................. 26 I. Different Steps in Bioanalytical Method Validation .......................................................................... 26 1.1. Different types of bioanalytical method validation .......................................................... 26 1.2. Methodology..................................................................................................................... 27 1.2.1. Relationship between concentration and response.................................................... 27 1.2.2. Specificity / selectivity.............................................................................................. 28 1.2.3. Accuracy and precision............................................................................................. 28 1.2.4. Lower limit of quantitation (LLOQ) ......................................................................... 29 1.2.5. Extraction recovery ................................................................................................... 29 1.2.6. Stability ..................................................................................................................... 30 1.2.7. Robustness / ruggedness ........................................................................................... 31

2

Page 3: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

1.2.8. Matrix effects ............................................................................................................ 31 1.3. Application of validated bioanalytical method to routine drug analysis .......................... 32 II. Assay for Simultaneous Determination of Two Antimalarial Monothiazolium Compounds in Biological Matrices by Liquid Chromatography – Electrospray Mass Spectrometry ........................... 33 2.1. Materials and methods ...................................................................................................... 33 2.1.1. Chemicals and reagents............................................................................................. 33 2.1.2. Equipment ................................................................................................................. 34 2.1.3. Liquid chromatography-mass spectrometry conditions ............................................ 36 2.1.4. Working standards .................................................................................................... 37 2.1.5. Preparation of calibration standards and quality control (QC) samples ................... 38 2.1.6. Sample preparation procedure................................................................................... 38 2.1.7. Data analysis ............................................................................................................. 39 2.1.8. Validation procedure................................................................................................. 39 2.1.9. Stability study ........................................................................................................... 40 2.1.10. Ion suppression / enhancement study...................................................................... 40 2.1.11. Pharmacokinetic study of T2 in rat ......................................................................... 40 2.2. Results of full validation in human plasma and whole blood........................................... 42 2.2.1. Drug / response relationship ..................................................................................... 42 2.2.2. Retention times and specificity ................................................................................. 44 2.2.3. Precision, accuracy, extraction efficiency and LLOQ .............................................. 47 2.2.4. Stability ..................................................................................................................... 49 2.2.5 Ion suppression / enhancement study......................................................................... 51 2.3. Results of partial validation in rat plasma and RBCs ....................................................... 51 2.3.1. Drug / response relationship ..................................................................................... 51 2.3.2. Retention times and specificity ................................................................................. 52 2.3.3. Precision, accuracy, extraction efficiency and LLOQ .............................................. 54 2.3.4. Stability ..................................................................................................................... 54 2.3.5 Ion suppression / enhancement study......................................................................... 54

2.4. Results of a pharmacokinetic study carried out after intravenous administration of T2 in rat ........................................................................................................................................... 54

Conclusion ............................................................................................................................................ 56 References ............................................................................................................................................. 57 Acknowledgements .............................................................................................................................. 63

3

Page 4: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

INTRODUCTION

Malaria remains one of the largest global health-care problems of the 21st century. Currently it

affects more than 40% of the world's population in tropical areas throughout sub-Saharan Africa,

South Africa, Southeast Asia, the Pacific Islands, India, and Central and South America.

The disaster of malaria is mainly due to the emergence of multi-drug resistant Plasmodium

strains. The current limited antimalarial drug repertoire, resistance of mosquitoes to various

insecticides and absence of efficient malaria vaccine indicate that new antimalarial compounds are

desperately needed.

In the first part of my work, the problem of malaria in the world, the complex life cycle of

Plasmodium spp., currently available antimalarial drugs (quinines, antifolates, atovaquone,

artemisinins and antibiotics) and their combinations are presented. The main attention is focused on

the new targets of Plasmodium and on the development of new potential antimalarial drugs. Among

them, mono- and bis-quaternary ammonium salts, mimicking the structure of choline and

inhibiting de novo phosphatidylcholine biosynthesis in the parasite have been developped. In

the Clinical Pharmacokinetic Laboratory (Faculty of Pharmacy, University Montpellier 1,

Montpellier, France), these compounds are being under investigation. I had an honour to contribute

to this research. My work has been focused on the development of monothiazolium compounds.

The goal of this work is to validate an analytical method to simultaneously quantify two

monothiazolium compounds (T1+ and T2+) in biological matrices and then to carry out

pharmacokinetic study in rat. In the experimental part of this work, the methodology of

bioanalytical method validation is documented. Then, the materials, methods and results of a full

validation in human plasma and whole blood and of a partial validation in rat plasma and red blood

cells by liquid chromatography-electrospray mass spectrometry (LC/ESI-MS) are presented. The

application of this validated bioanalytical method to a pharmacokinetic study carried out in rat is

highlighted.

The main tasks of this work are as follows:

• To show the problem of emergence of multi-drug resistant Plasmodium strains;

• To demonstrate potential Plasmodium targets for the development of new antimalarials;

• To present quaternary ammonium compounds as highly potential novel antimalarials;

• To validate bioanalytical methods in human plasma and whole blood as well as in rat

plasma and red blood cells for the simultaneous quantitation of monothiazolium compounds using

LC/ESI-MS;

• To use these validated bioanalytical methods to quantify the T2+ compound in biological

samples drawn during a pharmacokinetic study carried out in rat.

4

Page 5: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

PART 1 – LITERATURE REVIEW

I. THE PROBLEM OF MALARIA IN THE WORLD

Malaria is the world’s most important parasitic infection and one of the major causes of

morbidity and mortality in the third-world countries. There is an estimation of 500 million

infections (90% of them occurring in Africa) and 2.7 million deaths (75% of these deaths arising to

African children) from malaria each year – a death from malaria every 30 seconds [28, 17, 22, 26].

Malaria is a haematoprotozoan parasitic infection, caused by unicellular, apicomplexan

parasites of the genus Plasmodium and transmitted to human by female Anopheles mosquito. Four

species of Plasmodia are infectious to human and cause distinct disease patterns: Plasmodium

falciparum (malaria tropica), P.ovale (malaria quartana), P.vivax (malaria tertiana) and P.malariae

(malaria tertiana).

P.ovale and P.malariae are not lethal and are rare, most frequently found in parts of Africa

and Papua New Guinea. P.vivax and P.falciparum are the two most widespread species in the

world; P.falciparum is endemic in Africa, South and East Asia, South America, the Caribbean and

the Middle East, and is uniquely responsible for mortality and severe disease [28].

The common symptoms of uncomplicated malaria are headache, fatigue, abdominal

discomfort, joint and muscle aches, lassitude followed by fever, chills, perspiration, anorexia,

vomiting and worsening malaise. If not treated within few hours, uncomplicated malaria progresses

to coma (cerebral malaria), metabolic acidosis, severe anaemia, hypoglycaemia, acute renal failure

or acute pulmonary oedema. These are the common characteristics of severe disease, which is

almost always fatal, thus must be treated quickly [106].

The nature of the clinical disease depends on the pattern and intensity of the malaria

transmission in the area of residence. In areas with high malaria transmission rates (sub-Saharan

Africa, parts of Oceania) partial immunity is acquired early in childhood, therefore adolescents and

adults rarely suffer clinical disease and over 75% of deaths occur to children of less than 5 years

old. Reduced immunity and severe anaemia for pregnant women, exposed to malaria infection,

frequently result in low birthweight followed by roughly 25% of all neonatal mortality [112]. In

areas of unstable malaria (south and east of Asia, Latin America), where fluctuation of inoculation

rates over seasons and years retards the acquisition of immunity, people of all ages suffer from

clinical malaria [31, 106].

Malaria continues to affect public health, economic growth and livelihood in malaria endemic

areas of the world. Political instability, chronic regional economic difficulty, environmental impact,

5

Page 6: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

mosquito resistance to insecticides, Plasmodium resistance to established antimalarial drugs and

insufficient investment in the discovery of new drugs [112] have shattered the hope for rapid

malaria eradication and given rise to a very serious situation.

The increasing resistance of malaria parasites to the currently available antimalarial drugs,

absence of efficient malaria vaccine stress the importance of developing novel drugs and indicates

that drug development efforts should be geared towards obtaining compounds structurally unrelated

to existing antimalarial agents and with new independent mechanisms of action.

The completed genome sequence (in 2002) of P.falciparum

(http://www.plasmodb.org/plasmo/home.jsp) and the application of methods of modern drug design

promise to bring significant development in the fight against this disease.

6

Page 7: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

II. BRIEF PRESENTATION OF THE LIFE CYCLE OF PLASMODIUM

Plasmodium has a complex life cycle, involving both vertebrate and invertebrate hosts (Figure

1.1). The development of malaria parasites includes an asexual cycle in human host and a sexual

cycle in Anopheles mosquito.

Figure 1.1: The life cycle of malaria parasites in the human host and Anopheles

mosquito vector [33].

2.1. Asexual phase of development

2.1.1. Exoerythrocytic cycle:

7

Page 8: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

During a blood meal by female Anopheles mosquito, a few dozen of malaria sporozoites are

injected to human host subcutaneous tissues. Sporozoites enter the nearby blood vessel and reach

the liver. Once in the liver, parasite undergoes asymptomatic multiplication over a period of several

weeks. P.vivax and P.ovale can transform to hypnozoites (latent phase in the hepatocytes) that may

emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which

subsequently divides into several schizonts. The hepatic schizonts burst releasing thousands of

pathogenic merozoites into the bloodstream. This initiates intraerythrocytic phase, that gives rise to

malaria symptoms, morbidity and mortality [28, 11, 72].

2.1.2. Intraerythrocytic cycle:

Pathogenic merozoites gain entry into erythrocytes by a process that leaves the intracellular

parasite enclosed within parasitophorous vacuolar membrane (PVM). The parasite matures in the

erythrocyte through ring (0-24 h post-invasion), trophozoite (24-36 h post-invasion) and schizont

(36-48 h post-invasion) stages. At the end of 48 hours (72 hours in the case of P.malariae), the

schizont bursts and 8-32 merozoites are released, allowing the intraerythrocytic cycle to begin

again. A small percentage of the merozoites differentiate into sexual forms termed male and female

gametocytes. These stages are responsible for transmission through the mosquito [56].

2.2. Sexual phase of development

Gametocytes undergo sexual reproduction in the mosquito, when they are ingested through a

mosquito blood meal (3µl) that must contain both female and male gametocytes. Gamete fusion

(creating a zygote) and thus meiosis takes place in the mosquito’s mid gut. This brief diploid stage

in an otherwise haploid life cycle allows for sexual recombination of genetic material, including the

chromosomal genes responsible for most drug resistance. Within the mosquito mid gut the zygote

matures into an oocyst, which in turn releases sporozoites that then migrate to the mosquito salivary

glands, completing the life cycle [42].

8

Page 9: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

III. CURRENTLY AVAILABLE ANTIMALARIAL DRUGS

In this chapter the most extensively used antimalarial drugs (quinolines, antifolate drugs,

atovaquone, artemisinins, and antibiotics) are discussed, emphasizing their chemical structures,

mechanisms of action and resistance development.

3.1. Quinolines

This family of drugs includes (i) 4-amino quinolines: chloroquine (CQ) and amodiaquine

(Mannich base 4), (ii) aryl aminoalcohols: quinine, mefloquine, halofantrine and lumefantrine, and

(iii) 8-amino quinolines: primaquine.

3.1.1. Chemical structures:

Chloroquine Amodiaquine Quinine Mefloquine

Halofantrine Lumefantrine Primaquine

3.1.2. Effects on the stages of parasite’s life cycle and mode of action:

Quinolines are blood schizontocidal antimalarials [26], but chloroquine also acts against

young gametocytes. Primaquine is effective against intrahepatic stages of malaria parasites [106],

therefore it eradicates the liver hypnozoites of P.ovale and P.vivax [112]. Moreover, primaquine is

the only antimalarial drug known to act on mature infective gametocyte [28, 106].

The basic nature of quinolines allows them to be concentrated in the acidic food vacuole in

their membrane-impermeable protonated form [108].

Chloroquine and other quinoline derivatives act by joining with undimerized

ferriprotoporphyrin (FP) IX, which is released during the haemoglobin degradation process in the

9

Page 10: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

food vacuole [36; 37]. Thereby, chloroquine antagonises the polymerisation of FP IX into crystal

malaria pigment hemozoin [112, 35, 100]. This leads to intra-Plasmodium accumulation of highly

toxic chloroquine–FP IX complexes that are incorporated in the growing polymer hemozoin

resulting in the termination of chain extension [26]. Moreover, there are some evidences that

chloroquine inhibits glutathione that is responsible for the degradation of free FP IX [75].

There are available evidences that mefloquine and quinine inhibit the uptake of haemoglobin

from the host cell [108].

Primaquine’s mechanism of action is unknown [28], but treatment with primaquine causes

swelling and thickening of the mitochondria of intrahepatic parasites, suggesting that this drug

interferes with mitochondrial function [37].

3.1.3. Mechanism of resistance:

Resistance to chloroquine:

1st hypothesis: reduced concentration of chloroquine in the parasite vacuole can be attributed

to (i) an efflux of the drug from the vacuole due to an active verapamil-sensitive efflux pump (P-

glycoprotein 1 encoded by pfmdr1 gene) that extrudes chloroquine from the food vacuole (Figure

1.2, A) and/or (ii) a change in the structure of the transporter, importing the drug into the food

vacuole, due to mutations of the pfcrt gene encoding for this transporter (PfCRT) (Figure 1.2, B)

[63, 28, 115, 46].

A B C

Figure 1.2: Alternative models of the chloroquine (CQ) resistance [56].

2nd hypothesis: reduced passive diffusion of chloroquine into the vacuole due to elevation of

the vacuolar pH: this arises from a weakened vacuolar H+ pump, increased H+ leakage or perhaps

from an impaired Cl- conductance (Figure 1.2, C) [86, 56].

Resistance to mefloquine and other aryl-amino-alcohols results from amplifications in

Pfmdr1, which encodes an energy demanding P-glycoprotein 1 pump [106, 28, 37, 115].

10

Page 11: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Chloroquine resistance appeares to be associated with mefloquine hypersensitivity and vice versa

[115].

3.2. Antifolates

The most commonly used antifolates are pyrimethamine (2,4-diaminopyrimidine), proguanil,

chlorproguanil, and the sulfa-drugs: sulfadoxine and dapsone.

3.2.1. Chemical structures:

R Pyrimethamine Proguanil (R=H) Sulfadoxine Dapsone Chlorproguanil (R=Cl)

3.2.2. Effects on the stages of parasite’s life cycle and mode of action:

Antifolates are active against schizont stages within erythrocytes and hepatocytes [26].

Proguanil also has sporontocidal activity, rendering the gametocytes non-infective to the mosquito

[106].

Antifolates are antimetabolites, inhibiting the folate pathway and subsequently pyrimidine

synthesis. Sulfadoxine and dapsone, analogues of p-aminobenzoic acid, inhibit dihydropteroate

synthase, DHPS (a bifunctional enzyme in Plasmodia coupled with 2-amino-4-hydroxy-6-

hydroxymethyl-dihydropteridine pyrophosphokinase PPPK). Pyrimethamine is a selective,

competitive inhibitor of dihydrofolate redutase (DHFR) domain in bifunctional PfDHFR-TS

(thymidylate synthetase), a key enzyme in the redox cycle for production of tetrahydrofolate [14,

82]. Proguanil and chlorproguanil are metabolised by human CYP2C19 and CYP3A4 to the active

metabolites cycloguanil and chlorcycloguanil, respectively, which further inhibit parasite DHFR

[108, 112, 68].

3.2.3. Mechanism of resistance:

Slow elimination (e.g. t1/2 of pyrimethamine is roughly 100 hours [112]) and monotherapy of

antifolates have facilitated the development of resistance [113, 26]. Point mutations (codons: 51, 59,

108, 164) in the Pfdhfr gene and point mutations (codons: 436, 437, 540, 581, 613) in the Pfdhps

gene give moderate to high level of resistance to antifolates [63, 42].

11

Page 12: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

3.3. Atovaquone

Atovaquone is used orally in fixed, synergistically acting combination with proguanil

(Malarone®) to treat and prevent (especially among travellers) chloroquine-resistant uncomplicated

falciparum malaria, but high cost of atovaquone limits the usage of Malarone® in Africa.

3.3.1. Chemical structure:

Atovaquone (hydroxynaphthoquinone)

3.3.2. Effects on the stages of parasite’s life cycle and mode of action:

Atovaquone is a blood schizontocidal antimalarial drug also inhibiting pre-erythrocytic

development of Plasmodium species in the liver and oocyst development in the mosquito [106].

Atovaquone mimics the natural substrate ubiquinone (CoQ) and selectively inhibitis electron

transport in plasmodial mitochondria respirotory chain at the cytochrome bc1 oxidoreductase

(complex III) [28]. This results in the collapse of mitochondrial membrane potential [63, 93] and

inhibition of dihydroorotate dehydrogenase (DHOD), which is required in the biosynthesis of

pyrimidines [8].

In the combination with atovaquone, proguanil rather than its metabolite (cycloguanil) acts as

a biguanide and enhances the effect of atovaquone on the collapse of mitochondrial membrane

potential [98].

3.3.3. Mechanism of resistance:

In vitro and in vivo resistance of P.falciparum to atovaquone or atovaquone-proguanil has

been associated to two point mutations in the parasite cytochrome b gene (Tyr268Ser and

Tyr268Asn) [44].

3.4. Artemisinins

Natural artemisinin (quinghaosu) was first isolated in 1971 in China. It is a sesquiterpene

lactone, extracted from the dry leaves of Artemisia annua - the sweet wormwood tree [114, 49]. In

addition to artemisinin, the semisynthetic artemisinin derivatives (modifications made at C10

12

Page 13: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

position) – dihydroartemisinin, artemether, arteether (artemotil) and artesunate are used for the

treatment of malaria [108, 114].

The artemisinins are remarkable for their (i) high potency (they kill all human Plasmodium

species and are effective against multidrug-resistant P.falciparum strains), (ii) high therapeutic

index and (iii) rapid reduction in gametocyte carriage, and possess such features as (i) absence of

clinically important resistance, (ii) relative safety (few side effects in extensive clinical trials) and

(iii) good tolerability [112, 11, 116, 66].

3.4.1. Chemical structures:

Artemisinin Dihydroartemisinin Artemether Arteether Artesunate

3.4.2. Effects on the stages of parasite’s life cycle and mode of action:

Artemisinins have a broad spectrum of rapid activity against all parasite phases within

erythrocytes, in particular younger ring forms [26]. Furthermore, artemisinins are the most potent

gametocytocidal drugs destroying immature gametocytes [106].

Artemisinin derivatives are metabolised to the main bioactive compound, dihydroartemisinin

[108, 112, 114]. Artemisinins contain an endoperoxide trioxane moiety that undergoes (i) reductive

scission: Fe2+ catalyzed (by ferrous haem or exogenous free iron) cleavage into O-centred and

subsequently C-centred free radicals and (ii) peroxide ring-opening by protonation (H+) or complex

formation with Fe2+, resulting in an open hydroperoxide or metal peroxide. Reductive scission leads

to alkylation of plasmodial macromolecules such as Hb-derived haem or parasite proteins (Figure

1.3) and free-radical damage of P.falciparum. Ring-opening of peroxide bond results in

hydroxylation and oxidation of biomolecules [83, 41].

Figure 1.3: Hypothetical mechanism for the radical alkylation of haem by artemisinin

[108].

13

Page 14: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Recently it has been found that artemisinins accumulate in the cytosol of the parasite and

selectively, irreversibly inhibit an essential SERCA (sarco/endoplasmic reticulum Ca2+-ATPase)

orthologue of P.falciparum (PfATPase6) [106, 78, 114, 28].

3.4.3. Mechanism of resistance:

Although there has been no evidence for clinically relevant in vivo artemisinin resistance,

recently described emerging in vitro resistance to artemisinins in certain areas (French Guiana)

correlates with mutations in the SERCA-like sequence PfATP6 [115].

3.5. Antibiotics

The most commonly used are (i) tetracyclines: tetracycline, doxycycline and (ii)

lincosamides: clindamycin.

3.5.1. Chemical structures:

Tetracycline Doxycycline Clindamycin

3.5.2. Effects on the stages of parasite’s life cycle and mode of action:

Tetracycline and doxycycline are active against the asexual stages of all Plasmodium species

as well as against primary intrahepatic stages of P.falciparum. Clindamycin is a blood

schizontocide.

Antibiotics act through inhibition of protein synthesis in the parasite mitochondrion

(tetracyclines) and inside the apicoplast (tetracyclines, lincosamides). Inhibition of self-replication

of the apicoplast leads to the death of the parasite in the second replication cycle (‘delayed death’

phenomenon) [108, 89]. Thus, the action of antibiotics is slow and they are mainly combined with

quinine or other fast acting antimalarials [26, 64].

Tetracycline also depresses the activity of dihydroorototate dehydrogenase of the pyrimidine

pathway in P.falciparum [85].

14

Page 15: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

IV. COMBINATIONS OF ANTIMALARIAL DRUGS

As the resistance has been documented for Plasmodium falciparum (observed resistance to

amodiaquine, chloroquine, mefloquine, quinine, and sulfadoxine-pyrimethamine), Plasmodium

vivax (documented resistance to sulfadoxine-pyrimethamine and chloroquine) and, recently,

Plasmodium malariae (observed resistance to chloroquine), new approaches for the treatment of

malaria have been suggested [106]. The use of drug combinations is the current most important

strategy to fight developing resistance of Plasmodium to monotherapies and to improve the efficacy

of malaria treatment.

Artemisinin derivatives are the only antimalarial drugs to which clinically significant

resistance has not yet developed [11], therefore artemisinin-based combination therapy (ACT) has

been strongly suggested as the first-line treatment strategy of uncomplicated malaria to both

children and adults [106, 112, 114].

In this chapter different combinations of antimalarial drugs, used to treat uncomplicated

falciparum malaria are highlighted.

4.1. Artemisinin-based combination therapies (ACTs)

ACT comprises two or more blood schizontocidal antimalarial drugs that have different

mechanisms of action and therefore independent targets in Plasmodium [106].

Combinations with artemisinin derivatives have a strong impact on (i) rapid reduction of the

parasite biomass (by roughly 10000-fold per asexual cycle), that makes ACTs highly effective and

delays the possibility of the remaining parasite population to develop mutations to the second

partner drug [112, 111, 114], (ii) reduction of transmission of both drug-sensitive and drug-resistant

parasites as artemisinin derivatives have a gametocytocidal action [28].

ACTs present several drawbacks: (i) ACTs are not recommended for treatment of malaria

during the first three months of pregnancy because of embryolethality and dysmorphogenesis

observed in pregnant animals after artemisinin administration [67]; (ii) the use of ACT is

constrained due to high cost and limited availability in many regions of Africa.

Currently recommended ACTs are as follows [106]:

• Artemether-lumefantrine (Riamet®/Coartem®): the only fixed combination having the

advantage that one drug cannot be taken without the other [7]. It is not the case for other

combinations given below:

• Artesunate + amodiaquine (in areas where the cure rate of amodiaquine monotherapy is

greater than 80%);

15

Page 16: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

• Artesunate + mefloquine;

• Artesunate + sulfadoxine-pyrimethamine (in areas where the cure rate of sulfadoxine-

pyrimethamine is greater than 80%).

4.2. Non-artemisinin based combination therapies (non-ACTs)

Non-ACTs include amodiaquine + sulfadoxine-pyrimethamine (AQ+SP) and chloroquine +

sulfadoxine-pyrimethamine (CQ+SP). CQ+SP is no longer recommended for malaria treatment due

to high prevalence of CQ resistance. AQ+SP is an interim option in some Africa areas (especially

West Africa) that are unable to immediately use ACTs and where the efficacy of both compounds

remains high [69, 106, 50].

4.3. Synergistically acting combinations of antimalarial drugs

These combinations mainly include synergistic antifolate drug combinations, such as (i)

pyrimethamine-sulfadoxine (Fansidar®), (ii) chlorproguanil-dapsone (LapDap®) and (iii)

proguanil-atovaquone (Malarone®).

The parasite has already developed resistance to pyrimethamine-sulfadoxine. Both in vitro

and in vivo studies indicate that, despite the identical mechanism of action, chlorproguanil-dapsone

remains active against the Pfdhfr and Pfdhps genotypes in Africa that cause pyrimethamine-

sulfadoxine failure [42].

To further counteract the development and spreading of resistance, a fixed triple combination

of chlorproguanil-dapsone-artesunate is now being in phase III of clinical development [26, 17,

112].

16

Page 17: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

V. NEW TARGETS IN PLASMODIUM FOR THE DEVELOPMENT OF

NEW ANTIMALARIAL DRUGS

Due to emergence and spread of multidrug-resistant malaria parasites and unsuccessful efforts

to produce an effective vaccine, there is an urgent need for the identification and validation of new

drug targets in Plasmodium and for the development of novel antimalarial drugs.

The re-design of existing antimalarial drugs might result in cross-resistance within a short

period of time, thus effective and safe antimalarials with new mechanisms of action against new

targets are desperately needed [76, 84].

Metabolic pathways in Plasmodium, in particular those that do not occur or that are quite

different in human host, specific proteins that are unique and critical for cellular growth and

survival of the parasite provide important novel antimalarial drug targets [43, 26]. Figure 1.4

represents Plasmodium organelles and metabolic pathways as potential targets for antimalarial

compounds in development.

Figure 1.4: Potential drug targets in Plasmodium [14].

17

Page 18: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Ideally novel antimalarial drug should have: (i) high efficacy against multi-drug resistant

Plasmodium strains, (ii) low toxicity especially in children and pregnant women, (iii) low risk of

emergence of resistance. Curative regimens should be short: 1-3 days in length. In addition, new

antimalarials should be dosed orally and be effective with single-daily dosing. Furthermore, they

should be very inexpensive and affordable in the developing world [17, 93, 65].

The parasite’s miscellaneous biology at each stage and the ability to select drug-resistant

strains present a great challenge for new antimalarial drug development [28]. Therefore the

knowledge about parasite-specific organelles, metabolic pathways and membrane transport

mechanisms, the detailed structural and functional analysis of parasite enzymes, and their

comparison with their isofunctional counterparts in the host are indispensable features for rational

antimalarial drug design [13].

Brief description of new targets and lead inhibitors of specific metabolic pathways in the

parasite is provided in Table 1.1.

18

Page 19: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Table 1.1: New targets in Plasmodium Organelles

in Plasmodium

Pathway Potential targets Lead inhibitors in development Details

β-ketoacyl-acyl-carrier protein synthase (FabH)

Thiolactomycin

Thiolactomycin is a fungal secondary metabolite, selectively inhibiting FabH [14].

Type II fatty acid biosynthesis (FAS-II) (type I fatty acid synthase (FAS-I) exists in human)

Trans-2-enoyl-acyl-carrier protein reductase (FabI)

Triclosan [5-chloro-2-(2,4-dichlorophenoxy) phenol]

Triclosan is effective in vitro against CQ-sensitive and CQ-resistant strains, it invokes rapid death in parasites, but it has low bioavailability and share mechanisms of drug resistance with mefloquine [89, 77, 14].

Protein prenylation

Farnesyl transferase Peptidomimetics, biphenyl derivative FTI-2153, imidazole derivative FTI-2217, benzophenone based compounds (Schl-4116)

Farnesyl transferase is a heterodimeric zinc protein that catalyses the transfer of a farnesyl residue from farnesyl pyrophosphate to a cysteine side chain near the carboxy terminus of a number of G-proteins [39, 110, 55].

Isoprenoid synthesis via non-mevalonate pathway (mevalonate pathway is present in human)

1-deoxy-D-xylulose 5-phosphate (DOXP) reductoisomerase

Fosmidomycin (3-formylhydroxyamino-propylphosphonic acid mono-sodium salt), FR-900098 (N-acetyl homologue of fosmidomycin)

Fosmidomycin is a natural antibiotic isolated from Streptomyces lavendulae and currently being in a clinical development for the treatment of uncomplicated malaria. Advantages: low toxicity, activity against multiresistant strains. Limitations: short plasma half-life (2,5 h), moderate bioavailability, high recrudescence rate in monotherapy (evaluated synergistic combination with clindamycin). FR900098 is twice as effective as fosmidomycin [43, 107, 76, 65, 109].

Api

copl

ast

(chl

orop

last

-like

org

alle

with

a c

ircu

lar D

NA

of 3

5 kb

) ne

Shikimate pathway (absent in human)

Chorismate synthase; 5-enolpyruvyl shikimate 3-phosphate (EPSP) synthase

Glyphosate derivatives The shikimate pathway provides p-amino benzoic acid precursor (chorismate) for folate, ubiquinone, tryptophan, phenylalanine and tyrosine biosynthesis. Glyphosate is a well characterised herbicide that inhibits EPSP synthase [84, 38, 92].

Mio

-t

nd

cho

rion Electron

transport and de novo pyrimidine synthesis

Dihydroorotate dehydrogenase (DHODase); Ubiquinone (CoQ8)

2-amino-3-chloro-1,4-naphthoquinone, 2-nitro-3-methyl-benzamide, 2-phenyl quinoline-4-carboxylic acid derivatives, pyridones

Malarial DHODase active site and CoQ homolog (CoQ8) are different from the host. DHODase uses CoQ8 as an electron acceptor and catalyzes the reaction from dihydroorotate to orotate in de novo pyrimidine synthesis [10, 47, 20, 9, 53]. Pyridones show no cross-resistance with atovaquone [17].

19

Page 20: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Table 1.1 (continued): New targets in Plasmodium

Organelles in

Plasmodium Pathway Potential targets Lead inhibitors in development Details

Aspartic proteases (plasmepsins I and II)

Allophenylnorstatine (KNI-727), diphenylurea derivatives (WR268961), Ro 42-1118

Haemoglobin (Hb) hydrolysis

Cysteine proteases (falcipain 2, 2’ and 3)

Peptidyl fluoromethyl ketones, peptidyl vinyl sulfones, chalcones, phenothiazines, leupeptin

Undegraded Hb blocks parasite development. Peptidic inhibitors of cysteine proteases have some limitations: (i) susceptibility to hydrolysis by host proteases, (ii) low selectivity, (iii) irreversible mode of action. Synergistic effects are observed in vitro and in vivo when aspartic and cysteine protease inhibitors are combined [39, 108, 97].

Haem polymerization

Ferriprotoporphyrin IX (FP)

Triarylmethanol Ro 06-9075, benzophenone Ro 22-8014, N-desbutyl derivative of halofantrine, dihydroacridinedione WR243251; tafenoquine, isoquine, pyronaridine, diamidines (DB 289)

These compounds bound to FP IX and inhibit the formation of nontoxic hemozoin. Cardiotoxicity of halofantrine is eliminated in its N-desbutyl derivative. Pyronaridine shows synergistic effect in combination with artesunate. Tafenoquine also eliminated hypnozoites [108]. Hepatotoxicity of amodiaquine is eliminated in its analogue isoquine [17].

Food

vac

uole

Free radical generation, oxidative stress

Unknown

Artemisinin derivatives and peroxides: 1,2,4,5-tetraoxanes, oxazines, trioxaquines, Yingzahaosu A, artelinic acid

Carbon-centred radical formation is critical for the activity of these compounds. Artelinic acid is more stable than artesunate and is highly soluble in water; Yingzahaosu A is isolated from traditional Chinese medicinal plant Artabotrys uncinatus [108].

Membrane transport

Hexose transporter HT1, nucleoside transporter NT1, P-type Ca2+-ATPases, V-type H+-ATPases, H+-pumping pyrophosphatases

O-3 hexose derivatives inhibit PfHT1; cyclopiazonic acid inhibits endoplasmic reticulum P-type ATPases; Bafilomycin A1 inhibits V-type ATPases; aminomethylenediphosphonate inhibits H+-pumping pyrophosphatases

Inhibition of PfHT1 leads to ATP level decline and the loss of intracellular pH control in the parasite. PfNT1 transports purines from the erythrocyte to the parasite. Inhibition of PfNT1 or exploitation of PfNT1 ability to transport cytotoxic 3’-deoxynucleoside analogues (that are poor substrates for mammalian transporters) offer great promise in killing the parasite. P-type ATPases are responsible for the maintenance of Ca2+ and V-type ATPases - of H+ homeostasis [59, 18, 57, 19, 2, 32, 94].

Para

site

mem

bran

e

Phospholipid synthesis

Choline transporter Mono- and bisquaternary ammonium compounds

A detailed description of mono- and bisquaternary compounds participating in phospholipid metabolism is provided in the next chapter.

Cell cycle control

Cyclin-dependent protein kinases (CDKs): P.falciparum protein kinase (PfPK) 5, PfPK6, PfMRK and Pfnek-1

Purvalanol, hymenialdisine and indirubine-3’-monoxime inhibit PfPK5; phenylquinolinones, oxindoles, chalcones, tryptanthrins, thiophene sulfonamides inhibit PfMRK; Xestoquinone inhibits Pfnek-1

CDKs function as serine/threonine kinases and have crucial role in parasite growth and differentiation. Siginificant differences exist between Plasmodium and human CDKs [39, 38, 14]. Xestoquinone is isolated from Pacific marine sponges of the genus Xestospongia [62].

20

Page 21: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Table 1.1 (continued): New targets in Plasmodium

Organelles in

Plasmodium Pathway Potential targets Lead inhibitors in development Details

Purine salvage Adenosine deaminase (ADA), purine nucleoside phosphorylase (PNP) Hypoxanthine-guanine phosphoribosyl-transferase (HGPRT)

6-thioguanosine inhibits PfADA, 5’-methylthio-immucillin-H inhibits PfPNP, 6-thioguanine inhibits HGPRT

Plasmodium lacks de novo synthesis of purines and has to salvage purines from the host. PfADA, PfPNP and HGPRT permit the parasite to form hypoxanthine (precursor of all purines) from erythrocyte purine pools [102, 84].

Anaerobic glycolysis

Lactate dehydrogenase (LDH)

Gossypol derivatives – gossylic nitrile diacetate

NAD+ is regenerated by reduction of pyruvate to lactate by LDH that is variant from human LDH. The disesquiterpene gossypol is isolated from cotton seed, but is highly toxic; its derivatives show high efficacy against P.falciparum in vitro [81, 84, 90].

Folate metabolism

Dihydrofolate reductase - thymidylate synthase (DHFR-TS)

5-fluoroorotate, Eosin B (4’,5’-dibromo-2’,7’-dinitrofluorescein) derivatives

Even partial inhibition of either DHFR or TS leads to nucleotide imbalances and parasite death. DHFR domain significantly differs from its mammalian counterpart. Eosin B is a potent inhibitor of DHFR-TS as well as glutathione reductase and thioredoxin reductase in drug-resistant malaria strains [70]. 5-fluoroorotate is metabolised to 5-fluoro-2’-deoxyuridylate that binds to TS domain [48].

Cyt

osol

Polyamine biosynthesis

Bifunctional enzyme of ornithine decarboxylase (ODC) and S-adenosyl-methionine decarboxylase (AdoMetDC)

Difluoromethylornithine (DFMO), analogues of 3-amino-oxy-2-fluoro-1-propanamine inhibit ODC

Polyamines (putrescine, spermidine, spermine) that are essential for cell growth and differentiation are synthesized in the parasite. DFMO is incapable to inhibit parasitic growth in vivo due to newly expressed transporters of putrescine and spermidine in the infected erythrocytes. The potential compound therefore should inhibit polyamine biosynthesis and transport [68, 102, 88, 58].

New permeability pathways (NPP) - the intracellular parasite induces in the host erythrocyte membrane new permeation pathways that provide the major route of entry of some essential nutrients (glutamate, pantothenate) and they mediate the efflux of various metabolic wastes. NPPs are broadly anion selective, but with a significant permeability to both organic and inorganic cations [13, 40].

Furosemide analogues (H156, H158), niflumic acid, sulfo-NHS-LC-biotin (succinimidyl-6-(biotinamido)hexanoate), bioflavonoid glycoside phlorizin, sulfonyl ureas (glibenclamide, tolbutamide), several arylaminobenzoates; dinucleoside dimmers

Two approaches: (i) inhibition of the transport of nutrients essential for parasite development, (ii) selective transport of cytotoxic compounds - dinucleoside phosphate dimers are conjugated to antimalarial compounds to improve selective access to parasite targets [12, 18, 99, 14, 93].

21

Page 22: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

VI. POTENTIAL INHIBITORS OF PHOSPHOLIPID METABOLISM IN

PLASMODIUM

The metabolic pathway that is targeted in Plasmodium needs to be essential for parasite

survival and its inhibition should cause a nonreversible cytotoxic effect that is crucial to the parasite

but not to the mammalian host [104]. Phospholipid synthesis in Plasmodium is a unique and vital

pathway in membrane biogenesis of the asexual stages of malaria parasites.

Intraerythrocytic malaria parasites, especially in most metabolically active trophozoite stages,

synthesize considerable amounts of membranes with quasi-absence of cholesterol. These

membranes are necessary to enclose the parasitophorous vacuole, cytosol and multiple subcellular

compartments [93]. Therefore the infected-erythrocyte phospholipid (PL) content increases by as

much as 500% with phosphatidylcholine (PC) and phosphatidylethanolamine (PE) representing

about 85% of total PL content [3].

Host choline is required for de novo synthesis of PC in the parasite, thus inhibition of the

choline transport has been identified as a promising therapeutic strategy [93, 3].

6.1. De novo synthesis of phosphatidylcholine in Plasmodium

60-80% of PC is synthesized from choline via the Kennedy pathway that is also essential in

mammalian cells. CCT (choline-phosphate cytidylyltransferase) catalyzes the conversion of

phosphocholine into CDP-choline, and is the rate-limiting enzyme presenting substantial

differences with respect to its mammalian counterpart. Moreover, PC is also obtained via the

phosphatidylethanolamine N-methyltransferase pathway and from serine, which is incorporated into

phosphatidylserine and then decarboxylated to phosphatidylethanolamine [81, 61]. In this pathway

of de novo PC synthesis, parasitic CCT and choline transport, which supplies the precursor, are the

rate-limiting steps [5]. Figure 1.5 represents the PC synthesis pathway in Plasmodium.

22

Page 23: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Figure 1.5: The metabolic pathway of PC biosynthesis in Plasmodium [77]. Abreviations

used are: PA, phosphatidic acid; DAG, diacylglycerol; Ino, inositol; Ser, serine; Cho, choline, P-Cho, phosphocholine;

P-Etn, phosphoethanolamine; PtdIno, phosphatidylinositol; PtdSer, phosphatidylserine; PtdEtn,

phosphatidylethanolamine; PtdMMeEtn, phosphatidylmonomethylethanolamine, CDS, CDP-DAG synthase; PSD,

phosphatidylserine decarboxylase; PEMT, phosphatidylethanolamine methyltransferase; EK and CK, ethanolamine and

choline kinase, ECT and CCT, ethanolamine and choline cytidylyltransferase; EPT and CTP, ethanolamine and choline

phosphotransferase.

6.2. Potential inhibitors of PL metabolism

Series of compounds that mimic choline structure (quaternary ammonium compounds) have

been synthesized and tested in vitro for their activity against the intraerythrocytic stage of

P.falciparum and P.vivax by Henri J.Vial and colleagues. Three generations of compounds

possessing one or two quaternary ammonium ions and long lipophilic alkyl chains were rationally

designed [5]:

1st generation: mono- and bisquaternary ammonium salts (lead compounds: E10 and G25,

respectively).

2nd generation: amidine and guanidine compounds (MS1).

3rd generation: mono- and bisthiazolium compounds (lead compound: T3) and their disulfide

and thioester prodrugs (TE3).

Bisquaternary ammonium salts showed powerful antimalarial activity, with IC50 in the

nanomolar range, but had a very low oral bioavailability due to their permanently charged

quaternary ammonium moiety. However, oral administration of antimalarial compounds is essential

for dispensaries in endemic countries (due to lack of adequate facilities to safely give drug

injections), and is indispensable for prophylactic or curative treatments for travellers. The good

23

Page 24: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

bioavailability of orally delivered antimalarial compound is a prerequisite. Therefore, bioisosteric

analogues (bis-amidines), exhibiting similar powerful activities, and subsequently the third

generation of thiazolium salts and their non-ionic precursors (prodrugs) were synthesized [95].

Prodrugs are converted to active ionized bisthiazolium drugs in the presence of plasma esterases

[80].

The lead compound of bisthiazolium salt has potent in vitro as well as in vivo antimalarial

activities and its corresponding bioprecursor was found to completely cure P.cynomolgi infected

Rhesus monkeys after 4 days of treatment with daily oral doses of 3 mg/kg [80, 105].

6.3. Quaternary ammonium compounds exhibit dual mode of action against Plasmodium

Mono- and bisquaternary ammonium, mono- and bisthiazolium salts exert their antimalarial

activity by two ways: (i) inhibiting plasmodial PL metabolism via the blockage of choline

transporter, and (ii) interacting with FP IX and the parasite pigment hemozoin [18]. These two

distinct modes of action against Plasmodium should prevent the rapid development of resistance

[91].

Choline transporter constitutes a rate-limiting step in PC biosynthesis pathway and shows

specific biochemical properties that distinguish it from the high-affinity choline transporter

observed in neurons (P.falciparum choline transporter has a low sensitivity to hemicholinium-3

(HC-3) that is an inhibitor of the high-affinity choline transporter in neurons) [18]. Choline

penetrates infected erythrocyte membranes through the endogenous carrier and through parasite

induced NPP and then is delivered into the parasite via Plasmodium membrane choline transporter.

This transporter exhibits such properties: (i) it is inhibitor-sensitive, (ii) temperature-dependent, (iii)

Na+-independent, (iiii) it is driven by the proton-motive force and (iiiii) the Michaelis-Menten

constant for choline is Km = 25.0 ± 3.5 µM [16]. Competitive blockage of choline transporter in the

infected erythrocyte or parasite plasma membrane by quaternary ammonium compounds results in

PL metabolism inhibition.

The potential antimalarial activity of bisquaternary ammonium or bisthiazolium salts relies on

their high intracellular accumulation (CAR~500), which results from accumulation in the malarial

digestive vacuole and subsequent interaction with FP IX and hemozoin [91, 16]. Although this

mode of action correlates with the chloroquine mechanism of action, bis-quaternary compounds

have not shown any cross-resistance with chloroquine [15].

24

Page 25: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

6.4. Structure-activity relationship of quaternary ammonium compounds

Over recent years, more than 600 compounds that are choline analogues have been rationally

synthesised. The cationic charges of mono- and bis-quaternary ammonium and the presence of a

long lipophilic chain on the nitrogen atom are crucial for in vitro antimalarial activity. This activity

was dramatically increased by 2 orders of magnitude when the quaternary ammonium was

duplicated and the alkyl chain between the two nitrogen atoms contained at least 12 methylene

groups (IC50 of 10-9 to 10-12 M) [4]. Recently designed thiazolium salts with cyclic quaternary

ammonium and their nonionic precursors are optimum structures for antimalarial activity, tolerance

and specificity [91].

6.5. Monothiazolium salts T1 and T2

Today the bisthiazolium salt (T3) is in development as very potential antimalarial compound

to treat severe malaria. However, the evaluation of monothiazolium salts T1 and T2 appeared

interesting. T1 could be a possible metabolite of T2 in the body. T1 and T2 have about 50-times

lower in vitro efficacy against Plasmodium falciparum than bisthiazolium salts, but their higher

hydrophobic features might result in increased oral absorption in vivo. Moreover, these two

compounds have shown more potent activity than T3 in the treatment of some other parasitoses

such as leishmaniasis and trypanosomiasis.

In order to evaluate pharmacokinetic parameters of monothiazolium salts, analytical methods

for the quantification of T1 and T2 in biological matrices have been developed and validated. The

chemical structures of T1 and T2 are given in Figure 1.6.

3-dodecyl-5-(2-hydroxyethyl)-4-methyl- 3-dodecyl-5-(2-methoxyethyl)-4-methyl-

1,3-thiazol-3-ium bromide 1,3-thiazol-3-ium bromide

Figure 1.6: The chemical structures of monothiazolium salts T1 and T2.

25

Page 26: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

PART 2 – EXPERIMENTAL PART

Before the presentation of the methodology and results of this work, a summarized procedure

required to validate an analytical method in biological matrices according to the European and FDA

guidelines [103] is presented.

I. DIFFERENT STEPS IN BIOANALYTICAL METHOD VALIDATION

Analytical methods used for the quantitative analysis of drugs and their metabolites in

biological samples are the key determinants in generating reproducible and reliable data that in turn

are used in the evaluation and interpretation of bioavailability, bioequivalence, and pharmacokinetic

studies [96]. It is essential to use well-defined and fully validated analytical methods to obtain

reliable results that can be satisfactorily interpreted [23].

Bioanalytical method validation includes all of the procedures required to demonstrate that a

particular bioanalytical method used for the quantitative determination of analytes (or series of

analytes) in a particular biological matrix (such as blood, plasma, serum, or urine) is reliable and

reproducible for the intended application [103]. The ultimate objective of the method validation

process is to provide evidence that the method does what it is intended to do [23].

The bioanalytical method validation can be envisaged to consist of two distinct phases: (1) the

bioanalytical method development phase in which the assay is defined and validated and (2)

application to actual analysis of samples from pharmacokinetic, bioavailability, bioequivalence and

drug interaction studies [96].

The required extent of a validation must only go as far as it is needed for the goal of the

application of the method. This means that only the analytical parameters that are of importance for

the routine application need to be validated [45].

1.1. Different types of bioanalytical method validation

[96, 103]

Full validation: establishment of all acceptable validation parameters for the bioanalytical

method for each analyte in order to apply to sample analysis. This type of validation is performed

when developing and implementing a bioanalytical method for the first time, for a new drug entity,

or if metabolites are added to an existing assay for quantitation.

Partial validation: modifications of validated bioanalytical methods that do not necessarily

require full re-validation. Some of the typical bioanalytical method changes include: bioanalytical

26

Page 27: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

method transfers between laboratories or analysts, change in analytical methodology (e.g., change

in detection system), change in matrix within species (e.g., human plasma to human urine), change

in species within matrix (e.g. human plasma to rat plasma), selectivity demonstration of an analyte

in the presence of specific metabolites or concomitant medications, change in sample processing

procedure(s), change in relevant concentration range. The decision of which parameters have to be

revalidated depends on the logical consideration of the specific validation parameters likely to be

affected by the change made to the bioanalytical method.

Cross validation: Cross validation is a comparison of validation parameters when two or more

bioanalytical methods are used to generate data within the same study or across different studies.

1.2. Methodology

[103, 51, 23, 45, 27, 96]

The criteria considered in the validation are as follows:

Relationship between concentration and response

Specificity / selectivity

Accuracy and precision

Lower limit of quantitation (LLOQ)

Extraction recovery

Stability

Robustness / ruggedness

Matrix effects

1.2.1. Relationship between concentration and response:

The choice of an appropriate calibration model is necessary for reliable quantitation.

Therefore, the relationship between the concentration of the analyte in the sample and the

corresponding detector response must be investigated. This can be done by analyzing spiked

calibration samples and plotting the resulting ratios of peak area or peak height (analyte/internal

standard) versus the corresponding concentrations.

A sufficient number of standards should be used to adequately define the relationship between

concentration and response, and to demonstrate that the relationship is continuous and reproducible.

Concentrations of standards should be chosen on the basis of the concentration range expected in a

particular study. In many cases, six to eight concentrations (excluding blank – matrix sample

processed without analytes) can define the standard curve. For the evaluation of repeatability, it is

recommended to analyse at least six replicates of calibration set within a day (the same analyst, the

same stock solution, the same material). Reproducibility can be evaluated if one set of calibration is

27

Page 28: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

analysed per day following at least 6 different days (the same analyst, different stock solution,

different material).

Standard curve fitting is determined by applying the simplest model (unweighted or weighted

least-squares linear regression y = ax + b, quadratic equation y = ax² + bx + c, etc.) that adequately

describes the concentration-response relationship. Statistical tests must be used to verify goodness

of fit. The quality of fit can be determined (i) by comparing back-calculated concentrations (i.e.,

concentrations obtained by entering the measured response for each standard into the regression

equation) to the nominal ones, (ii) by evaluating the correlation coefficient, (iii) by calculating the

bias or mean residual value (i.e. difference between back-calculated and added concentrations; the

bias should not be statistically different from 0), (iv) by studying the distribution of the residuals

and etc. Selection of a weighing factor and use of a complex regression equation should be justified.

If the relationship between response and concentration is linear, the test of “lack of fit” can be used.

The following conditions should be met in developing a calibration curve: 20% deviation of

the lower limit of quantitation (LLOQ) from nominal concentration and 15% deviation of standards

other than LLOQ from nominal concentration. At least five non-zero standards should meet the

above criteria, including the LLOQ and the highest concentration of the calibration curve.

Excluding the standards (leaving at least 5 points in the calibration curve) should not change the

model used.

1.2.2. Specificity / selectivity:

Selectivity is the ability of an analytical method to differentiate and quantify the analyte in the

presence of other components (metabolites, impurities, degradation products, co-administered

drugs) in the sample. Specificity is the ability to assess unequivocally the analyte in the presence of

endogenous compounds.

The specificity of the assay methodology should be established using a minimum of six

independent sources of the same matrix (exceptions can be made for hyphenated mass

spectrometry-based methods as matrix effects should be investigated).

For chromatographic procedures, representative chromatograms should be used to

demonstrate that there is no interference across the time windows of peaks of interest (i.e.,

comparison of retention times of endogenous compounds of the matrix to those of the drugs to be

analysed).

1.2.3. Accuracy and precision:

Precision and accuracy are the most important factors in assessing the quality of an analytical

method. They are determined by analysing quality control (QC) samples against a calibration curve.

28

Page 29: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

The accuracy of a bioanalytical method is a measure of the systematic error or bias and is

defined as the degree of closeness of the determined value to the nominal or known true value under

prescribed conditions. Accuracy is calculated as follows: [mean found concentration/nominal

concentration] x 100. Accuracy is best reported as percentage bias and is sometimes termed

trueness.

The precision of a bioanalytical method is a measure of the random error and expresses the

closeness of agreement (degree of scatter) between a series of measurements obtained from multiple

sampling of the same homogenous sample under the prescribed conditions. Precision is usually

measured in terms of imprecision expressed as the percentage coefficient of variation (% CV) or

relative standard deviation (R.S.D.) of the replicate measurements.

Accuracy and precision are further subdivided into: (i) within-run, intra-batch, intra-assay

precision or accuracy that are assessed under the same operating conditions (the same analyst, the

same reagents and material) over a short interval of time (intra-day) and (ii) between-run, inter-

batch, inter-assay precision or accuracy that are measured on different days, and may involve

different analysts, material, reagents, and laboratories.

The accuracy and precision should be determined with a minimum of 6 determinations per

QC sample. A minimum of three concentrations (one within 3x the LLOQ (low QC sample), one

near the center (middle QC), and one near the upper boundary of the standard curve (high QC)) in

the range of expected concentrations is recommended. The concentrations of QC samples are

determined against a calibration curve (n = 6). The precision around the mean value should not

exceed 15% CV and the accuracy should be between 85-115%. It is desirable that these tolerances

be provided both for intra-day and inter-day experiments. At the LLOQ, 20% of precision and 80-

120% of accuracy are acceptable limits.

1.2.4. Lower limit of quantitation (LLOQ):

LLOQ is the lowest amount of an analyte in a sample that can be quantitatively determined

with suitable precision and accuracy. The LLOQ should be determined using at least five samples

containing an analyte. The LLOQ should serve as the lowest concentration on the calibration curve.

The following conditions for LLOQ should be met: (i) the analyte response at the LLOQ

should be at least 5 times the response compared to blank response; (ii) analyte peak (response)

should be identifiable, discrete, and reproducible with a precision of 20% and accuracy of 80-120%.

1.2.5. Extraction recovery:

Absolute recovery of a bioanalytical method is measured as the response of a processed

spiked matrix standard, expressed as a percentage of the response of a pure standard, which has not

29

Page 30: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

been subjected to sample pre-treatment. It indicates whether the method provides a response for the

entire amount of the analyte that is present in the sample. Generally, the absolute recovery is

calculated at each concentration of QC samples analysed in replicates (at least three times per

concentration).

In order to study the effect of co-extracted biological material, relative recovery is computed

by comparing responses of replicates of extracted QC samples with those of extracted blank matrix

to which analyte has been added at the same nominal concentration.

If an internal standard is used, its recovery should be determined independently at the

concentration level used in the method.

Recovery of the analyte need not to be 100%, but the extent of recovery of an analyte and of

the internal standard should be consistent, precise, and reproducible. It is most important to reach a

reproducible recovery, which is high enough to satisfy the requirements of quantifying low sample

concentrations, even when the recovery itself is low. Values of recovery not less than 50, 80 and

90% have been reported as numerical acceptance limits [54].

1.2.6. Stability:

Stability of pure analyte and/or solutions of the analyte must be studied in replicate (n > 3)

under normal laboratory conditions of heat, humidity, light and air exposure by comparison with

fresh solutions.

Drug stability in a biological matrix is function of the storage conditions, the chemical

properties of the drug, the type of matrix, and the type of container. Stability data are based on

replicate determinations (at least 3) of QC samples at two or three concentrations (low, medium and

high).

Two essential types of stability studies should be performed: 1) short term stability, including

bench-stop storage, +4°C, -20°C and stability during three freeze-thaw cycles, and 2) long-term

stability. Long-term stability must be proven over at least the maximum period of storage of study

samples under the temperature conditions to be used for study samples (i.e., -20°C, -80°C) and

using the exact type of container (e.g., glass, polypropylene) as for the study samples. Generally,

the assessment of long-term stability of frozen samples is achieved by comparing the responses of

frozen and freshly prepared (time 0) QC samples.

At least three aliquots of each of the low, middle and high concentrations (QCs) should be

investigated for each type of stability studies.

Stability of the analyte should be also evaluated during all steps of the analytical procedure

(solvent used for the extraction, light exposure, temperature, autosampler).

30

Page 31: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

1.2.7. Robustness / ruggedness:

The robustness of an analytical procedure is a measure of its capacity to remain unaffected by

small, but deliberate variations in method parameters and provides an indication of its reliability

during normal use. Typical variations in the case of liquid chromatography are: variations of pH

values in a mobile phase, variations in mobile phase composition, different columns, temperature,

flow rate. Full validation must not necessarily include ruggedness testing; it can however be very

helpful during the method development/prevalidation phase, as problems that may occur during

validation are often detected in advance. Ruggedness should be tested, if a method is supposed to be

transferred to another laboratory.

1.2.8. Matrix effects:

Matrix effects are attributed to those organic and/or inorganic components of a sample that

co-elute with an analyte affecting the ionisation efficiency in the mass spectrometer and, therefore,

the response of the analyte [24]. Endogenous co-eluting components present in any biological

matrix (e.g., salts, amines, fatty acids, etc.), compounds deriving from the LC separation (e.g.

mobile phase additives, ion-pairing agents) and exogenous materials (e.g. polymers, contained in

different brands of plastic tubes, Li-heparin, a commonly used anticoagulant) can be responsible for

this phenomenon [52, 74].

The exact mechanism of matrix effects is unknown, but it probably originates from the

competition among analyte ions and the co-eluting undetected non-volatile matrix components for

access to the droplet surface for gas phase emission [101]. These competitive mechanisms in the

droplet can cause a positive (ion enhancement) or negative (ion suppression) influence on the

resulting signal and have deleterious impact on methods precision, accuracy and sensitivity [29].

Therefore, according to recent guidelines for bioanalytical method validation, there is a need to

evaluate matrix effects during development and validation of LC-MS or LC-MS-MS methods.

In most cases, method validations are performed using standard and quality control samples

prepared from unique source of pooled matrix (whole blood, urine, etc.). In such a case, the matrix

is homogenous and thus can be assumed that prepared samples will have the same absolute matrix

effects (the response of solutes to be measured will be modified in an identical way). The use of

homogenous samples for validation does not take into account the inter- and intra-patient matrix

variability (relative matrix effect) that is present in the clinical setting [101].

The two common ways to assess matrix effects are (i) the post-extraction addition method or

(ii) the post-column infusion method. The post-extraction addition technique requires sample

extracts with the analyte of interest added post-extraction compared with pure solutions prepared in

mobile phase and containing equivalent amounts of the analyte of interest. The response of post-

31

Page 32: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

extraction sample / response of pure solution ratio determines the degree of matrix effect occurring

to the analyte in question under chromatographic conditions. This ratio should be close to 1 (a limit

of 0.85-1.15 is generally admitted). A more dynamic technique for determining matrix effects is

post-column infusion. An infusion pump is used to deliver a constant flow of the analyte into the

HPLC eluent at a point after the chromatographic column and before the mass spectrometer

ionization source. A sample extract (without added analyte) is injected into the HPLC portion of the

instrument under the desired chromatographic conditions and the response from the infused analyte

recorded. The post-infusion technique enables the influence of the matrix on analyte response to be

investigated over the entire chromatographic run [101].

A simple experimental approach to identify and study the relative (i.e., variation of absolute

matrix effect between different lots of matrix) matrix effect has been recently described by Bogdan

Matuszewski [73]. This author suggests to investigate the precision of standard line slopes

constructed in five different sources of matrix (n = 5). Both precision (i.e., CV of the slope) and

accuracy (difference between the highest and the lowest slope divided by the lowest slope) are

calculated. A slope precision value of <3-4% is a guide for reliable method that is free from relative

matrix effect and is applicable to support clinical studies [73].

1.3. Application of validated bioanalytical method to routine drug analysis

[103, 23]

A calibration curve should be generated to quantify each analyte in each analytical run and

should be used to calculate the concentration of the analyte in the unknown samples in the run. The

spiked samples can contain more than one analyte. The calibration curve should cover the expected

unknown sample concentration range and should consist of a minimum of six standard points,

excluding blanks. An analytical run should consist of QC samples (at least duplicated), calibration

standards, and the processed samples to be analysed. The QC samples should be used to accept or

reject the run. The same analyst prepares calibration curve and samples to be analysed.

Matrix-based standard calibration samples: 75%, or a minimum of six standards, when back-

calculated should fall within ±15%, except for LLOQ, when it should be ±20% of the nominal

value. At least 67% (4 out of 6) of the QC samples should be within ±15% of their respective

nominal values; 33% of the QC samples (not all replicates at the same concentration) may be

outside the ±15% respective nominal value.

If study samples are run singly, a single calibration curve should normally be performed. If

replicates of study samples are analysed, identical replication of standard curves is recommended.

However, in practice, calibration curves are performed in duplicate even if the samples are analysed

singly.

32

Page 33: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

II. ASSAY FOR SIMULTANEOUS DETERMINATION OF TWO

ANTIMALARIAL MONOTHIAZOLIUM COMPOUNDS IN BIOLOGICAL

MATRICES BY LIQUID CHROMATOGRAPHY – ELECTROSPRAY MASS

SPECTROMETRY

2.1. Materials and methods

2.1.1. Chemicals and reagents:

T1 (3-dodecyl-5-(2-hydroxyethyl)-4-methyl-1,3-thiazol-3-ium bromide), T2 (3-dodecyl-5-(2-

methoxyethyl)-4-methyl-1,3-thiazol-3-ium bromide) and T3 (internal standard, 1,12-bis[4-methyl-

5-(2-hydroxyethyl)-thiazol-3-ium-3-yl]dodecane dibromide, MW 614.7) were obtained from the

Laboratoire des Aminoacides, Peptides et Proteines (University Montpellier I and II). The purity of

these standards was evaluated by elemental analysis; results were within ± 0.4 of calculated values.

They were stored at ambient temperature with protection from light.

Trifluoroacetic acid (TFA) was obtained from Sigma (St Louis, MO, USA). Acetonitrile,

methanol, ammonium formate, formic acid and trimethylamine were purchased from Merck

(Darmstadt, Germany). All chemicals were of analytical grade. In-house deionised water was

further purified with a Milli-Q water-purifying system (Millipore, Bedford, MA, USA). Oasis HLB

cartridges (30 mg of sorbent, average particle diameter 30 µm) were supplied by Waters (Saint

Quentin, France).

For method validation, human whole blood and plasma were obtained from pooled blood

samples collected from healthy volunteers not undergoing drug therapy (Etablissement Français du

sang, Montpellier, France). Coagulation was prevented by adding EDTA-sodium salt. Blood

samples from Sprague-Dawley rats (Charles River Laboratories) were collected with sodium

heparin. The blood was centrifuged at 2000 x g for 10 min to obtain plasma. Red blood cells

(RBCs) were washed twice with an equal volume of 0.9% sodium chloride before storage. The

drug-free whole blood, RBCs and plasma were aliquoted, stored at -80°C and then used during the

study for the preparation of standards and quality control (QC) samples.

Individual stock solutions of T1 (45 mg/L), T2 (45 mg/L) and T3 (65 mg/L), expressed in the

form of charged compounds, were prepared by accurately weighing the required amounts in

separate volumetric flasks and dissolving in purified water. For each compound, two separate stock

standard solutions were prepared: one was used for the preparation of the calibration curve

standards and the second was used for the preparation of quality control (QC) samples.

33

Page 34: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

2.1.2. Equipment:

The liquid chromatography – mass spectrometry (LC-MS) analysis was performed using an

Agilent 1100 quadrupole mass spectrometer (Agilent Technologies, Lesulis, France) (mass range:

m/z 50-2000) working with an electrospray ionisation (ESI) source in the positive mode.

The main parts of the mass spectrometer are the ion source, where the molecules are ionized,

the mass analyzer, where molecules are separated according to their mass-to-charge (m/z) ratio, and

the detector, where the ion signal is transferred to qualitative and quantitative data [87].

Electrospray is currently the most widely used ionisation source. ESI is a “soft” ionisation

technique (producing primarily molecular species with little fragmentation), forming protonated

[M+zH]z+or de-protonated [M-zH]z- ions for positive and negative ionisation mode, respectively

[60]. Positively charged ions such as inorganic cations and protonated organic bases are analyzed in

the positive ion mode, whereas negatively charged ions such as inorganic anions and deprotonated

organic acids are analyzed in the negative ion mode.

ESI is an interface that: (i) is relatively easy to use, (ii) exhibits low solvent consumption, (iii)

is applicable to a wide range of polar and thermally labile analytes of both low and high molecular

weight (up to 100 kDa) and (iv) is compatible with a wide range of HPLC conditions (mobile phase

flow rates from 20 nL/min to in excess of 1 mL/min). The electrospray interface produces singly or

multiply charged ions directly from an aqueous–organic solvent system. For analysis in the positive

ion mode (the most frequently used), solutions of water with a high percentage of either methanol

or acetonitrile are generally preferred. A volatile, weak acid, such as acetic acid, is added to this

solvent to increase conductivity and to facilitate the protonation of the analyte [25, 1].

The compounds that may be studied are as follows: (i) ionic compounds that are intrinsically

charged in solution; (ii) neutral/polar compounds that may be protonated (for positive-ion mass

spectra) or deprotonated (for negative-ion mass spectra) under the solution conditions employed,

e.g. appropriate pH; (iii) non-polar compounds that undergo oxidation (positive-ion mass spectra) or

reduction (negative-ion mass spectra) at the electrospray capillary tip.

The mechanism of electrospray ionisation includes three main steps [21, 25, 6] that are

presented in Figure 2.1:

1. The formation of charged droplets at the tip of the spray needle. A liquid, in which the

analyte(s) of interest has been dissolved, is passed at atmospheric pressure through a metal needle,

maintained at high voltage (2–5 kV). This voltage can be either negative or positive, depending on

the analytes chosen. The applied voltage provides the electrical field gradient (between the needle

tip and a counter electrode) required to charge the surface of the liquid and produce charge

separation. As a result, a so-called Taylor cone protrudes. Consequently, a small liquid filament is

34

Page 35: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

established and small highly charged droplets are generated in a fine mist. A concentric flow of

nitrogen gas assists in the nebulization process.

2. Evaporation of solvent from the droplets. When the charged droplets travel towards the

counter electrode, their radius decreases as the solvent evaporates, but their charge remains

constant. At a certain point, called the Rayleigh limit, the Coulombic force overcomes the surface

tension of the liquid and the droplet undergoes fission into several smaller droplets with a radius

approximately 10% of that of the parent droplet. This so-called Coulombic fission can be repeated

in several cycles, leading to very small highly charged droplets. These droplets move through the

atmosphere towards the capillary sampling orifice and subsequently into the mass analyzer (e.g.,

quadrupole). There is a counterflow of heated nitrogen drying gas that shrinks the droplets and

carries away the uncharged material.

Figure 2.1: Schematic representation of electrospray ionization process [25].

3. The formation of gas phase ions. The actual mechanism for the transfer from solvated ions

to gas phase ions is not fully understood and has been under discussion for a long time. Two main

theories have been suggested: the charged residue mechanism (CRM) and the ion evaporation

mechanism (IEM).

The CRM mechanism implies that the gas-phase ions are produced when solvent evaporation

causes a series of Coulomb fission events that ultimately leads to the production of final small

droplets bearing one or more excess charges, but only a single analyte molecule. This analyte

molecule retains the ‘residual’ droplet charge, also when the last solvent molecule evaporates thus

forming a free gas-phase ion (Figure 2.2, A). On the other hand, the IEM mechanism asserts that

solvated ions are emitted directly from the charged droplets when the electric field on the surface of

35

Page 36: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

the charged droplets is sufficiently high, well before the Rayleigh limit (radius <10 nm) is reached

(Figure 2.2, B) [30].

A B

Figure 2.2: Formation of gas phase ions. A – the charged residue mechanism (CRM); B -

the ion evaporation mechanism (IEM).

The mass spectrometer, used in the studies, was coupled to a Hewlett Packard LC system

equipped with a quaternary pump, a degasser and an autosampler set at 4°C. HPChem software

(version 8.04) from Agilent Technologies was used for data acquisition and handling.

Separation of the analytes was performed on a Zorbax Eclipse XD8 C8 column 150 x 4.6 mm,

i.d., packed with particles of 5 µm size (Agilent technologies). A C8 symmetry column (20 x 3.9

mm, i.d., 5 µm-size) from Waters Corporation was used as a guard column. The column was

thermostated at 20 °C.

2.1.3. Liquid chromatography-mass spectrometry conditions:

Chromatography was carried out using the gradient system of two solvents: A, 130 µL/L

trimethylamine in acetonitrile, and B, 2 mM ammonium formate buffer (126 mg/L of ammonium

formate were dissolved in purified water, the pH was adjusted to 3 ± 0.05 with formic acid). The

starting eluent consisted of 15% A and 85% B; during the first 10 min the proportion of eluent A

was increased linearly to 100% and then held for 2 min in order to wash the column. Initial

composition of eluent A (15%) and B (85%) was returned in subsequent 2 min and then held for 5

min in order to re-equilibrate the column.

The flow rate was 800 µL/min and the injection volume was 10 µL.

The mass spectrometer was calibrated in the positive ion mode using a mixture of NaI and CsI

(peak width of the mass: 0.6-0.7 amu). The MS system was operated with a capillary voltage of 4.0

kV. The sampling cone voltage was set at 60 V for T1 and T2, and at 80 V for T3. The drying gas

temperature and flow were maintained at 350°C and 12.0 L/min, respectively, and the nebulizer

pressure was set at 35 psig.

Mass spectrometric data were acquired using selected ion monitoring (SIM) mode (SIM dwell

time: 98 ms). T1 and T2 were detected through the (M+) ions at m/z 312 and m/z 326, respectively.

36

Page 37: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

The T3 compound was detected by use of the quaternary ammonium salt (M²+/2) at m/z 227.3. The

mass spectra (scan mode) of T1, T2 and T3 are shown in Figure 2.3.

Figure 2.3: Mass spectra (scan mode) for T1 (A), T2 (B) and T3 (C).

2.1.4. Working standards:

Stock solutions of T1 and T2 were appropriately diluted in purified water to obtain the 14

working standard solutions (0.056, 0.111, 0.223, 0.56, 1.113, 2.225, 5.56, 11.125, 22.5 mg/L) used

to spike calibrators and QC samples. The stock solution of T3 was diluted 8-fold (8.125 mg/L) in

purified water before use. A reference standard solution containing T1, T2 (2.25 mg/L of each compound), and the

internal standard T3 (1.62 mg/L) was prepared daily in a mixture of 1 mL/L TFA in water and 1

mL/L TFA in acetonitrile (50:50, v/v). This solution was injected before each run to verify the

performance of the LC/ESI-MS system.

2.1.5. Preparation of calibration standards and quality control (QC) samples:

Calibration standards were prepared in drug-free plasma (0.5 mL), drug-free whole blood or

drug-free RBCs (0.25 g diluted with 0.25 mL of purified water). The calibration set consisted of 7-9

concentrations, prepared by adding 20 µL of the relevant working standard solution to drug-free

matrices. Concentrations across the range of 2.25 to 900 µg/L (expressed in the form of charged

compound) for T1 and T2 in plasma (2.25, 4.5, 9.0, 22.5, 45, 90, 225, 450 and 900 µg/L) and of 4.5

to 900 µg/kg for T1 and T2 in whole blood and RBCs (4.5, 9, 18, 45, 90, 450 and 900 µg/kg) were

obtained. A calibration set also included a blank matrix. QC samples at four different levels (3.75,

11.2, 112 and 675 µg/L in plasma; 7.5, 22.5, 225 and 600 µg/kg in whole blood and RBCs) were

37

Page 38: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

prepared in the same way as the calibration standards by mixing drug-free matrices with 20 µL of

appropriate working solutions.

2.1.6. Sample preparation procedure:

Plasma, whole blood and RBC samples were subjected to a solid-liquid extraction for the

removal of proteins and interfering components. The 1 cm3 Waters Oasis HLB cartridges (sorbent:

N-vinylpyrrolidone-divinylbenzene copolymer, that retains both ionised and non-ionised

compounds) were conditioned with 1 mL of methanol followed by 1 mL of purified water before

use.

In a 5 mL polypropylene tube, after adding 20 µL of internal standard solution (8.125 mg/L)

and 0.5 mL of water containing 10 ml/L TFA (to precipitate the proteins) to 0.5 mL plasma sample

aliquot, the mixture was vortex-mixed for 15 s and then centrifuged for 10 min at 1500 g (4°C). The

supernatant was loaded onto the conditioned cartridge under a light vacuum (approximately 86 kPa)

using a Vac Elut 20® (Varian). The cartridge was then rinsed with 1 mL of purified water and dried

for 2 min by vacuum aspiration (approximately 27 kPa). The elution was carried out with 2 x 1 mL

of acetonitrile containing 1 mL/L TFA under a light vacuum (approximately 86 kPa). The eluate

fractions were collected in a 5 mL polypropylene tube and evaporated to dryness under the stream

of nitrogen using a Zymark Turbovap LV evaporator (Zymark Corporation, Roissy, France):

thermostatically controlled water-bath was maintained at 40°C and the evaporation process lasted

for about 40 min. The dried residue was reconstituted in 100 µL of a mixture of water containing 1

mL/L TFA and acetonitrile containing 1 mL/L TFA (50:50, v/v).

20 µL of internal standard (8.125 mg/L) were added to 0.5 mL of diluted whole blood or RBC

samples (0.25 g of whole blood or RBCs plus 0.25 mL of purified water) and mixed with 0.5 ml of

water containing 10 mL/L TFA. The acidic solution was added drop-wise while the mixture was

vortex-mixed in order to obtain smaller precipitate particles. The mixture was centrifuged for 20

min at 17,562 g (4°C). Thereafter, the assay procedure was as described above for the plasma

samples except that the cartridge was washed with 1.5 mL of purified water instead of 1 mL.

2.1.7. Data analysis:

From recorded peak areas, the ratios of each analyte and internal standard were calculated.

The obtained ratios were linked to the theoretical concentrations of each analyte in the matrix

according to a quadratic relationship (Y=ax2+bx+c, where Y is the peak area ratio and x is the

concentration of the analyte) using the PK-fit program [34]. The regression curve was not forced

through zero. The resulting equation parameters (a, b and c) were used to calculate the ‘back-

calculated’ concentrations and the obtained values were statistically evaluated. The normal

38

Page 39: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

distribution of the residuals (i.e. difference between nominal and back-calculated concentrations)

was verified. For the comparison of the back-calculated concentrations (Ccal) and the theoretical

concentrations (Cref), the bias (mean predictor error; formula 2.1) was computed and compared to

zero (Student t-test).

Bias = ∑=

=

−ni

iREFCALC CC

n 1][1 (2.1)

Where n - the number of values; Ccal – the back-calculated concentration; Cref - the theoretical

concentration.

The 95% confidence interval for bias was also computed.

2.1.8. Validation procedure:

The validation criteria considered here are specificity, accuracy, precision, extraction recovery

and lower limit of quantitation.

The specificity of the analytical method was determined by the analysis of ten different

independent sources of the same biological matrix. The retention times of endogenous compounds

in the matrices were compared with those of the compounds of interest.

Both intra- and inter-assay precision and accuracy by analysing QC samples at four

concentrations (3.75, 11.2, 112 and 675 µg/L in plasma; 7.5, 22.5, 225 and 600 µg/kg in whole

blood and RBCs) were assessed. For intra-assay, replicate analyses (n = 6) of each QC sample were

performed the same day. For inter-assay, a set of QC samples was analysed once a day following 6

different days (n = 6). The back-calculated concentrations of QC samples were determined from a

calibration curve prepared for each set of QC samples. The accuracy was evaluated as [mean found

concentration/nominal concentration] x 100. Precision was given by the percent relative standard

deviation (R.S.D.).

Extraction recoveries of T1 and T2 in plasma, whole blood and RBCs were measured six

times at each concentration of QC samples. The areas under the peaks of extracted QC samples

were compared with those of the authentic (unextracted) standards in the relevant concentration

range prepared in a mixture containing 1 mL/L TFA in water and 1 mL/L TFA in acetonitrile

(50:50, v/v). The extraction recovery was also determined for the internal standard.

The LLOQ was defined as the lowest concentration that could be determined with accuracy

within 80-120% and precision < 20% on a day-to-day basis. To determine the analytical error in the

LLOQ, QC samples containing compounds of interest were used.

39

Page 40: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

2.1.9. Stability study:

The stability of stock solutions was determined at 4°C. Appropriately diluted stock solutions

were injected in triplicate into LC-MS system immediately after preparation (time 0) and at periodic

intervals after the storage at 4°C over a span of 1 month.

For stability studies in plasma, whole blood and RBCs, QC samples (3.75, 11.2, 112 and 675

µg/L in plasma; 7.5, 22.5, 225 and 600 µg/kg in whole blood and RBCs) were used. The short-term

stability study included the stability of samples stored at 4°C and 20°C during 6 hours and the

stability during three freeze-thaw cycles. The long-term stability study was carried out after 1

month of storage at -20°C and -80°C.

Spiked QC samples were analyzed immediately after preparation (reference values – T0) and

after storage. The back–calculated concentrations (according to freshly prepared calibration curve)

were computed; precision (%) and accuracy (%) were determined.

Each result represented the mean of three separate samples. Compounds were considered

stable when losses were < 15%.

2.1.10. Ion suppression / enhancement study:

The absence of ion suppression / enhancement attributable to the matrix effects was

demonstrated by the method of Matuszewski et al. [71].

For this procedure, six different batches of each drug-free matrix were treated as described

above in duplicate (n = 12 per matrix and per concentration studied).

A set of QC samples (4 concentrations) was prepared using extracted blank matrices (the

blank matrix was mixed with 0.5 mL of water containing 1% TFA and then centrifuged, extracted

and evaporated). The appropriate working solutions of T1, T2 and the internal standard T3,

prepared in 1 mL/L TFA in water and 1 mL/L TFA in acetonitrile (50:50, v/v), were added to the

extracted blank plasma, whole blood or RBCs.

The reference samples were prepared by adding the drugs to the solvent (1 mL/L TFA in

water and 1 mL/L TFA in acetonitrile (50:50, v/v)) to obtain the same final concentrations.

The reconstituted extracts (in 100 µL of a mixture of 1 mL/L TFA in water and 1 mL/L TFA

in acetonitrile (50:50, v/v)) and reference solutions were injected onto the analytical column. Peak

areas obtained from the reconstituted extracts were compared with the corresponding peak areas

produced by the reference solutions. The CV% at each concentration level was calculated.

2.1.11. Pharmacokinetic study of T2 in rat:

The pharmacokinetic study was carried out according to the Principles of Laboratory Animal

Care [79] and was approved by the local Animal Use Committee.

40

Page 41: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Before initiation of the study, Sprague-Dawley male rats were housed in cages (6 rats per

cage) and given rodent food and free access to water. Acclimatisation and stabilisation in the

environment of 20-24ºC and 40-70% of humidity lasted for one week. Just before the study rats

were weighted (approximately 220-250 g) and identified by the number.

The formulation of T2 for intravenous administration was prepared as follows: T2 was

dissolved in 0.9% NaCl solution to obtain 0.5 mg/ml solution.

The T2 doses of 2.1 mg/kg (expressed in the form of charged compound) for single

intravenous administration was adjusted according to the exact body weight of rats. Intravenous

administration was carried out to anesthetized (with diethyl ether) rats in the vein of penis.

Blood samples (one sample per rat) were drawn in lithium-heparinized polypropylene tubes at

the following time-points (3 animals per time-point): before drug administration, 5, 10, 30 minutes

and 1, 2, 4, 8, 12, 24 and 36 hours after drug administration. Blood samples were collected after the

sacrifice of the animal by the section of the carotid artery. Animals were anesthetized with diethyl

ether 2 minutes before sampling.

Obtained blood samples were centrifuged at 4°C (2000 g for 10 min). Plasma samples were

transferred into polypropylene tubes and stored at -80°C until assay. RBCs were washed twice with

an equal volume of 0.9% NaCl to limit plasma contamination before storage at –80°C.

The samples were processed and T2 quantified following the procedure of validated method

for the quantification of T2 in the biological matrices.

Pharmacokinetic parameters were computed using a compartmental approach from the

average concentration values at each time points using the Pk-fit software.

41

Page 42: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

2.2. Results of full validation in human plasma and whole blood

2.2.1. Drug / response relationship:

For each analyte, peak area ratios (analyte / internal standard) were linked to concentrations

using a quadratic equation. Mean coefficients of determination (r2) were 0.991 (R.S.D., 0.4%) for

T1 and 0.995 (R.S.D., 0.4%) for T2. For each compound, mean parameters of the quadratic

equation are presented in Table 2.1.

Table 2.1: Mean parameters of the quadratic equation

Analyte a (mean) ± S.D. b (mean) c (mean) ± S.D.

Human plasma (n=6)

T1 -1.90 x 10-6 ± 6.5 x 10-7 0.0060 (R.S.D., 6.1%) -0.0105 ± 0.0048 intra-day

T2 -2.16 x 10-6 ± 6.4 x 10-7 0.00815 (R.S.D., 9.3%) -0.0136 ± 0.0045

T1 -1.39 x 10-6 ± 2.01 x 10-7 0.00487(R.S.D., 6.6%) -0.00773 ± 0.0077 inter-day

T2 -1.65 x 10-6 ± 2.56 x 10-7 0.00627(R.S.D., 7.2%) -0.0123 ± 0.0101

Human whole blood (n=6)

T1 -3.13 x 10-7± 2.18 x 10-8 0.0034 (R.S.D., 10.4%) -0.000184 ± 0.0032 intra-day

T2 -6.97 x 10-7± 4.89 x 10-8 0.00498 (R.S.D., 12.5%) -0.010183 ± 0.0053

T1 -6.09 x 10-7± 4.74 x 10-8 0.00305 (R.S.D., 11.7%) -0.00506± 0.0070 inter-day

T2 -5.84 x 10-7± 6.44 x 10-8 0.00395 (R.S.D., 7.6%) -0.00780 ± 0.0056

For each point on the calibration curve, the concentrations were back-calculated from the

corresponding quadratic equation parameters. Mean back-calculated concentrations together with

precision and recovery (computed from mean back-calculated concentrations) are presented in

Tables 2.2 (human plasma data) and 2.3 (human whole blood data).

42

Page 43: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Back-calculated concentrations from calibration curves constructed in human whole blood

Inter-day (n=6) T1+ T2+

Nominal conc. µg/kg

Mean back-calcul.

conc. µg/kg

R.S.D. (%)

Recovery (%)

Mean back-calcul.

conc. µg/kg

R.S.D. (%)

Recovery (%)

4.50 4.24 14.3 94.3 4.59 11.7 1029.0 8.92 12.0 99.1 8.60 9.9 95.6

18.0 17.2 7.3 95.3 17.8 7.2 98.945.0 43.0 6.7 95.7 42.1 7.4 93.690.0 89.2 12.5 99.1 88.7 10.4 98.6180 179 3.4 99.4 181 4.0 101450 415 8.7 92.2 434 9.9 96.5900 899 1.9 99.9 898 2.0 99.8

43

Table 2.2: Mean back-calculated concentrations from calibration curves in human plasma

Back-calculated concentrations from calibration curves constructed in human plasma Intra-day (n=6)

T1+ T2+ Nominal

conc. µg/L

Mean back-calcul.

conc. µg/L

R.S.D. (%)

Recovery (%)

Mean back-calcul.

conc. µg/L

R.S.D. (%)

Recovery (%)

2.25 2.28 15.4 101 2.35 14.2 1054.5 4.13 12.1 91.7 4.08 7.1 90.79.0 8.62 5.0 95.7 8.37 4.2 93.0

22.5 22.8 4.2 101 22.4 6.1 99.545 43.9 6.8 97.5 44.2 6.1 98.190 90.6 4.0 101 90.1 4.0 100

225 228 5.7 101 216 6.0 96.4450 437 5.2 97.3 431 4.2 95.7900 895 2.7 99.5 878 4.4 97.6

Back-calculated concentrations from calibration curves constructed in human plasma Inter-day (n=6)

T1+ T2+Nominal

conc. µg/L

Mean back-calcul.

conc. µg/L

R.S.D. (%)

Recovery (%)

Mean back-calcul.

conc. µg/L

R.S.D. (%)

Recovery (%)

2.25 2.02 10.1 89.8 2.34 19.3 1044.5 4.55 15.4 109 4.73 14.0 1059.0 8.98 3.8 99.7 9.23 9.0 103

22.5 22.3 2.7 98.9 22.6 3.8 10145 45.6 3.3 101 46.0 2.9 10290 89.5 2.5 99.4 91.6 1.7 102

225 221 3.9 98.1 225 3.9 99.9450 444 3.5 98.6 448 6.6 99.5900 896 1.3 99.5 904 0.9 100

Table 2.3: Mean back-calculated concentrations from calibration curves in human whole blood

Back-calculated concentrations from calibration curves constructed in human whole blood

Intra-day (n=6) T1+ T2+

Nominal conc. µg/kg

Mean back-calcul.

conc. µg/kg

R.S.D. (%)

Recovery (%)

Mean back-calcul.

conc. µg/kg

R.S.D. (%)

Recovery (%)

4.50 4.24 12.3 94.2 4.82 12.1 1079.0 8.60 4.9 95.7 9.07 11.3 101

18.0 17.6 5.5 97.5 17.4 9.6 96.545.0 44.8 3.6 99.4 40.5 7.0 90.290.0 93.9 13.2 104 90.9 9.7 101180 178 3.1 98.7 177 4.7 98.2450 462 6.4 103 456 6.8 101900 891 0.7 99.1 908 1.0 101

Page 44: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

For each analyte, the goodness of fit between back-calculated concentrations and nominal

concentrations was statistically evaluated (i) by comparing the regression line of back-calculated

versus nominal concentrations to the reference line (slope = 1 and intercept = 0); no significant

difference was observed; (ii) by studying the frequency distribution histogram of the residuals,

which were normally distributed and centered around zero, the number of positive and negative

values being approximately equal; and (iii) by comparing the bias to zero; a t-test showed that this

parameter was not statistically different from zero; moreover, the 95% confidence interval included

the zero value.

2.2.2. Retention times and specificity:

Figures 2.4 and 2.5 show typical chromatograms obtained from extracts of drug-free human

matrices spiked with the three analytes. Under the chromatographic conditions described above,

peaks were adequately separated. Observed retention times (n = 30) were 9.03 min (R.S.D. =

0.12%) for T1, 9.98 min (R.S.D. = 0.14%) for T2 and 5.11 min (R.S.D. = 0.09%) for the internal

standard T3.

The values of capacity factor (k’) were 3.1, 3.5 and 0.22 for T1, T2 and T3, respectively, and

selectivity factors were αT1, T2 = 1.13, αT3, T1 = 2.38. T1, T2 and T3 exhibited well-separated (R T1, T2

= 2.33, R T3, T1 = 9.12), narrow (wT1 = 0.25 min, wT2 = 0.23 min, wT3 = 0.18 min) and symmetrical

peaks under the chromatographic conditions described. The peak skew was evaluated using the

asymmetry coefficient As = b / a, where b is the distance from the center line of the peak to the back

slope and a is the distance from the center line of the peak to the front slope, both a and b being

measured at 10% of the total peak height. The asymmetry coefficient was 0.96 for T1, 0.87 for T2

and 0.90 for T3. The number of theoretical plates (calculated from the T3 peak) was approximately

23,600. The guard column was exchanged every 200 sample runs and the column was replaced

when the number of theoretical plates decreased below 10,000 (approximately after 1000 analyses).

44

Page 45: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

min5 6 7 8 9 10 11

0

200000

400000

600000

800000

A

min9.1

2

10.0

3

min

TIC 1

mi5 6 7 8 9 1 1

0

20000

40000

60000

80000

100000

120000

140000B3

21

TIC

mi5 6 7 8 9 1 1

0

20000

40000

60000

80000

3m/z 326SIM

0

20000

40000

60000

800002

m/z 312SIM

min5 6 7 8 9 10 11

SIM

2

m/z 31212000

1400010000

12000

800010000

6000

4000

2000

0

Figure 2.4: Typical chromatograms of blank human plasma spiked with T1 and T2 at the following concentrations: (A) 2.25 µg/L

(LLOQ); (B) 450 µg/L. Peak 1 = T3 (internal standard); peak 2 = T1 and peak 3 = T2 (TIC, total ion current; SIM, single ion monitoring).

min5 6 7 8 9 10 11

SIM17500

14000m/z 326

1500012000

12500

0

2500

5000

7500

100003 10000

5 6 7 8 9 1 1 mi

min5 6 7 8 9 10 11

SIM 1m/z 227.3

mi5 6 7 8 9 1 1

20000

40000

60000

800001

m/z 227.3SIM 14000

800000

12000

600000 10000

400000

200000

0

0

45

Page 46: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Figure 2.5: Typical chromatograms of blank human whole blood spiked with T1 and T2 at the following concentrations: (A) 4.5 µg/kg

(LLOQ); (B) 450 µg/kg. Peak 1 = T3 (internal standard); peak 2 = T1 and peak 3 = T2 (TIC, total ion current; SIM, single ion monitoring).

mi5 6 7 8 9 1 1

0

200

400

600

800

1000

1200

1400

mi5 6 7 8 9 1 1

0

10000

20000

30000

40000

50000

60000TIC

SIM

3

1

mi mi0

2 3

9.1 10.0

mi5 6 7 8 9 1 1

0

200

400

600

800

1000

1200

1400SIM

2

m/z 312

m/z 326

mi5 6 7 8 9 1 1

0

10000

20000

30000

40000

50000

60000SIM 1

m/z 227.3

A

min5 6 7 8 9 10 11

0

100000

200000

300000

400000

500000

600000

B1

3

2

TIC

min5 6 7 8 9 10 11

0

100000

200000

300000

400000

500000

600000

2

m/z 312

SIM

min5 6 7 8 9 10 11

0

100000

200000

300000

400000

500000

600000 3m/z 326

SIM

min5 6 7 8 9 10 11

0

100000

200000

300000

400000

500000

600000 1m/z 227.3

SIM

46

Page 47: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

The specificity of this method was demonstrated by representative chromatograms of blank

matrices (Figure 2.6), which indicated that each analyte was well resolved from the matrix

endogenous peaks.

Figure 2.6: Chromatograms of blank human matrices A) plasma, B) whole blood.

2.2.3. Precision, accuracy, extraction efficiency and LLOQ:

Results for intra-day and inter-day precision and accuracy are presented in Table 2.4. All

values of accuracy and precision were within recommended limits. For all matrices, the precision

values were less than 15%. The percent recoveries from the nominal values for accuracy were

between 92.7-111% for T1, and between 95.1-108% for T2.

For T1 and T2, the mean extraction recoveries were i) 87% (R.S.D. = 10%, n = 24) and 85%

(R.S.D. = 9.5%, n = 24) from human plasma, ii) 53% (R.S.D. = 14%, n = 24) and 56% (R.S.D. =

8.6%, n = 24) from human whole blood, respectively. They were not statistically different over the

range of concentrations studied. The extraction recovery of the internal standard was 89% (R.S.D. =

4.6%, n = 24) from human plasma and 51% (R.S.D. = 8%, n = 24) from human whole blood.

min5 6 7 8 9 1

ATIC

3 21

1

1750

1500

1250

1000

750

500

250

0

mi5 6 7 8 9 1 1

0

B

12

3

TIC 1400

1200

1000

800

600

400

200

47

Page 48: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Table 2.4: Precision and accuracy of the method

Accuracy and precision of the method in human plasma Intra-day (n=6)

T1+ T2+

Theoretical concentration

µg/L

Mean back-calculated

concentration µg/L

Precision (%)

Accuracy (%)

Mean back-calculated

concentration µg/L

Precision (%)

Accuracy (%)

3.75 3.96 8.0 106 3.90 8.6 104 11.2 12.4 4.9 111 12.12 10.4 108 112 114 5.8 102 111 9.6 99.1 675 684 6.3 101 647 9.9 95.9

Accuracy and precision of the method in human plasma Inter-day (n=6)

T1+ T2+

Theoretical concentration

µg/L

Mean back-calculated

concentration µg/L

Precision (%)

Accuracy (%)

Mean back-calculated

concentration µg/L

Precision (%)

Accuracy (%)

3.75 3.71 8.5 98.9 3.66 12.7 97.6 11.2 11.4 5.3 102 10.9 4.3 97.3 112 112 4.9 100 114 2.9 102 675 689 6.1 102 674 2.8 99.9

Accuracy and precision of the method in human whole blood Intra-day (n=6)

T1+ T2+

Theoretical concentration

µg/kg

Mean back-calculated

concentration µg/kg

Precision (%)

Accuracy (%)

Mean back-calculated

concentration µg/kg

Precision (%)

Accuracy (%)

7.5 7.45 9.0 99.3 7.57 7.2 101 22.5 21.3 12.3 94.7 21.4 9.8 95.1 225 214 7.6 95.1 215 11.0 95.6 600 577 9.0 96.2 591 9.2 98.5

Accuracy and precision of the method in human whole blood Inter-day (n=6)

T1+ T2+

Theoretical concentration

µg/kg

Mean back-calculated

concentration µg/kg

Precision (%)

Accuracy (%)

Mean back-calculated

concentration µg/kg

Precision (%)

Accuracy (%)

7.5 7.41 14.3 98.8 7.58 9.8 101 22.5 22.4 11.6 99.5 22.3 9.6 99.1 225 213 12.4 94.7 226 10.0 100 600 556 10.0 92.7 583 9.0 97.2

48

Page 49: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Based upon the analysis of low concentration replicate standards in each validation run, the

LLOQ value was 2.25 µg/L for both T1 and T2 in human plasma when using a 0.5 mL plasma

sample for extraction and a 10 µL injection volume for LC/ESI-MS analysis. In human whole

blood, the corresponding values were 4.5 µg/kg when using a 0.25 g whole blood for extraction and

the same injection volume. At these levels, the precision was <19.5% R.S.D. and accuracy was

89.8-107%.

2.2.4. Stability:

When stored at 4°C in the refrigerator for a period of 1 month, stock solutions of T1 and T2

did not reveal any appreciable degradation.

Human plasma and whole blood samples spiked with T1 and T2 were stored at room

temperature (20°C) or at 4°C for 6 h; no sign of decrease in the nominal starting concentration

(Table 2.5) was shown.

Both in human plasma and whole blood at least three freeze-thaw cycles can be tolerated

without losses higher than 10%. After the third freezing-thawing process, mean percent recoveries

ranged from 95.5 to 109% for T1 and from 94.7 to 113% for T2 (Table 2.5).

Frozen QC human plasma and whole blood (-20 and -80°C) samples, tested over 1 month

period, showed no sign of either degradation or losses (Table 2.5).

49

Page 50: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Table 2.5: Stability studies in human plasma and whole blood Stability studies in human plasma (n=3)

T1+ T2+Nominal conc.

µg/L Mean back-calcul. conc. µg/L

Precision (%)

Accuracy (%)

Mean back-calcul. conc. µg/L

Precision (%)

Accuracy (%)

After 6 h storage at 4°C 3.75 3.60 11.7 96.0 4.09 4.8 109 11.2 11.3 9.3 101 11.9 8.9 106 112 108 7.0 96.4 111 3.7 99.1 675 682 9.8 101 667 7.7 98.8

After 6 h storage at 20°C 3.75 3.86 4.2 103 4.13 4.1 110 11.2 10.5 3.9 93.7 11.3 3.0 101 112 115 8.9 103 114 9.9 102 675 694 9.2 103 659 7.5 97.6

After three freeze/thaw cycles 3.75 3.58 11.3 95.5 4.18 2.2 111 11.2 12.2 2.4 109 12.6 1.1 113 112 116 7.1 104 122 1.7 109 675 694 12.1 103 656 7.7 97.2

After 1 month storage at -20°C 3.75 3.97 15.6 106 3.78 4.3 101 11.2 11.3 7.3 101 11.0 8.5 98.2 112 115 1.2 103 116 4.0 104 675 699 9.7 104 702 3.6 104

After 1 month storage at -80°C 3.75 3.77 8.6 101 4.22 5.4 113 11.2 11.8 2.7 105 12.1 2.9 108 112 115 8.1 103 117 9.6 104 675 710 5.4 105 709 7.2 105

Stability studies in human whole blood (n=3)T1+ T2+

Nominal conc. µg/kg Mean back-calcul.

conc. µg/kg Precision

(%) Accuracy

(%) Mean back-calcul.

conc. µg/kg Precision

(%) Accuracy

(%) After 6 h storage at 4°C

7.5 7.97 8.0 106 7.82 6.8 104 22.5 23.1 1.6 103 21.9 9.8 97.3 225 234 11 104 204 6.6 90.7 600 639 6.2 107 593 9.6 98.8

After 6 h storage at 20°C 7.5 7.16 7.3 95.4 7.66 8.1 102

22.5 20.5 4.5 91.0 22.2 8.3 98.6 225 210 6.1 93.3 201 0.6 89.3 600 584 6.4 97.3 571 4.4 95.1

After three freeze/thaw cycles 7.5 8.16 4.5 109 7.99 3.9 107

22.5 23.9 4.8 106 23.8 8.5 106 225 228 6.8 101 213 5.5 94.7 600 649 6.3 108 649 5.9 108

After 1 month storage at -20°C 7.5 7.68 5.1 102 6.94 11.0 92.5

22.5 21.5 10.0 95.6 22.4 2.8 99.6 225 230 13.5 102 226 11.2 100 600 591 2.9 98.5 622 1.6 104

After 1 month storage at -80°C 7.5 7.86 7.3 105 7.12 7.8 94.9

22.5 20.6 11.5 91.6 19.4 14.4 86.2 225 223 4.5 99.1 227 1.1 101 600 596 5.0 99.3 653 3.5 109

50

Page 51: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

2.2.5 Ion suppression / enhancement study:

The results of ion suppression / enhancement study for T1, T2 and the internal standard T3 are

presented in Table 2.6.

Table 2.6: Data of ion suppression/enhancement study

Ion suppression / enhancement

T1+ T2+Theoretical concentration Response (%) * CV (%) Response (%)* CV (%)

In human plasma (n=12 per concentration level) 3.75 µg/L 104 2.2 92.8 4.8 11.2 µg/L 107 1.1 104 1.1 112 µg/L 105 2.2 93.7 2.1 675 µg/L 106 2.2 97.0 0.8

In human whole blood (n=12 per concentration level) 7.5 µg/kg 102 3.0 93.5 5.9 22.5 µg/kg 110 2.5 92.8 3.8 225 µg/kg 102 3.2 102 4.5 600 µg/kg 94.5 1.5 99.3 4.0 * [peak area extract / peak area reference] x 100

T3++: 93% (CV 5%) (n=48) in human plasma and 113% (CV 6%) (n=48) in human whole

blood.

These findings confirmed that human plasma and whole blood had no influence on the

detection of T1, T2 and the internal standard T3.

2.3. Results of partial validation in rat plasma and RBCs

Only the results of partial validation carried out in rat RBCs are reported below.

2.3.1. Drug / response relationship:

Mean parameters of the quadratic equation, used to link peak area ratios (analyte / internal

standard) and analyte concentrations, are presented in Table 2.7.

Table 2.7: Mean parameters of the quadratic equation

Analyte a (mean) ± S.D. b (mean) c (mean) ± S.D.

Rat RBCs (n=7)

T1 -6.43 x 10-8 ± 9.75 x 10-7 0.00396 (R.S.D., 7.5%) 0.0113 ± 0.0147 Inter-day

T2 -1.45 x 10-6 ± 1.52 x 10-6 0.00860 (R.S.D., 7.6%) -0.0264 ± 0.0437

Mean back-calculated concentrations and percent recovery are reported in Table 2.8.

51

Page 52: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

Table 2.8: Back-calculated concentrations from calibration curves performed in rat

RBCs

Back-calculated concentrations from calibration curves constructed in rat RBCs

Inter-day (n=7)

T1+ T2+Nominal conc.

µg/kg R.S.D. (%) Recovery (%) R.S.D. (%) Recovery (%) 4.50 8.4 108 18.1 101 9.0 10.2 97.5 10.8 101

18.0 12.6 96.2 11.3 102 45.0 10.9 100 8.8 104 90.0 8.5 92.3 11.5 96.8 180 9.7 102 13.2 93.3 450 7.8 106 4.4 100 900 7.5 95.5 1.2 101

From 9 to 900 µg/kg, R.S.D. values did not exceed 15% (1.2 to 13.2%) and recoveries were

between 93.3 and 106%. At the lowest concentrations (4.5 µg/kg), R.S.D. values were 8.4% for T1+

and 18.1% for T2+. The good adjustment between back-calculated and added concentrations has

been statistically proven: (i) a linear regression of the back-calculated concentrations versus the

nominal ones provided a unit slope and an intercept equal to 0; (ii) the distribution of the residuals

showed random variations, the number of positive and negative values being approximately equal;

and (iii) residuals were normally distributed and centred around zero.

2.3.2. Retention times and specificity:

The specificity of the method was demonstrated by representative chromatograms of blank rat

matrix (Figure 2.7). Each analyte was well resolved from the matrix endogenous peaks.

Figure 2.8 shows typical chromatograms obtained from extracts of drug-free RBCs spiked

with the three analytes at 4.5 µg/kg (LLOQ) and 450 µg/kg.

3 2

1

TIC

Figure 2.7: Chromatogram of a blank rat RBCs.

52

Page 53: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

1 SIM m/z 227

SIM

SIM

TIC

m/z 326

m/z 312

3

2

2 3

1

1 SIM m/z 227

SIM

SIM

TIC

m/z 326

m/z 312

3

2

2 3

1 A

2

SIMm/z 312

3SIM m/z 326

1SIM

m/z 227

31

2

TIC B

2

SIMm/z 312

3SIM m/z 326

1SIM

m/z 227

31

2

TIC

2

SIMm/z 312

3SIM m/z 326

2

SIMm/z 312

2

SIMm/z 312

3SIM m/z 326

3SIM m/z 326

1SIM

m/z 2271SIM

m/z 227

31

2

31

2

TIC B

Figure 2.8: Typical chromatograms of blank rat RBCs spiked with T1 and T2 at the following concentrations: (A) 4.5 µg/kg; (B) 450

µg/kg. Peak 1 = T3 (internal standard); peak 2 = T1 and peak 3 = T2 (TIC, total ion current; SIM, single ion monitoring).

53

Page 54: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

2.3.3. Precision, accuracy, extraction efficiency and LLOQ:

Inter-day precision and accuracy of the method are presented in Table 2.9. Obtained values for

accuracy (97.8-103% for T1 and 98.7-102% for T2) and precision (<10.4%) are in agreement with

the recommendation limits [103].

Table 2.9: Precision and accuracy of the method Accuracy and precision of the method in rat RBCs

Inter-day (n=7)

T1+ T2+Nominal conc.

µg/kg Precision (%) Accuracy (%) Precision (%) Accuracy (%) 7.5 10.4 101 10.0 102

22.5 10.0 97.8 9.1 98.7 225 6.8 103 8.5 101 600 9.0 98.8 9.4 101

The mean extraction recoveries from rat RBCs were 53.5% (R.S.D., 9.2%, n = 24) and 55.3%

(R.S.D., 9.4%, n = 24) for T1 and T2, respectively. The mean extraction recovery of the internal

standard T3 was 51.8% (R.S.D., 8.7%, n = 24).

The LLOQ in rat RBCs was 4.5 µg/kg for both T1 and T2 when using 0.25 g of RBCs for the

extraction and 10 µL of the reconstituted extract for the injection onto LC/ESI-MS.

2.3.4. Stability:

QC samples of rat RBCs (i) stored at room temperature (20°C) or at 4°C for 6 h, (ii) stored

frozen at -20 and -80°C and tested over a span of one month or (iii) frozen-and-thawed for three

times, showed no sign of either degradation or losses >15%.

2.3.5 Ion suppression / enhancement study:

In rat RBCs, the peak area ratios of the reconstituted extracts and the reference solutions

ranged from 0.9 to 1.1. This ratio is close to 1, therefore matrix had no effect on the detection of the

analytes.

2.4. Results of a pharmacokinetic study carried out after intravenous administration of

T2 in rat

Semilogarithmic plots of the mean (± SD) T2 plasma and RBC concentration-time profiles

after intravenous administration (2.1 mg/kg) of T2 in rat are shown in Figure 2.9.

54

Page 55: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

0.001

0.010

0.100

1.000

10.000

0 2 4 6 8 10

Time (hours)

T2 mg/L

Plasma RBCs

Figure 2.9: Mean (SD; error bars) plasma and RBCs concentration-time curves after

single intravenous administration of 2.1 mg/kg T2 in rat.

Five minutes after intravenous administration of T2, concentrations were 0.332 ± 0.056 mg/L

in rat plasma and 2.05 ± 0.19 mg/kg in rat RBCs. Data were consistent with a two-compartment

model following first order kinetics. The half-lives of the terminal part of the curves were 2.33 h

from plasma data and 2.62 h from RBC data. The total plasma clearance was 10 L/h/kg and the

volume of distribution at steady state was 33.8 L/kg. The RBC/plasma AUC ratio was 7.9.

In opposite to the bisquaternary ammonium salts, T2 accumulated strongly within

unparasitized RBCs. In rat, T1 was the main metabolite of T2.

55

Page 56: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

CONCLUSION

Liquid chromatography / electrospray ionisation – mass spectrometry methods with good

precision and accuracy have been developed for the simultaneous determination of two antimalarial

monothiazolium compounds, T1 and T2, in biological matrices. Full validation of T1 and T2 in

human plasma and whole blood as well as partial validation in rat plasma and RBCs have been

carried out. Distinct advantages of the method include simplicity and rapidity of sample

preparation, good resolution between the analytes, high sensitivity, reliability and specificity.

This highly sensitive, accurate and precise method was suitable to analyse plasma and RBC

samples, collected during preclinical pharmacokinetic study carried out after intravenous

administration of T2 in rat.

The study of monothiazolium compounds in the context of developmental work of

Plasmodium phospholipid metabolism inhibitors is definitely significant. Strong accumulation of

T2 in unparasitized rat RBCs indicate that further development of this potential class of antimalarial

drugs is needed.

56

Page 57: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

REFERENCES 1. Ackermann BL, Berna MJ, Murphy AT. Recent Advances in use of LC/MS/MS for

Quantitative High-Throughput Bioanalytical Support of Drug Discovery. Current Topics in Medicinal Chemistry 2002, 2, 53-66.

2. Alleva LM and Kirk K. Calcium regulation in the intraerythrocytic malaria parasite Plasmodium falciparum. Molecular & Biochemical Parasitology 117 (2001) 121–128.

3. Ancelin ML, Calas M, Bompart J, Cordina G, Martin D, Bari MB et al. Antimalarial Activity of 77 Phospholipid Polar Head Analogs: Close Correlation Between Inhibition of Phospholipid Metabolism and In Vitro Plasmodium Falciparum Growth. Blood, Vol 91, No 4 (February 15), 1998: pp 1426-1437.

4. Ancelin ML, Calas M, Bonhoure A, Herbute S, Vial HJ. In Vivo Antimalarial Activities of Mono- and Bis Quaternary Ammonium Salts Interfering with Plasmodium Phospholipid Metabolism. Antimicrobial Agents and Chemotherapy, Aug. 2003, Vol. 47, No. 8, p. 2598–2605.

5. Ancelin ML, Calas M, Vidal-Sailhan V, Herbute S, Ringwald P, Vial HJ. Potent Inhibitors of Plasmodium Phospholipid Metabolism with a Broad Spectrum of In Vitro Antimalarial Activities. Antimicrobial Agents and Chemotherapy, Aug. 2003, Vol. 47, No. 8, p. 2590–2597.

6. Ardrey RE. Liquid Chromatography –Mass Spectrometry: An Introduction. John Wiley & Sons, Ltd., 2003, pp. 98-122.

7. Ashley EA, Lwin KM, McGready R, Simon WH, Phaiphun L, Proux S et al. An opel label randomized comparison of mefloquine-artesunate as separate tablets vs. a new co-formulated combination for the treatment of uncomplicated multidrug-resistant falciparum malaria in Thailand. Tropical Medicine and International Health, Nov 2006, Vol.11 No.11, pp. 1653-1660.

8. Baggish AL and Hill DR. Antiparasitic agent atovaquone. Antimicrobial Agents and Chemotherapy, May 2002, Vol.46, No.5, p. 1163-1173.

9. Baldwin J, Farajallah AM, Malmquist NA, Rathod PK, Phillips MA. Malarial dihydroorotate dehydrogenase. The Journal of Biological Chemistry, November 1, 2002, Vol. 277, No. 44, pp. 41827–41834.

10. Baldwin J, Michnoff CH, Malmquist NA, White J, Roth MG, Rathod PK et al. High-throughput screening for potent and selective inhibitors of Plasmodium falciparum dihydroorotate dehydrogenase. The Journal of Biological Chemistry, June 10, 2005, Vol. 280, No. 23, pp. 21847–21853.

11. Barnes KI, White NJ. Population biology and antimalrial resistance: the transmission of antimalarial drug resistance in Plasmodium falciparum. Acta Tropica 94 (2005) 230-240.

12. Baumeister S, Endermann T, Charpian S, Nyalwidhe J, Duranton C, Huber S et al. A biotin derivative blocks parasite induced novel permeation pathways in Plasmodium falciparum-infected erythrocytes. Molecular & Biochemical Parasitology 132 (2003) 35–45.

13. Becker K and Kirk K. Of malaria, metabolism and membrane transport. Trends in Parasitology, December 2004, Vol.20 No.12, pp. 590-596.

14. Biagini GA, O’Neill PM, Nzila A, Ward SA, Bray PG. Antimalarial chemotherapy: young guns or back to the future? Trends in Parasitology November 2003, Vol.19 No.11. 479-487.

15. Biagini GA, Richier E, Bray PG, Calas M, Vial H, Ward SA. Heme Binding Contributes to Antimalarial Activity of Bis-Quaternary Ammoniums. Antimicrobial Agents and Chemotherapy, Aug. 2003, Vol. 47, No. 8, p. 2584–2589.

16. Biagini GA, Pasini EM, Hughes R, De Koning HP, Vial HJ, O’Neill PM et al. Characterization of the choline carrier of Plasmodium falciparum: a route for the selective delivery of novel antimalarial drugs. Blood, 15 Nov 2004, Vol. 104, No. 10, p.3372-3377.

17. Biagini GA, O’Neill PM, Bray PG, Ward SA. Current drug development portfolio for antimalarial therapies. Current Opinion in Pharmacology 2005, 5:473-478.

18. Biagini GA, Ward SA, Bray PG. Malaria parasite transporters as a drug-delivery strategy. Trends in Parasitology, July 2005, Vol.21 No.7, pp. 299-301.

57

Page 58: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

19. Bissati KE, Zufferey R, Witola WH, Carter NS, Ullman B, Mamoun CB. The plasma membrane permease PfNT1is essential for purine salvage in the human malaria parasite Plasmodium falciparum. PNAS, June 13, 2006, Vol.103, No. 24, 9286–9291.

20. Boa AN, Canavan SP, Hirst PR, Ramsey C, Stead AMW, McConkey GA. Synthesis of brequinar analogue inhibitors of malaria parasite dihydroorotate dehydrogenase. Bioorganic & Medicinal Chemistry 13 (2005) 1945–1967.

21. Bokman CF. Analytical Aspects of Atmospheric Pressure Ionization in Mass Spectrometry. Acta Universitatis Upsaliensis. Comprehensive Summaries of Uppsala Dissertations from the Faculty of Science and Technology 748. 2002, 48 pp.

22. Breman JG, Alilio MS, Mills A. Conquering the intolerable burden of malaria: what’s new, what’s needed: a summary. Am. J. Trop. Med. Hyg., 71 (Suppl 2), 2004, pp. 1-15.

23. Bressolle F, Bromet-Petit M, Audran M. Validation of LC and GC methods. Applications to pharmacokinetics. Journal of Chromatography B, 686 (1996) 3-10.

24. Careri M, Mangia A. Validation and qualification: the fitness for purpose of mass spectrometry-based analytical methods and analytical systems. Anal Bioanal Chem (2006) 386: 38–45.

25. Cech NB and Enke CG. Practical implications of some recent studies in electrospray ionization fundamentals. Mass Spectrometry Reviews, 2001, 20, 362– 387.

26. Cunha-Rodrigues M, Prudencio M, Mota MM, Haas W. Antimalarial drugs – host targets (re)visited. Biotechnol. J. 2006, 1, 321-332.

27. Dadgar D, Burnett PE, Choc MG, Gallicano K, Hooper JW. Application issues in bioanalytical method validation, sample analysis and data reporting. Journal of Pharmaceutical and Biomedical Analysis, 1995, Vol. 13, No. 2, pp.89-97.

28. Daily JP. Antimalarial drug therapy : the role of parasite biology and drug resistance. J Clin Pharmacol 2006;46:1487-1497.

29. Dams R, Huestis MA, Lambert WE, Murphy CM. Matrix Effect in Bio-Analysis of Illicit Drugs with LC-MS/MS: Influence of Ionization Type, Sample Preparation, and Biofluid. J Am Soc Mass Spectrom 2003, 14, 1290–1294.

30. Di Tullio A, Reale S, De Angelis F. Molecular recognition by mass spectrometry. J. Mass Spectrom. 2005; 40: 845–865.

31. Drakeley C, Sutherland C, Bousema JT, Sauerwein RW, Targett GAT. The epidemiology of Plasmodium falciparum gametocytes : weapons of mass dispersion. Trends in Parasitology 2006, Vol.22 No.9. 424-430.

32. Elandalloussi LM, Adams B, Smith PJ. ATPase activity of purified plasma membranes and digestive vacuoles from Plasmodium falciparum. Molecular & Biochemical Parasitology 141 (2005) 49–56.

33. Ersmark K, Samuelsson B, Hallberg A. Plasmepsins as potential targets for new antimalarial therapy. Medicinal Research Reviews 2006, Vol.26, No.5, 626-666.

34. Farenc C, Fabreguette JR, Bressolle F. Pk-fit: a pharmacokinetic/pharmacodynamic and statistical data analysis software. Comput. Biomed. Res. 2000, 33:315-330.

35. Fitch CD. Ferriprotoporphyrin IX, phospholipids, and the antimalarial actions of quinoline drugs. Life Sciences 74 (2004) 1957-1972.

36. Fitch CD, Russell NV. Accelerated denaturation of hemoglobin and the antimalarial action of chloroquine. Antimicrobial Agents and Chemotherapy, July 2006, Vol.50, No.7, p. 2415-2419.

37. Foley M and Tilley L. Quinoline antimalarials: mechanisms of action and resistance and prospects for new agents. Pharmacol. Ther. 1998, Vol.79, No.1, pp. 55-87.

38. Geyer JA, Prigge ST, Waters NC. Targeting malaria with specific CDK inhibitors. Biochimica et Biophysica Acta (2005) 1754 160 – 170.

39. Go ML. Novel antiplasmodial agents. Medicinal Research Reviews, 2003, Vol. 23, No. 4, 456-487.

58

Page 59: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

40. Go ML, Liu M, Wilairat P, Rosenthal PJ, Saliba KJ, Kirk K. Antiplasmodial chalcones inhibit sorbitol-induced hemolysis of Plasmodium falciparum-infected erythrocytes. Antimicrobial Agents and Chemotherapy, Sept. 2004, Vol. 48, No. 9, p. 3241–3245.

41. Golenser J, Waknine JH, Krugliak M, Hunt NH, Grau GE. Current perspectives on the mechanism of action of artemisinins. International Journal for Parasitology 36 (2006) 1427-1441.

42. Gregson A and Plowe CV. Mechanisms of resistance of malaria parasites to antifolates. Pharmacol Rev 2005, 57:117-145.

43. Haemers T, Wiesner J, Poecke SV, Goeman J, Henschker D, Beck E, Jomaa H, Calenbergh SV. Synthesis of a-substituted fosmidomycin analogues as highly potent Plasmodium falciparum growth inhibitors. Bioorganic & Medicinal Chemistry Letters 16 (2006) 1888–1891.

44. Happi CT, Gbotosho GO, Folarin OA, Milner D, Sarr O, Sowunmi A et al. Confirmation of emergence of mutations associated with atovaquone-proguanil resistance in unexposed Plasmodium falciparum isolates from Africa. Malaria Journal 2006, 5:82.

45. Hartmann C, Smeyers-Verbeke J, Massart DL, McDowall RD. Validation of bioanalytical chromatographic methods. Journal of Pharmaceutical and Biomedical Analysis 17 (1998) 193–218.

46. Hayward R, Saliba KJ, Kirk K. Mutations in pfmdr1 modulate the sensitivity of Plasmodium falciparum to the intrinsic antiplasmodial activity of verapamil. Antimicrobial Agents and Chemotherapy, Feb 2005, Vol.49, No.2, p. 840-842.

47. Heikkila T, Thirumalairajan S, Davies M, Parsons MR, McConkey AG, Fishwick CWG, Johnson AP. The first de novo designed inhibitors of Plasmodium falciparum dihydroorotate dehydrogenase. Bioorganic & Medicinal Chemistry Letters 16 (2006) 88–92.

48. Hekmat-Nejad M and Rathod PK. Kinetics of Plasmodium falciparum Thymidylate Synthase: Interactions with High-Affinity Metabolites of 5-Fluoroorotate and D1694. Antimicrobial Agents and Chemotherapy, July 1996, Vol. 40, No. 7, p. 1628–1632.

49. Hsu E. Reflections on the ‘discovery’ of the antimalarial qinghao. Br J Clin Pharmacol (2006) 61:6, 666-670.

50. Hwang J, Bitarakwate E, Pai M, Reingold A, Rosenthal PJ, Dorsey G. Chloroquine or amodiaquine combined with sulfadoxine-pyrimethamine for uncomplicated malaria: a systematic review. Tropical Medicine and International Health, June 2006, Vol.11 No.6, pp. 789-799.

51. ICH Harmonised Tripartite Guideline. Validation of Analytical Procedures: Text and Methodology Q2(R1). November, 2005.

52. Jemal M. High-throughput quantitative bioanalysis by LC/MS/MS. Biomed. Chromatogr. (2000) 14: 422–429.

53. Kapadia GJ, Azuine MA, Balasubramanian V, Sridhar R. Aminonaphthoquinones – a novel class of compounds with potent antimalarial activity against Plasmodium falciparum. Pharmacological Research, 2001, Vol. 43, No. 4, p. 363-367.

54. Karnes HT, Shiu G, Shah VP. Validation of bioanalytical methods. Pharm. Res., (1991) Apr; 8(4): 421-6.

55. Kettler K, Wiesner J, Silber K, Haebel P, Ortmann R, Sattler I et al. Non-thiol farnesyltransferase inhibitors: N-(4-aminoacylamino-3-benzoylphenyl)-3-[5-(4-nitrophenyl)-2 furyl]acrylic acid amides and their antimalarial activity. European Journal of Medicinal Chemistry 40 (2005) 93–101.

56. Kirk, K. Membrane transport in the malaria-infected erythrocyte. Physiological Reviews 2001, Vol.81, No.2.

57. Kirk K. Channels and transporters as drug targets in the Plasmodium-infected erythrocyte. Acta Tropica 89 (2004) 285–298.

58. Krause T, Ersen KL, Wrenger C, Gilberger TW, Ller SM, Walter RD. The ornithine decarboxylase domain of the bifunctional ornithine decarboxylase/S-adenosylmethionine decarboxylase of Plasmodium falciparum : recombinant expression and catalytic properties of two different constructs. Biochem. J. (2000) 352, 287-292.

59

Page 60: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

59. Krishna S, Eckstein-Ludwig U, Joet T, Uhlemann AC, Morin C, Webb R et al. Transport processes in Plasmodium falciparum-infected erythrocytes: potential as new drug targets. International Journal for Parasitology 32 (2002) 1567–1573.

60. Lagerwerf FM, Dongen WD, Steenvoorden RJJM, Honing M, Jonkman JHG. Exploring the boundaries of bioanalytical quantitative LC-MS-MS. Trends in analytical chemistry, 2000, vol. 19, no. 7, pp.418-427.

61. Larvor MP, Cerdan R, Gumila C, Maurin L, Seta P, Roustan C et al.. Characterization of the lipid-binding domain of the Plasmodium falciparum CTP:phosphocholine cytidylyltransferase through synthetic-peptide studies. Biochem. J. (2003) 375, 653–661.

62. Laurent D, Jullian V, Parenty A, Knibiehler M, Dorin D, Schmitt S et al. Antimalarial potential of xestoquinone, a protein kinase inhibitor isolated from a Vanuatu marine sponge Xestospongia sp. Bioorganic & Medicinal Chemistry 14 (2006) 4477–4482.

63. Le Bras J, Musset L, Clain J. Antimalarial drug resistance. Medecine et maladies infectieuses 36 (2006) 401-405.

64. Lell B and Kremsner PG. Clindamycin as an antimalarial drug: review of clinical trials. Antimicrob Agents Chemother. Aug 2002, Vol.46 No.8, p. 2315-2320.

65. Lell B, Ruangweerayut R, Wiesner J, Missinou MA, Schindler A, Baranek T et al. Fosmidomycin, a novel chemotherapeutic agent for malaria. Antimicrobial Agents and Chemotherapy, Feb. 2003, Vol. 47, No. 2, p. 735–738.

66. Liu C, Zhao Y, Wang Y. Artemisinin: current state and perspectives for the biotechnological production of antimalarial drug. Appl Microbiol Biotechnol (2006) 72: 11-20.

67. Longo M, Zanoncelli S, Torre PD, Riflettuto M, Cocco F, Pesenti M et al. In vivo and in vitro investigations of the effects of the antimalarial drug dihydroartemisinin (DHA) on rat embryos. Reproductive Toxicology 22 (2006) 797-810.

68. Macreadie I, Ginsburg H, Sirawaraporn W, Tilley L. Antimalarial drug development and new targets. Parasitology Today 2000, Vol.16, No.10. 438-444.

69. Majori G. Combined antimalarial therapy using artemisinin. Parassitologia, June 2004, 46(1-2):85-7 (abstract).

70. Massimine KM, McIntosh MT, Doan LT, Atreya CE, Gromer S, Sirawaraporn W et al. Eosin B as a novel antimalarial agent for drug-resistant Plasmodium falciparum. Antimicrobial Agents and Chemotherapy, Sept. 2006, Vol. 50, No. 9, p. 3132–3141.

71. Matuszewski BK, Constanzer ML, Chavez-Eng CM. Matrix effect in quantitative LC/MS/MS analyses of biological fluids: a method for determination of finasteride in human plasma at picogram per milliliter concentrations. Anal Chem, 1998 Mar 1; 70(5):882-9.

72. Matuschewski K. Vaccine development against malaria. Current Opinion in Imunology 2006, 18:449-457.

73. Matuszewski BK. Standard line slopes as a measure of a relative matrix effect in quantitative HPLC–MS bioanalysis. Journal of Chromatography B, 830 (2006) 293–300.

74. Mei H, Hsieh Y, Nardo C, Xu X, Wang S, Ng K, Korfmacher WA. Investigation of matrix effects in bioanalytical high-performance liquid chromatography/tandem mass spectrometric assays: application to drug discovery. Rapid Commun. Mass Spectrom. 2003; 17: 97-103.

75. Meierjohann S, Walter RD, Muller S. Regulation of intracellular glutathione levels in erythrocytes infected with chloroquine-sensitive and chloroquine-resistant Plasmodium falciparum. Biochem. J. (2002) 368, 761-768.

76. Missinou MA, Borrmann S, Schindler A, Issifou S, Adegnika AA, Matsiegui PB et al. Fosmidomycin for malaria. Lancet 2002; 360: 1941–42.

77. Mitamura T and Palacpac NMQ. Lipid metabolism in Plasmodium falciparum-infected erythrocytes: possible new targets for malaria chemotherapy. Microbes and Infection 5 (2003) 545–552.

78. Mugittu K, Genton B, Mshinda H, Beck HP. Molecular monitoring of Plasmodium falciparum resistance to artemisinin in Tanzania. Malaria Journal 2006, 5:126

60

Page 61: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

79. National Institutes of Health. Principles of laboratory animal care, revised. NIH publication 1985, 85-23. Bethesda, MD: NIH.

80. Nicolas O, Margout D, Taudon N, Wein S, Calas M, Vial HJ, Bressolle F. Pharmacological Properties of a New Antimalarial Bisthiazolium Salt, T3, and a Corresponding Prodrug, TE3. Antimicrobial Agents and Chemotherapy, Sept. 2005, Vol. 49, No. 9, p. 3631–3639.

81. Olliaro PL and Yuthavong Y. An overview of chemotherapeutic targets for antimalarial drug discovery. Pharmacol. Ther. 1999, Vol. 81, No. 2, pp. 91–110.

82. Olliaro P. Mode of action and mechanisms of resistance for antimalarial drugs. Pharmacology and Therapeutics 89 (2001) 207-219.

83. Olliaro PL, Haynes RK, Meunier B, Yuthavong Y. Possible modes of action of the artemisinin-type compounds. Trends in Parasitology, March 2001, Vol. 17, No. 3, pp. 122-126.

84. Pattanaik P, Raman J, Balaram H. Perspectives in drug design against malaria. Current Topics in Medicinal Chemistry 2002, 2, 483-505.

85. Pradines B, Spiegel A, Rogier C, Tall A, Mosnier J, Fusal T et al. Antibiotics for prophylaxis of Plasmodium falciparum infections: in vitro activity of doxycycline against Senegalese isolates. Am. J. Trop. Med. Hyg., 62(1), 2000, pp. 82-85.

86. Ramanitrahasimbola D, Rasaonaivo P, Ratsimamanga S, Vial H. Malagashanine potentiates chloroquine antimalarial activity in drug resistant Plasmodium malaria by modifying both its efflux and influx. Molecular & Biochemical Parasitology 146 (2006) 58-67.

87. Ramstrom M. Analysis of complex biological samples using liquid chromatography-fourier transform ion cyclotron resonance mass spectrometry. Acta Universitatis Upsaliensis. Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Science and Technology 31. 2005, 62 pp.

88. Ramya TNC, Surolia N, Surolia A. Polyamine synthesis and salvage pathways in the malaria parasite Plasmodium falciparum. Biochemical and Biophysical Research Communications 348 (2006) 579–584.

89. Ramya TNC, Mishra S, Karmodiya K, Surolia N, Surolia A. Inhibitors of non-housekeeping functions of the apicoplast defy ‘delayed death’ in Plasmodium falciparum. Antimicrob Agents Chemother. Jan 2007, Vol.51 No.1, p. 307-316.

90. Razakantoanina V, Phung NKP, Jaureguiberry G. Antimalarial activity of new gossypol derivatives. Parasitol Res, 2000, 86: 665-668.

91. Richier E, Biagini GA, Wein S, Boudou F, Bray PG, Ward SA et al. Potent Antihematozoan Activity of Novel Bisthiazolium Drug T16: Evidence for Inhibition of Phosphatidylcholine Metabolism in Erythrocytes Infected with Babesia and Plasmodium spp. Antimicrobial Agents and Chemotherapy, Oct. 2006, Vol. 50, No. 10, p. 3381–3388.

92. Roberts CW, Roberts F, Lyons RE, Kirisits MJ, Mui EJ, Finnerty J et al. The Shikimate pathway and its branches in Apicomplexan parasites. The Journal of Infectious Diseases 2002;185(Suppl 1):S25–36.

93. Rosenthal PJ. Antimalarial drug discovery: old and new approaches. The Journal of Experimental Biology 206 (2003) 3735-3744.

94. Saliba KJ, Krishna S, Kirk K. Inhibition of hexose transport and abrogation of pH homeostasis in the intraerythrocytic malaria parasite by an O-3-hexose derivative. FEBS Letters 570 (2004) 93–96.

95. Salom-Roig XJ, Hamze A, Calas M, Vial HJ. Dual Molecules as New Antimalarials: Combinatorial Chemistry & High Throughput Screening, Volume 8, Number 1, February 2005, pp. 49-62(14) (abstract).

96. Shah VP, Midha KK, Findlay JWA, Hill HM, Hulse JD, McGilveray IJ et al. Bioioanalytical Method Validation - A Revisit with a Decade of Progress. Pharmaceutical Research, 2000, Vol. 17, No. 12, pp. 1551-1557.

97. Singh N, Sijwali PS, Pandey KC, Rosenthal PJ. Plasmodium falciparum: Biochemical characterization of the cysteine protease falcipain-2. Experimental Parasitology 112 (2006) 187–192.

61

Page 62: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

98. Srivastava IK and Vaidya AB. A mechanism for the synergistic antimalarial action of atovaquone and proguanil. Antimicrob. Agents and Chemoth., June 1999, Vol.43, No.6, p. 1334-1339.

99. Staines HM, Dee BC, O’Brien M, Lang HJ, Englert H, Horner HA, Ellory JC, Kirk K. Furosemide analogues as potent inhibitors of the new permeability pathways of Plasmodium falciparum-infected human erythrocytes. Molecular & Biochemical Parasitology 133 (2004) 315–318.

100. Sullivan DJ. Theories on malarial pigment formation and quinoline action. International Journal for Parasitology 32 (2002) 1645-1653.

101. Taylor PJ. Matrix effects: The Achilles heel of quantitative high performance liquid chromatography–electrospray–tandem mass spectrometry. Clin. Bioch. 38 (2005) 328– 334.

102. Ting LM, Shi W, Lewandowicz A, Singh V, Mwakingwe A, Birck MR et al. Targeting a novel Plasmodium falciparum purine recycling pathway with specific immucillins. The Journal of Biological Chemistry, March 11, 2005, Vol. 280, No. 10, pp. 9547–9554.

103. US Food and Drug Administration. Guidance for industry. Bioanalytical method validation. May 2001. http://www.fda.gov/cder/guidance/index.htm (Accessed May 2004).

104. Vial HJ, Eldin P, Tielens AGM, Hellemond JJ. Phospholipids in parasitic protozoa. Molecular & Biochemical Parasitology 126 (2003) 143–154.

105. Vial HJ, Wein S, Farenc C, Kocken C, Nicolas O, Ancelin ML et al. Prodrugs of bisthiazolium salts are orally potent antimalarials. PNAS, October 26, 2004, Vol. 101, No. 43, 15458–15463.

106. WHO. (2006) Guidelines for the treatment of malaria. 107. Wiesner J, Henschker D, Hutchinson DB, Beck E, Jomaa H. In vitro and in vivo synergy

of fosmidomycin, a novel antimalarial drug, with clindamycin. Antimicrobial Agents and Chemotherapy, Sept. 2002, Vol. 46, No. 9, p. 2889–2894.

108. Wiesner J, Ortmann R, Jomaa H, Schlitzer M. New antimalarial drugs. Angew. Chem. Int. Ed. 2003, 42, 5274-5293.

109. Wiesner J, Borrmann S, Jomaa H. Fosmidomycin for the treatment of malaria. Parasitol Res (2003) 90: S71–S76.

110. Wiesner J, Kettler K, Sakowski J, Ortmann R, Katzin AM, Kimura EA et al. Farnesyltransferase inhibitors inhibit the growth of malaria parasites in vitro and in vivo. Angew. Chem. Int. Ed. 2004, 43, 251 –254.

111. Winstanley PA. Chemotherapy for falciparum malaria: the armoury, the problems and the prospects. Parasitology Today 2000, Vol.16, No.4.

112. Winstanley P. Modern chemotherapeutic options for malaria. Lancet Infectious Diseases 2001; 1: 242-50.

113. Winstanley P. The contribution of clinical pharmacology to antimalarial drug discovery and development. Br J Clin Pharmacol (2003) 55, 464-468.

114. Woodrow CJ, Haynes RK, Krishna S. Artemisinins. Postgrad. Med. J. 2005;81;71-78. 115. Woodrow CJ and Krishna S. Antimalarial drugs: recent advances in molecular

determinants of resistance and their clinical significance. Cell. Mol. Life Sci. 63 (2006) 1586-1596. 116. Yeung S and White NJ. How do patients use antimalarials drugs? A review of the

evidence. Tropical Medicine and International Health, Feb 2005, Vol.10, No.2, pp. 121-138.

62

Page 63: BIOANALYTICAL METHOD VALIDATION FOR QUANTITATIVE ... · emerge years later. Inside the hepatocyte the sporozoite is converted to trophozoite, which subsequently divides into several

ACKNOWLEDGEMENTS

First of all I would like to deeply thank my supervisors for all support during the work on this

thesis. From start, Prof. Dr. Francoise Bressolle accepted me to join the team in the Clinical

Pharmacokinetic Laboratory (Faculty of Pharmacy, University Montpellier 1, Montpellier, France),

showed never-ending enthusiasm in the performance of experimental projects, encouraged in

writing my thesis and helped to find new solutions to various problems. I am very grateful for your

professional guidance. Prof. Dr. Vitalis Briedis suggested and encouraged to carry out the

experiments of my thesis in foreign country, advised in the field of science and showed

unbelievable understanding. I am very glad I could realize some of your many ideas.

Secondly, I am very thankful to Nicolas Taudon for fruitful scientific collaborations, advises

and consultations. I have really appreciated all lovely and good-natured support from Delphine

Margout, her kindness and willingness to teach the subtleties of laboratory work and correct French

language. I am very happy that I could join your team!

I would like to thank my colleagues of university for your friendship and collaboration.

Finally, I want to thank my family: my parents and my sister for all enormous support. And

my beloved Mindaugas: for understanding, support, care and happiness you bring into my life.

63


Recommended