The Pennsylvania State University

The Graduate School

College of Agricultural Sciences

IN VITRO DRUG INTERACTIONS BETWEEN TAFENOQUINE AND

CURRENT ANTIMALARIALS IN PLASMODIUM FALCIPARUM PARASITES

A Thesis in

Entomology

by

Karen Kemirembe

 2015 Karen Kemirembe

Submitted in Partial Fulfillment of the Requirements for the Degree of

Master of Science

December 2015

The thesis of Karen Kemirembe was reviewed and approved* by the following:

Liwang Cui Professor of Entomology Thesis Adviser

Kelli Hoover Professor of Entomology

Jason L. Rasgon Associate Professor of Entomology and Disease Epidemiology

Cristina Rosa Associate Professor of Plant Virology

Gary W. Felton Professor of Entomology Head of the Department of Entomology

*Signatures are on file in the Graduate School

iii

ABSTRACT

Malaria, caused by Plasmodium spp. parasites, is one of the top ten causes of death in low-income tropical/sub-tropical countries. There are five species of human malaria but of these,

Plasmodium falciparum and Plasmodium vivax are the most prevalent. The 2014 World Malaria

Report estimates that about half of all countries with ongoing malaria transmission are co- endemic for P. vivax and P. falciparum malaria. Special features of P. vivax and P. falciparum respectively are that P. vivax has a latent liver stage that can be re-activated 6 months to several years after initial clearance of an infection with antimalarial drugs, and P. falciparum, although lacking in a dormant liver stage, has the slowest growing infectious sexual stage (gametocytes) of the human malarias. In addition, P. falciparum has prolonged gametocyte clearance within an infected patient due to the inefficacy of current antimalarials at this stage, posing an increased risk of transmission from infected humans to female mosquito vectors that carry malaria.

Currently, an 8-aminoquinoline class drug, primaquine (PMQ) is the only antimalarial licensed to target the liver and gametocyte stages of these two malaria species; the first- line treatments, termed combination therapy (ACT) preferentially target the asexual stages in a human host that cause the majority of clinical symptoms of the disease. To address this drug shortage, GlaxoSmithKline and the Walter Reed Army Institute of Research have developed an 8- aminoquinoline 5-phenoxyl PMQ derivative, WR 238605/SB-252263, herein referred to as tafenoquine (TFQ). TFQ is currently in late-stage clinical development for the radical cure of liver stage, asexual and sexual stage P. vivax, administered as a single 300 mg dose following a 3- day chloroquine (CQ) regimen to patients pre-screened for a >70% glucose 6-phosphate- dehydrogenase (G6PD) activity in order to minimize severe hemolytic anemia from oxidative stress, a side effect of the drug in deficient individuals.

iv

On account of 1) a high prevalence of mixed P. vivax and P. falciparum co-infections observed in field clinical trials, 2) possible misdiagnosis of P. falciparum as P. vivax and 3) widespread and emerging CQ resistance in P. falciparum and P. vivax respectively, leading to a shift in antimalarial use from CQ to ACT, studying the drug combination interactions of TFQ with ACT for both P. falciparum and P. vivax parasites is especially important in order to assess the best drug combinations of antimalarial regimens; 3 days of ACT for asexual stages, followed by a single dose of TFQ, that will achieve enhanced treatment efficacy, or synergy.

Since there is currently no continuous in vitro culture protocol for P. vivax, this thesis focusses on TFQ’s effect on P. falciparum when given post ACTs for a P. vivax infection.

Although TFQ is being developed primarily for P. vivax, it is important to note that studies in P. falciparum have shown causal prophylactic, schizonticidal and gametocytocidal activity of TFQ, suggesting possible off-target inhibition of P. falciparum, should the drug be deployed for P. vivax.

In order to assess how TFQ interacts with current ACT antimalarials in vitro, both asexual and sexual stage parasite replication was assessed in the presence of a single drug or combined ACT- partner drugs with TFQ. A SYBR Green I fluorescent dye was used to quantify asexual stage replication whereas a flow cytometry based method was used to quantify the drug inhibition of transgenic parasites expressing green fluorescent protein (GFP) in sexual stage parasites. Fractional inhibitory indices were calculated from growth inhibition curves of single or combined drugs at fixed ratios and used to determine synergistic, additive or antagonistic drug interactions of parasite strains with differing genetic backgrounds. In general, synergism, whereby a given drug in combination with TFQ is more potent than when used alone would be the desired result. Five sensitive or resistant parasite strains to either CQ or Artemisinin (ART) were tested against a panel of long-lasting six ACT component drugs namely amodiaquine

(AMQ), lumefantrine (LMF), mefloquine (MFQ), naphthoquine (NQ), piperaquine (PPQ) and

v pyronaridine (PND) using a fixed- ratio method based on published Cmax values to mimic in vivo human pharmacokinetics. The short-lived ART and its derivatives were excluded from the experiment on the basis that TFQ peak plasma levels are achieved after their elimination in vivo.

Results showed mostly synergistic relationships in all strains at the asexual stage, regardless of

CQ or ART sensitivity. Some gametocyte interactions were however found to be antagonistic.

Here for the first time, TFQ interactions with ACTs have been investigated. Taken together, TFQ appears to have a positive effect on P. falciparum parasite inhibition, at least in vitro, and patients with mixed malaria infection, will likely benefit from taking TFQ in addition to the standard ACTs. Each malaria-infected region will therefore have to select an ACT-TFQ pair that will likely give the most effective treatment in patients. In vivo drug interaction work in humanized mice with varying G6PD activity as well as clinical drug interaction trials in humans are a necessary follow-up to these claims since host factors such as hematocrit, gender, immunity, drug activation as well as diet might alter the results of drug-drug interaction studies.

This work portrays the urgent need for more in vitro studies to perform interaction analyses on both sexual and asexual parasites, not just the former, as the interactions appear to be stage-dependent; there is currently only one publication that includes the sexual stages, perhaps due to previous difficulty in obtaining these in sufficient amounts compared to their asexual stage counterparts. The flow cytometry based method used here, as has been previously shown, is a reproducible way to do this in vitro, although different methods may have to be applied to field parasite isolates. Additionally, the disagreement between results obtained here using ratios based on in vivo plasma concentrations and those reported previously for CQ-TFQ interactions supports the notion of a switch from the more common use of fixed ratios based on in vitro drug inhibitory concentrations to physiologically relevant fixed drug ratios.

vi

TABLE OF CONTENTS

List of Figures ...... vii

List of Tables ...... viii

List of Abbreviations ...... ix

Acknowledgements ...... x

Chapter 1 An introduction to human malaria, Plasmodium spp...... 1

1.1 Introduction ...... 1 1.2 P. falciparum and P. vivax life cycle and implications for chemotherapy ...... 4 1.3 Thesis aim, objectives and rationale ...... 9 1.4 References ...... 12

Chapter 2 Tafenoquine Drug Combinations in Asexual and Sexual parasites ...... 21

2.1 Introduction ...... 21 2.2 Materials and methods ...... 27 2.3 Results ...... 33 2.4 Discussion ...... 38 2.5 Conclusions ...... 43

Chapter 3 Conclusions and future directions ...... 50

3.1 Summary of findings and relevance ...... 50 3.2 Perspectives on malaria control ...... 52 3.3 References ...... 55

Appendix A ...... 60

A1: Gametocyte induction ...... 60

A1.1 Equipment and Materials ...... 60 A1.2 Reagents ...... 60 A1.3 Procedure (Modified from Fivelman et al., 2007; Lucantoni et al., 2013) ..... 62

A.2 Flow cytometry method to determine gametocyte drug inhibition ...... 64

A2.1 Materials and Equipment ...... 64 A2.2 Reagents ...... 65 A2.3 Procedure (Modified from Wang et al., 2014) ...... 66 A2.4 References ...... 71

Appendix B: Drug-Drug interaction isobolograms for each lab strain ...... 72

vii

LIST OF FIGURES

Figure 1-1. Life-cycle of Plasmodium falciparum...... 6

Figure 1-2. Methods flow-chart for asexual and sexual drug interactions ...... 11

Figure 4-1. Dot plots of control and fluorescent parasites...... 67

Figure 5-1. Isobolograms of asexual and sexual parasite strains...... 72

viii

LIST OF TABLES

Table 1-1. Comparison between P. falciparum and P. vivax malaria parasites ...... 7

Table 1-2. Resistance markers of drugs used in this study...... 10

Table 2-1. Structural classification of antimalarials used in this study...... 25

Table 2-2. Summary of available artemisinin combination therapies ...... 26

Table 2-3. Pharmacokinetics of drugs used in this study...... 32

Table 2-4. Ratios used for asexual TFQ- ACT-partner drug interactions ...... 33

Table 2-5. Drug susceptibilities of asexual parasites ...... 35

Table 2-6. Median TFQ inhibitory concentrations by gametocyte stage ...... 35

Table 2-7. Drug susceptibilites of 3D7αtubIIGFP gametocytes ...... 36

Table 2-8. Summary of TFQ-ACT- partner drug interactions...... 37

ix

LIST OF ABBREVIATIONS

Acronyms of Antimalarial drugs

ACT Artemisinin Combination Therapy

AMQ Amodiaquine

ART Artemisinin

CQ Chloroquine

DHA Dihydroartemisinin

LMF Lumefantrine

MFQ Mefloquine

NQ Naphthoquine

PMQ Primaquine

PND Pyronaridine

PPQ Piperaquine

TFQ Tafenoquine

Acronyms of Materials and Methods

MCM Malaria Complete Medium

G6PD Glucose-6-Phosphate Dehydrogenase

GFP Green Fluorescent Protein

CM Conditioned medium

IC50 Median inhibitory drug concentration

FIC Fractional Inhibitory Concentration

x

ACKNOWLEDGEMENTS

Many thanks to my advisor, Dr. Liwang Cui for his guidance and invested resources in me whilst in his lab. I am grateful to my committee members Dr. Cristina Rosa, Dr. Jason L.

Rasgon and Dr. Kelli Hoover for their generous time and advice. I am grateful to all Cui lab members for helpful discussions and to Dr. Mynthia Cabrera Goss for her mentorship. To Dr.

Gary W. Felton without whose leadership the Dept. of Entomology would not be the same, I am gratified. I am indebted to my parents for their continued support and patience with me being thousands of miles away from home to obtain a good education. I also wish to thank my house- mates, Jennifer T. Yang and Meredith T. Hanlon for always making our grad student home a fun and comfortable environment to live. I acknowledge my boyfriend, Troy F. Carl, and all my friends for their continued love and support. Lastly, this work would not be possible without the

National Institutes of Health’s (fund U19AIO89672) financial support.

1

Chapter 1

An introduction to human malaria, Plasmodium spp.

1.1 Introduction

Malaria, in addition to other infectious diseases like tuberculosis and HIV, is one of the top ten causes of death in low-income countries (WHO, 2014). The name, derived from Italian mal aria or ‘bad air’ came about due to previous beliefs that its cause was air-borne, rather than vector-borne (Wahlgren and Bejarano, 1999). Malaria parasites are single cell eukaryotes or protozoans (not to be confused by popular media as viruses or bacteria) and are spread by female anopheles mosquitos as a by-product of blood feeding behavior (Nilsson et al., 2015). Mosquitoes find their hosts using odorant receptors sensitive to carbon dioxide in exhaled air (Turner et al.,

2011; Pellegrino et al., 2011).

The disease is reported to have killed approximately 584,000 in the year 2013 according to the most recent World Malaria report (WHO, 2014). Anopheles mosquitos (Diptera: Culicidae) are widespread worldwide with Anopheles gambiae, An. funestus and An. arabiensis being co- dominant in Africa, where most of the malaria fatalities occur. In S. E. Asia, An. dirus is the most prevalent amidst a larger selection of vectors compared to the few in Africa. An. darlingi is prevalent in S. America, An. Albimanus in Central America, and An. fluviatilis and An. stephensi in India/Western Asia (Sinka et al., 2012). Further research is necessary to determine whether these and all other species identified in the field are competent malaria vectors.

There are five known Plasmodium (Haemosporida: Plasmodiidae) species of malaria that infect humans, namely Plasmodium ovale, Plasmodium malariae, Plasmodium knowlesi,

2 Plasmodium vivax and Plasmodium falciparum, with the latter two being the most prevalent (Li et al., 2004; Moreno and Joyner, 2015; Biamonte et al., 2013 WHO, 2014). P. vivax is the most wide-spread malaria owing to its lower temperature threshold for sporogony and subsequent parasite development within a mosquito of 16°C, compared to 20°C for P. falciparum, making it withstand temperate, in additional to tropical climate provided that by chance it comes into contact with a competent vector (Weihe et al., 1991; WHO, 2015). All human malarias with the exception of P. malariae which has a 72 hr life- cycle have a 48 hr life-cycle (Moreno and

Joyner, 2015). P. knowlesi is predominantly found in macaque monkeys with a few human infections a year (Lee et al., 2001). P. ovale is mostly found in West Africa and is often misdiagnosed as P. vivax (Roucher et al., 2014; Mueller et al., 2007). P. vivax and P. ovale, unlike P. falciparum have an advantage of having a dormant liver stage that can cause recrudescence or relapses after a few months or years after initial treatment of a malaria infection

(Li et al., 2014; Leroy et al., 2014). This is not only detrimental to people in P. vivax endemic areas, but also for tourists, expats and military personnel who may not know that they harbor dormant malaria until a relapse in their home countries that may be difficult to diagnose.

When infected with P. falciparum malaria, fevers and joint pain are common symptoms, and if left untreated, severe cerebral malaria may develop as well as multi-organ failure

(Biamonte et al., 2013). Due to their underdeveloped immune system, children under 5 years of age account for >75% of malaria deaths and are prone to malaria-related complications like hypoglycemia and anemia, and these can often be fatal if left untreated (WHO, 2014). Rapid diagnostic tests of malaria include a finger prick followed by a thick blood smear that is stained and viewed by microscopy, as well as the more sensitive Real-Time Nucleic Acid Sequence-

Based Amplification to detect sub-microscopic levels of malaria (Schneider et al., 2004).

Wide-spread control of malaria has included the use of pyrethroid- based insecticide treated mosquito nets, indoor residual spraying, draining of all possible larval sites to target the

3 female Anopheles vector, although this has been hindered by the presence of vectors that bite earlier and outdoors (Russell et al., 2011; Mwangangi et al., 2013; Strode et al., 2014; Yohannes and Boelee, 2012), thereby evading any contact with any indoor vector-control methods. To manage the malaria parasite, mass drug administration and chemoprophylaxis for vulnerable populations, such as pregnant women have been successfully carried out (Seidlein and

Greenwood, 2003; Tesfazghi et al., 2015; Steketee, 2014). Other control strategies in development are whole organism and partial protein-in-adjuvant vaccines, use of transgenic mosquitoes, photonic fences to target females using their wing beat frequencies and fungal treatment, among others (Moreno and Joyner 2015; Wells et al; 2015; Heinig and Thomas, 2015;

Marsden et al., 2013; Hughes et al., 2014; Hyde et al., 2014). A multi-faceted target of both vector and parasite is essential if we are to further reduce malaria prevalence.

Unfortunately, there have been issues with the control of malaria in the last 50 years due to the evolution of resistance to both insecticides used against the vector, and to antimalarial drugs used against the blood stage parasite in infected humans. There is wide-spread resistance to chloroquine, as well as to sulfadoxine- pyrimethamine, partial mefloquine (MFQ) resistance along the Thai-Cambodia border and emerging resistance to the first- line antimalarial treatment artemisinin (ART) in 6 countries in S. E Asia due to either non-synonymous point mutations of particular genes like Pfcrt in chloroquine and Pfkelch13 mutations in ART resistance or increased copy numbers like in the Pfmdr1gene in the case of MFQ (Ariey et al., 2014; Ranson et al., 2011;

White, 2004; Phyo et al., 2012; Price et al., 2004; Sidhu et al., 2002). Currently, chemotherapy of antimalarial drugs is employed in a co-formulated combination to reduce the evolution of resistance to one drug, with artemisinin combination therapy (ACT) being the first line treatment for malaria. ACTs consist of a fast-acting, short half-life and artemisinin-based component and a longer lasting partner drug to eliminate those parasites that are left-over from the initial killing

(Eastman and Fidock, 2009). In addition to ACT, the WHO recommends prescribing a single

4 0.25mg base/kg dose of the 8-aminoquinoline, primaquine (PMQ), the only licensed gametocytocide, to malaria-infected individuals in areas with emerging artemisinin resistance to curb transmission of gametocytes from infected humans to mosquitoes (WHO, 2014). There is widespread CQ resistance in P. falciparum and ACT is the most used treatment whereas CQ is still efficacious and used commonly against P. vivax in all but 9 countries due to its cheaper price compared to ACT (White, 2013; Price et al., 2014; WHO, 2014). This slow emergence of CQ resistance in P. vivax has been attributed to a shorter duration between peak asexual and gametocyte densities, thus exposing both asexual and sexual parasites to antimalarial drugs when taken doing the symptomatic asexual phase, whereas in P. falciparum., the extended difference in asexual and sexual parasite peak times leads to evasion of an acute febrile response and the release of tumor necrosis factor 2, as well as preferential exposure of only the symptom-causing asexual parasites to antimalarials allowing those asexual parasites that survive drug treatment to differentiate into infectious gametocytes. Furthermore, the reduced parasite numbers in P. vivax versus P. falciparum infections likely plays a role in how many parasites can survive drug treatment, since the evolution of resistance is directly proportional to initial parasite biomass.

Another hypothesis is that since P. vivax gametocytes appear early during infection, transmission may occur successfully before exposure to antimalarial drugs, lessening the likelihood of resistance developing (White, 2004; Mueller et al., 2009; Drakely et al., 2006, Douglas et al.,

2013).

1.2 P. falciparum and P. vivax life cycle and implications for chemotherapy

The malaria life cycle is very complex, with the parasite spending some time both in a human host and in a mosquito. A female Anopheles spp. mosquito, in search of nutrient supplements for egg-laying, bites a person infected with malaria and draws blood containing male

5 and female sexual-stage parasites, named gametocytes. Gametocytes on encountering a change in pH and xanthurenic acid in the mosquito gut are activated in a preparation for gametogenesis where the male and female gametocytes are transformed to gametes by mitosis and meiosis respectively (Delves et al., 2013; Khan et al., 2005). The male gametocyte undergoes three rounds of DNA replication in preparation for the formation of eight motile gametes that each go on to fertilize female gametes, which they encounter through a cell surface adhesion process mediated by Pfs48/45 in a process called exflagellation. Female gametocytes once activated in the mosquito, egress from the red blood cell (RBC), de-repress a lot of silenced transcripts that were under the control of a DOZI gene (Mair et al., 2006). A HAP-2 Arabidopsis plant sterility gene homolog dependent formation of a diploid zygote occurs. The zygote matures into a tetraploid ookinete, meiosis occurs to form to form multi-cellular oocysts that burst to release haploid sporozoites that then travel to the mosquito salivary glands within mosquito hamocoel

(Josling and Llinas, 2015). Mature sporozoites are then passed on from mosquitoes to humans when a female mosquito takes her next blood meal.

Whilst in the human body, sporozoites travel to the liver where they infect hepatocytes and mature into liver schizonts. These schizonts mature in the liver within 7 days and eventually release up to 40,000 merozoites that then initiate the asexual intra-erythrocytic blood stage when they invade red blood cells. In the case of P. vivax, a small proportion of liver parasites become dormant in the liver awaiting activation at a later time point ranging from months to years; these are termed hypnozoites (Mikolaiczak et al., 2015). Within the red blood cells, the malaria parasites replicate asexually from ring stage, progressing through the highly metabolic trophozoite stage, gradually digesting the host red blood cell hemoglobin as they develop over the course of a 48 hour life-cycle with the cycle repeating after a blood-stage schizont bursts to release 8-32 merozoites, and proceeds to invade new red blood cells (Flannery et al., 2013). Due to the rapid replication and exponential growth of these asexual parasites, an immune response is

6 triggered that causes fever and other malaria-induced clinical symptoms like joint pain. Anemia occurs due to the increased rupture of host red blood cells. A small proportion of <10% these haploid asexual blood parasites undergo gametocytogenesis, mediated by the transcription factor,

PfAP2-G (Sinha et al., 2014). P. falciparum gametocytes take 10-12 days to mature within a human host in five distinct stages I-V, and peak in number 7-10 days after the initial asexual stage peak (McKenzie et al., 2006; Talman et al., 2004). Stages II-IV are sequestered in the bone marrow, presumably to avoid clearance by the spleen, and only return to peripheral circulation as mature stage V gametocytes that are then infectious to a mosquito within 2 days (Nilsson et al.,

2015). P. vivax gametocytes on the other hand mature within 3-4 days and reach peak parasitemia simultaneously with asexual parasites and there is no evidence of sequestration

(Mueller et al., 2009; Bousema and Drakely, 2011; White et al., 2008). They are therefore likely to come into contact with antimalarial drugs taken during the symptomatic asexual stage.

Fig 1-1 Life cycle of Plasmodium falciparum. Both mosquito and human stages are shown. P. vivax liver hypnozoites are indicated to differentiate the two species. Adapted by permission from

7 Macmillan Publishers Ltd: [Nature Publishing Group] (Michalakis and Renaud, 2009), copyright

(2009). RightsLink license number: 3741480055847. License date: Nov 3rd, 2015.

Table 1-1 Comparison between P. falciparum and P. vivax malaria parasites. Data is from WHO,

2015 and Mueller et al., 2009.

Plasmodium falciparum P. vivax

18ºC sporogony 14ºC sporogony

12 d gametocyte maturation 3-4d gametocyte maturation

Crescent-shaped gametocytes Round gametocytes

Peak gametocyte day 7-11 Peak gametocyte day 1-2

No dormant liver stage parasites Dormant liver stage parasites

Gametocytes more tolerant to ACT Gametocytes killed by ACT

Infects all RBCs Infects reticulocytes

Schüfnner’s dots absent on RBC Schüfnner’s dots present

BOTH REQUIRE HEMOGLOBIN DIGESTION SO SIMILAR HEME-MEDIATED

DRUG TARGETS

Owing to the fact that malaria symptoms are initiated at the asexual intra-erythrocytic blood stage, most antimalarial drugs on the market target this stage, with most drugs depending on the hemoglobin degradation pathway for their action. Gametocytes minimize their hemoglobin digestion machinery past stage III so only drugs affecting other organelles beside the digestive vacuole, such as the mitochondria are effective against them (Butcher, 1997). Only one drug, namely primaquine (PMQ) is licensed to kill gametocyte sexual stages. A derivative of PMQ,

8 tafenoquine (TFQ), is active against the afore-mentioned liver stage that occurs in some P. vivax infections (Li et al., 2014). TFQ is the subject of study in this thesis, because it is currently in late stage clinical trials for the radical treatment of P. vivax, and encounters P. falciparum in areas co- endemic for both malaria species. The problem with both PMQ and TFQ is that they cause hemolysis in individuals with a Glucose-6-Phosphate Dehydrogenase (G6PD) deficiency.

Many studies are underway to identify other drug compounds that are safe for humans and possess the transmission blocking benefit of killing sexual stage gametocytes, but work in this area is relatively in its infancy because it wasn’t until recently that methods were developed to readily obtain large numbers of gametocytes in vitro (Fivelman et al., 2007). Numerous studies have since investigated the effect of potential antimalarials that are gametocytocidal

(Delves et al., 2013; Reader et al., 2015; Peatey et al., 2012). The problem currently lies in the fact that due to differences in methodology, very few of these studies have data in agreement with one another (Reader et al., 2015). One thing that is for certain is that the oldest antimalarial, methylene blue, has received renewed interest because of its inhibitory effects at both the asexual and sexual blood stages of the malaria parasite and is currently in clinical trials (Coulibaly et al.,

2015).

As mentioned earlier, the only drug licensed for gametocytocidal clearance is PMQ, an 8- aminoquinoline. It’s derivative, TFQ, another 8-aminoquinoline is in late stage clinical trials for the treatment of P. vivax relapse, but in vitro studies have shown its efficacy in all stages of P. falciparum as well (Shanks et al., 2001; Crockett and Cain, 2007; Brueckner et al., 1998). Due to a high prevalence of mixed infections of P. vivax and P. falciparum, a person taking a TFQ prescription for P. vivax is likely to be co-infected with both parasites (Snounou and White.,

2004; Douglas et al., 2013). My thesis therefore aims to investigate the off-target effects of TFQ on P. falciparum, when taken with ACT or chloroquine treatment. The recommendation is that a person will be given 3 days of CQ or ACT followed by a single dose of 300mg TFQ (Llanos-

9 Cuentas et al., 2013). The hypothesis is that the TFQ will come into contact with the longest lasting ACT partner drug in vivo and will either enhance, maintain or diminish the efficacy of these drugs on P. falciparum parasites. Although TFQ is being developed primarily for P. vivax, this study will focus only on P. falciparum due to the lack of a continuous in vitro culture method for P. vivax parasite proliferation due to the requirement of fresh reticulocytes (Roobsong et al.,

2015). Follow up studies ex-vivo or in vivo will have to be carried out to further verify the results of this study.

1.3 Thesis aim, objectives and rationale

Aim: To determine if there is an off-target treatment benefit of TFQ against P. falciparum parasites when administered with ACT for P.vivax in areas where mixed infections occur.

Objective 1

To determine asexual stage TFQ-ACT-partner drug combination interactions

Rationale: First line ACT targets asexual stage parasites. ACT consists of a fast-acting and rapidly eliminated artemisinin derivative such as dihydroartemisinin or and a long lasting partner drug, such as the bisquinoline, piperaquine. TFQ, if administered after 3 days of

ACT will encounter the long lasting drug compound in peripheral blood. The resulting TFQ-drug combinations’ ability to inhibit growth and proliferation of asexual parasites will either be enhanced (synergy), maintained (additivity) or diminished (antagonism).

Method: SYBR Green I fluorescent dye is used to stain DNA of the parasite with higher fluorescence corresponding to more parasite survival post-drug treatment (Smilkstein et al.,

2004).

10 Asexual strains used:

- IPC5202 (Cambodia: DHA resistant [Pfkelch13 R539T mutation])

- 7G8 (Brazil: CQ resistant [Pfcrt K76T SVMNT haplotype])

- DD2 (Indo-China: CQ resistant [Pfcrt K76T CVIET haplotype, Pfmdr1 copy number: 4])

- 3D7 (Africa: CQ sensitive)

- HB3 (Honduras: CQ sensitive)

Table 1-2 Resistance markers of drugs used in this study.

DRUG RESISTANCE NOTES REFERENCE

MARKERS

LMF, MFQ Pfmdr1 Multi-drug resistance Sidhu et al., 2006;

amplification, N86Y transporter, influx pump Folarin et al., 2011

CQ Pfcrt K76T Chloroquine resistance Valderramos et al.,

transporter, efflux pump 2010

ART R539T (Pfkelch13) KLHL12 homolog likely Ariey et al., 2014

involved in ubiquitination

Objective 2

To determine sexual stage TFQ-schizonticide combination interactions

Rationale: Most antimalarial drug treatment (ACT) targets the asexual stage of parasites. A single drug, an 8-aminquinoline, PMQ additionally targets both sexual stage gametocyte parasites of P. falciparum, P. vivax and liver stage dormant parasites in P. vivax. TFQ, a PMQ derivative is in late stage clinical trials to carry out PMQ’s role in P. vivax, taken on the third day of antimalarial treatment. Since mixed infections are common in the field, TFQ will encounter P.

11 falciparum gametocytes. Although TFQ is being developed for P. vivax, a desired off-target effect on P. falciparum, either additivity or synergy is desired. In addition, past drug-drug interaction studies have only been conducted in asexual parasites.

Method: A transgenic line of parasite expressing GFP under the α-tubulin II promoter is used to determine cell viability after drug treatment by flow cytometry (Wang et al., 2014).

Gametocyte strain used: 3D7αtubIIGFP [PlasmoDB: PF3D7_042230]

Drugs used for both asexual and sexual drug interactions include the 4-aminoquinolines chloroquine (CQ), amodiaquine (AMQ) and naphthoquine (NQ), the bisquinoline piperaquine

(PPQ), aryl amino-alcohols lumefantrine (LMF) and mefloquine (MFQ) and the mannich base, pyronaridine (PND).

Figure 1-2 Methods flow-chart for asexual and sexual drug interactions.

12 1.4 References

Ariey, Frédéric, Benoit Witkowski, Chanaki Amaratunga, Johann Beghain, Anne-Claire

Langlois, Nimol Khim, Saorin Kim et al. "A molecular marker of artemisinin-resistant

Plasmodium falciparum malaria." Nature 505, no. 7481 (2014): 50-55.

Biamonte, Marco A., Jutta Wanner, and Karine G. Le Roch. "Recent advances in malaria drug discovery." Bioorganic & medicinal chemistry letters 23, no. 10 (2013): 2829-2843.

Bousema, Teun, and Chris Drakeley. "Epidemiology and infectivity of Plasmodium falciparum and Plasmodium vivax gametocytes in relation to malaria control and elimination." Clinical microbiology reviews 24, no. 2 (2011): 377-410.

Brueckner, Ralf P., Kenneth C. Lasseter, Emil T. Lin, and Brian G. Schuster. "First-time- in-humans safety and pharmacokinetics of WR 238605, a new antimalarial." The American journal of tropical medicine and hygiene 58, no. 5 (1998): 645-649.

Butcher, G. A. "Antimalarial drugs and the mosquito transmission of

Plasmodium." International journal for parasitology 27, no. 9 (1997): 975-987.

Coulibaly, Boubacar, Michael Pritsch, Mamadou Bountogo, Peter E. Meissner, Eric

Nebié, Christina Klose, Meinhard Kieser et al. "Efficacy and safety of triple combination therapy with -amodiaquine-methylene blue for falciparum malaria in children: a randomised controlled trial in Burkina Faso." Journal of Infectious Diseases (2014): jiu540.

Crockett, Maryanne, and Kevin C. Kain. "Tafenoquine: a promising new antimalarial agent." (2007): 705-715.

Delves, Michael J., Andrea Ruecker, Ursula Straschil, Jöel Lelièvre, Sara Marques, María

José López-Barragán, Esperanza Herreros, and Robert E. Sinden. "Male and female Plasmodium falciparum mature gametocytes show different responses to antimalarial drugs." Antimicrobial agents and chemotherapy 57, no. 7 (2013): 3268-3274.

13 Douglas, Nicholas M., Julie A. Simpson, Aung Pyae Phyo, Hadjar Siswantoro, Armedy

R. Hasugian, Enny Kenangalem, Jeanne Rini Poespoprodjo et al. "Gametocyte dynamics and the role of drugs in reducing the transmission potential of Plasmodium vivax." Journal of Infectious

Diseases 208, no. 5 (2013): 801-812.

Drakeley, Chris, Colin Sutherland, J. Teun Bousema, Robert W. Sauerwein, and Geoffrey

AT Targett. "The epidemiology of Plasmodium falciparum gametocytes: weapons of mass dispersion." Trends in parasitology 22, no. 9 (2006): 424-430.

Eastman, Richard T., and David A. Fidock. "Artemisinin-based combination therapies: a vital tool in efforts to eliminate malaria." Nature Reviews Microbiology 7, no. 12 (2009): 864-

874.

Fivelman, Quinton L., Louisa McRobert, Sarah Sharp, Cathy J. Taylor, Maha Saeed,

Claire A. Swales, Colin J. Sutherland, and David A. Baker. "Improved synchronous production of

Plasmodium falciparum gametocytes in vitro." Molecular and biochemical parasitology 154, no.

1 (2007): 119-123.

Flannery, Erika L., Arnab K. Chatterjee, and Elizabeth A. Winzeler. "Antimalarial drug discovery approaches and progress towards new medicines." Nature Reviews Microbiology 11, no. 12 (2013): 849-862.

Folarin, O. A., C. Bustamante, G. O. Gbotosho, A. Sowunmi, M. G. Zalis, A. M. J.

Oduola, and C. T. Happi. "In vitro amodiaquine resistance and its association with mutations in pfcrt and pfmdr1 genes of Plasmodium falciparum isolates from Nigeria." Acta tropica 120, no. 3

(2011): 224-230.

Hughes, Grant L., Brittany L. Dodson, Rebecca M. Johnson, Courtney C. Murdock,

Hitoshi Tsujimoto, Yasutsugu Suzuki, Alyssa A. Patt et al. "Native microbiome impedes vertical transmission of Wolbachia in Anopheles mosquitoes." Proceedings of the National Academy of

Sciences 111, no. 34 (2014): 12498-12503.

14 Hyde, Roderick A., Jordin T. Kare, Artyom Makagon, Emma Rae Mullen, Nathan P.

Myhrvold, Thomas J. Nugent, Nathan John Pegram, Nels R. Peterson, Phillip Rutschman, and

Lowell L. Wood. "Photonic fence." U.S. Patent Application 14/255,119, filed April 17, 2014.

Josling, Gabrielle A., and Manuel Llinás. "Sexual development in Plasmodium parasites: knowing when it's time to commit." Nature Reviews Microbiology 13, no. 9 (2015): 573-587.

Khan, Shahid M., Blandine Franke-Fayard, Gunnar R. Mair, Edwin Lasonder, Chris J.

Janse, Matthias Mann, and Andrew P. Waters. "Proteome analysis of separated male and female gametocytes reveals novel sex-specific Plasmodium biology." Cell 121, no. 5 (2005): 675-687.

Lee, Kim-Sung, P. C. Divis, Siti Khatijah Zakaria, Asmad Matusop, Roynston A. Julin,

David J. Conway, Janet Cox-Singh, and Balbir Singh. "Plasmodium knowlesi: reservoir hosts and tracking the emergence in humans and macaques." PLoS Pathog 7, no. 4 (2011): e1002015.

Li, Qigui, Michael O'Neil, Lisa Xie, Diana Caridha, Qiang Zeng, Jing Zhang, Brandon

Pybus, Mark Hickman, and Victor Melendez. "Assessment of the prophylactic activity and pharmacokinetic profile of oral tafenoquine compared to primaquine for inhibition of liver stage malaria infections." Malaria Journal 13, no. 141 (2014): 10-1186.

Llanos-Cuentas, Alejandro, Marcus V. Lacerda, Ronnatrai Rueangweerayut, Srivicha

Krudsood, Sandeep K. Gupta, Sanjay K. Kochar, Preetam Arthur et al. "Tafenoquine plus chloroquine for the treatment and relapse prevention of Plasmodium vivax malaria

(DETECTIVE): a multicentre, double-blind, randomised, phase 2b dose-selection study." The

Lancet 383, no. 9922 (2014): 1049-1058.

McKenzie, F. Ellis, Chansuda Wongsrichanalai, Alan J. Magill, J. Russ Forney, Barnyen

Permpanich, Carmen Lucas, Laura M. Erhart et al. "Gametocytemia in Plasmodium vivax and

Plasmodium falciparum infections." Journal of Parasitology 92, no. 6 (2006): 1281-1285.

Mair, Gunnar R., Joanna AM Braks, Lindsey S. Garver, Joop CAG Wiegant, Neil Hall,

Roeland W. Dirks, Shahid M. Khan, George Dimopoulos, Chris J. Janse, and Andrew P. Waters.

15 "Regulation of sexual development of Plasmodium by translational repression." Science 313, no.

5787 (2006): 667-669.

Michalakis, Yannis, and François Renaud. "Malaria: Evolution in vector control." Nature 462, no. 7271 (2009): 298-300.

Mikolajczak, Sebastian A., Ashley M. Vaughan, Niwat Kangwanrangsan, Wanlapa

Roobsoong, Matthew Fishbaugher, Narathatai Yimamnuaychok, Nastaran Rezakhani et al.

"Plasmodium vivax liver stage development and hypnozoite persistence in human liver-chimeric mice." Cell host & microbe 17, no. 4 (2015): 526-535.

Moreno, Alberto, and Chester Joyner. "Malaria vaccine clinical trials: what's on the horizon." Current opinion in immunology 35 (2015): 98-106.

Mueller, Ivo, Mary R. Galinski, J. Kevin Baird, Jane M. Carlton, Dhanpat K. Kochar,

Pedro L. Alonso, and Hernando A. del Portillo. "Key gaps in the knowledge of Plasmodium vivax, a neglected human malaria parasite." The Lancet infectious diseases 9, no. 9 (2009): 555-

566.

Mueller, Ivo, Peter A. Zimmerman, and John C. Reeder. "Plasmodium malariae and

Plasmodium ovale–the ‘bashful’malaria parasites." Trends in parasitology23, no. 6 (2007): 278-

283.

Marsden, Clare D., Anthony Cornel, Yoosook Lee, Michelle R. Sanford, Laura C. Norris,

Parker B. Goodell, Catelyn C. Nieman et al. "An analysis of two island groups as potential sites for trials of transgenic mosquitoes for malaria control." Evolutionary applications 6, no. 4 (2013):

706-720.

Mwangangi, Joseph M., Ephantus J. Muturi, Simon M. Muriu, Joseph Nzovu, Janet T.

Midega, and Charles Mbogo. "The role of Anopheles arabiensis and Anopheles coustani in indoor and outdoor malaria transmission in Taveta District, Kenya." Parasit Vectors 6 (2013): 114.

16 Nilsson, Sandra K., et al. "Targeting human transmission biology for malaria elimination." PLoS Pathog 11.6 (2015): e1004871.

Peatey, Christopher L., Didier Leroy, Donald L. Gardiner, and Katharine R. Trenholme.

"Anti-malarial drugs: how effective are they against Plasmodium falciparum gametocytes." Malaria Journal 11 (2012): 34.

Pellegrino, Maurizio, Nicole Steinbach, Marcus C. Stensmyr, Bill S. Hansson, and Leslie

B. Vosshall. "A natural polymorphism alters odour and DEET sensitivity in an insect odorant receptor." Nature 478, no. 7370 (2011): 511-514.

Phyo, Aung Pyae, Standwell Nkhoma, Kasia Stepniewska, Elizabeth A. Ashley, Shalini

Nair, Rose McGready, Carit ler Moo et al. "Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study." The Lancet 379, no. 9830 (2012): 1960-1966.

Price, Ric N., Anne-Catrin Uhlemann, Alan Brockman, Rose McGready, Elizabeth

Ashley, Lucy Phaipun, Rina Patel et al. "Mefloquine resistance in Plasmodium falciparum and increased pfmdr1 gene copy number." The Lancet 364, no. 9432 (2004): 438-447.

Price, Ric N., Lorenz von Seidlein, Neena Valecha, Francois Nosten, J. Kevin Baird, and

Nicholas J. White. "Global extent of chloroquine-resistant Plasmodium vivax: a systematic review and meta-analysis." The Lancet Infectious Diseases 14, no. 10 (2014): 982-991.

Ranson, Hilary, Raphael N’Guessan, Jonathan Lines, Nicolas Moiroux, Zinga Nkuni, and

Vincent Corbel. "Pyrethroid resistance in African anopheline mosquitoes: what are the implications for malaria control?." Trends in parasitology 27, no. 2 (2011): 91-98.

Reader, Janette, Mariëtte Botha, Anjo Theron, Sonja B. Lauterbach, Claire Rossouw,

Dewaldt Engelbrecht, Melanie Wepener et al. "Nowhere to hide: interrogating different metabolic parameters of Plasmodium falciparum gametocytes in a transmission blocking drug discovery pipeline towards malaria elimination." Malaria Journal 14, no. 1 (2015): 213.

17 Roobsoong, Wanlapa, Chayada S. Tharinjaroen, Nattawan Rachaphaew, Porpimon

Chobson, Louis Schofield, Liwang Cui, John H. Adams, and Jetsumon Sattabongkot.

"Improvement of culture conditions for long-term in vitro culture of Plasmodium vivax." Malaria

Journal 14, no. 1 (2015): 297.

Russell, Tanya L., Nicodem J. Govella, Salum Azizi, Christopher J. Drakeley, S. Patrick

Kachur, and Gerry F. Killeen. "Increased proportions of outdoor feeding among residual malaria vector populations following increased use of insecticide-treated nets in rural Tanzania." Malaria

Journal 10, no. 80 (2011): 80.

Schneider, Petra, Gerard Schoone, Henk Schallig, Danielle Verhage, Denise Telgt,

Wijnand Eling, and Robert Sauerwein. "Quantification of Plasmodium falciparum gametocytes in differential stages of development by quantitative nucleic acid sequence-based amplification." Molecular and biochemical parasitology 137, no. 1 (2004): 35-41.

Sidhu, Amar Bir Singh, Dominik Verdier-Pinard, and David A. Fidock. "Chloroquine resistance in Plasmodium falciparum malaria parasites conferred by pfcrt mutations." Science 298, no. 5591 (2002): 210-213.

Smilkstein, Martin, Nongluk Sriwilaijaroen, Jane Xu Kelly, Prapon Wilairat, and Michael

Riscoe. "Simple and inexpensive fluorescence-based technique for high-throughput antimalarial drug screening." Antimicrobial agents and chemotherapy 48, no. 5 (2004): 1803-1806.

Snounou, Georges, and Nicolas J. White. "The co-existence of Plasmodium: sidelights from falciparum and vivax malaria in Thailand." Trends in parasitology 20, no. 7 (2004): 333-

339.

Staines, Henry M., and Sanjeev Krishna. Treatment and prevention of malaria: antimalarial drug chemistry, action and use. Springer Science & Business Media, 2012.

Steketee, Richard W. "Malaria Prevention during Pregnancy—Is There a Next Step

Forward?." (2014): e1001734.

18 Roucher, Clémentine, Christophe Rogier, Cheikh Sokhna, Adama Tall, and Jean-François

Trape. "A 20-year longitudinal study of Plasmodium ovale and Plasmodium malariae prevalence and morbidity in a West African population."PloS one 9, no. 2 (2014): e87169.

Shanks, G. Dennis, Aggrey J. Oloo, Gladys M. Aleman, Colin Ohrt, Francis W. Klotz,

David Braitman, John Horton, and Ralf Brueckner. "A new primaquine analogue, tafenoquine

(WR 238605), for prophylaxis against Plasmodium falciparum malaria." Clinical infectious diseases 33, no. 12 (2001): 1968-1974.

Sidhu, Amar Bir Singh, Anne-Catrin Uhlemann, Stephanie G. Valderramos, Juan-Carlos

Valderramos, Sanjeev Krishna, and David A. Fidock. "Decreasing pfmdr1 copy number in

Plasmodium falciparum malaria heightens susceptibility to mefloquine, lumefantrine, halofantrine, quinine, and artemisinin." Journal of Infectious Diseases 194, no. 4 (2006): 528-

535.

Sinha, Abhinav, Katie R. Hughes, Katarzyna K. Modrzynska, Thomas D. Otto, Claudia

Pfander, Nicholas J. Dickens, Agnieszka A. Religa et al. "A cascade of DNA-binding proteins for sexual commitment and development in Plasmodium."Nature 507, no. 7491 (2014): 253-257.

Sinka, Marianne E., Michael J. Bangs, Sylvie Manguin, Yasmin Rubio-Palis, Theeraphap

Chareonviriyaphap, Maureen Coetzee, Charles M. Mbogo et al. "A global map of dominant malaria vectors." Parasit Vectors 5, no. 1 (2012): 69.

Strode, Clare, Sarah Donegan, Paul Garner, Ahmad Ali Enayati, and Janet Hemingway.

"The impact of pyrethroid resistance on the efficacy of insecticide-treated bed nets against

African anopheline mosquitoes: systematic review and meta-analysis." PLoS Med 11, no. 3

(2014): e1001619.

Talman, Arthur M., Olivier Domarle, F. Ellis McKenzie, Frédéric Ariey, and Vincent

Robert. "Gametocytogenesis: the puberty of Plasmodium falciparum."Malaria Journal 3, no. 1

(2004): 24.

19 Tesfazghi, Kemi, Jenny Hill, Caroline Jones, Hilary Ranson, and Eve Worrall. "National malaria vector control policy: an analysis of the decision to scale-up larviciding in

Nigeria." Health policy and planning (2015): czv055.

Turner, Stephanie Lynn, Nan Li, Tom Guda, John Githure, Ring T. Cardé, and

Anandasankar Ray. "Ultra-prolonged activation of CO2-sensing neurons disorients mosquitoes." Nature 474, no. 7349 (2011): 87-91.

Valderramos, Stephanie G., Juan-Carlos Valderramos, Lise Musset, Lisa A. Purcell,

Odile Mercereau-Puijalon, Eric Legrand, and David A. Fidock. "Identification of a mutant

PfCRT-mediated chloroquine tolerance phenotype in Plasmodium falciparum." PLoS Pathog 6, no. 5 (2010): e1000887.

von Seidlein, Lorenz, and Brian M. Greenwood. "Mass administrations of antimalarial drugs." Trends in parasitology 19, no. 10 (2003): 452-460.

Wahlgren, Mats, and Maria Teresa Bejarano. "Malaria A blueprint of ‘bad air’."Nature 400, no. 6744 (1999): 506-507.

Weihe, Wolf H., and Raf Mertens. "Human well-being, diseases and climate." NASA

19990036600 (1991).

Wells, Timothy NC, Rob Hooft van Huijsduijnen, and Wesley C. Van Voorhis. "Malaria medicines: a glass half full?." Nature Reviews Drug Discovery (2015).

Wang, Zenglei, Min Liu, Xiaoying Liang, Salil Siriwat, Xiaolian Li, Xiaoguang Chen,

Daniel M. Parker, Jun Miao, and Liwang Cui. "A flow cytometry-based quantitative drug sensitivity assay for all Plasmodium falciparum gametocyte stages." PloS one 9, no. 4 (2014): e93825.

White, Nicholas J. "Antimalarial drug resistance." Journal of Clinical Investigation 113, no. 8 (2004): 1084.

20 White, Nicholas J. "The role of anti-malarial drugs in eliminating malaria." Malaria

Journal 7, no. Suppl 1 (2008): S8.

White, Nicholas J. "Primaquine to prevent transmission of falciparum malaria." The

Lancet infectious diseases 13, no. 2 (2013): 175-181.

World Health Organization. "Control and elimination of plasmodium vivax malaria: a technical brief." (2015).

World Health Organization. "World Malaria Report 2014 (2014)." WHO: Geneva (2014).

Yohannes, Mekonnen, and Eline Boelee. "Early biting rhythm in the afro‐tropical vector of malaria, Anopheles arabiensis, and challenges for its control in Ethiopia." Medical and veterinary entomology 26, no. 1 (2012): 103-105.

21

Chapter 2

Tafenoquine Drug Combinations in Asexual and Sexual parasites

*Status: In prep for submission to Antimicrobial Agents and Chemotherapy

2.1 Introduction

The current first- line treatment for Plasmodium malaria parasites is artemisinin combination therapy (ACT), comprising a co-formulated fixed-dose tablet of a fast-killing and rapidly eliminated artemisinin (ART) derivative, combined with a slower-acting partner drug with a longer elimination half-life in humans [1]. The majority of ACTs however, are highly effective against the asexual blood parasites that cause clinical symptoms, but ineffective against the infectious sexual stage gametocytes in P. falciparum and the dormant liver stage parasites, namely hypnozoites of Plasmodium vivax [2]. To address this, the World Health Organization recommends prescribing a 0.25mg base dose/kg of the only licensed gametocytocidal and liver schizonticidal drug, primaquine (PMQ), an 8-aminoquinoline, following either chloroquine (CQ) or ACT to suppress human to mosquito transmission and P. vivax relapses respectively in areas of emerging P. vivax CQ resistance and emerging P. falciparum ART resistance [3]. Owing to the shortage of antimalarial therapies with similar parasite stage targets as PMQ, the Walter Reed

Army Institute of Research in collaboration with GlaxoSmithKline began testing the antimalarial efficacy of a 5-phenoxyl PMQ derivative, tafenoquine (TFQ), also denoted as WR 238605/SB-

252263. [4]. Compared to PMQ, TFQ has a longer elimination half- life of 2 weeks compared to the 6-8 hours of PMQ, appears to have better bioavailability and enhanced hypnozoite suppression [5, 6]. TFQ, is currently in late stage clinical trials for the radical cure of P. vivax [5]

22 as a single 300mg dose after either 3 days of chloroquine, or ACT, a drug regimen similar to the current one of PMQ for P. vivax (Unpublished data: Clinical Trials.gov identifier: NCT01376167 and NCT02184637 respectively). A completed clinical trial showed a higher protection against P. vivax relapse for a CQ+TFQ regimen compared to CQ alone [7].

TFQ’s mechanism of action is unknown but studies in Leishmania spp. and Trypanosoma spp. have yielded possible hypotheses that it acts by inducing mitochondrial dysfunction [8, 9].

Studies in Plasmodium are yet to reveal the link between mitochondrial function and TFQ. Both

TFQ and PMQ have the disadvantage of causing severe hemolytic anemia in individuals with glucose-6-phosphate dehydrogenase (G6PD) deficiency [7]. Screening for this common X-linked genetic disorder must be carried out prior to dosing with PMQ or TFQ for transmission blocking and/or killing dormant liver stage parasites. The classic Fluorescent Spot Test and the recently developed CareStart (AccessBio, Somerset, NJ) G6PD kit are two effective rapid diagnostic tests for this, although test results must be taken with caution since heterozygous women carriers may show false negative results [10].

Should TFQ be deployed for P. vivax radical treatment in a P. falciparum and P. vivax co-endemic zone, knowledge of drug-drug interactions between either ACT or CQ in the presence of TFQ will be essential, since drug-drug interactions within the human host could enhance or reduce the efficacy of all the drugs involved. In order to study this, interactions in P. vivax would have to be investigated ex-vivo or in-vivo since continuous P. vivax in vitro culture is challenging and unstable, requires a constant supply of reticulocytes and yields very low parasitemia [11, 12].

This study therefore focused on only P. falciparum in vitro drug-drug interactions with ACT- partner drugs and since mixed P. falciparum and P. vivax malaria infections are extremely common in co-endemic countries, the goal was to determine off-target benefits/limitations of

TFQ dosage to P. falciparum parasite inhibition.

23 Few in vitro studies pertaining to TFQ drug-drug interactions have been performed. One study reports either antagonism or additivity of TFQ when combined with CQ or AQ respectively, whereas another conflicting study reports synergism when combined with CQ [13,

14]. Another study reports synergism when TFQ is combined with Artemisinin at a single 1: 1 ratio [15]. Neither of these tested drug-drug interactions against the full panel of ACT- partner drugs and neither of them tested these interactions in sexual stage gametocytes. Drug-drug interactions of these are expected to differ since asexual parasites are sometimes as much as 2000 fold more susceptible to most antimalarials especially the first- line ACTs, compared to their sexual gametocyte-stage parasite counterparts [16]. With regard to PMQ, a recently published study investigated the effects of PMQ-ACT partner drugs in vitro on asexual stage and sexual stage gametocyte P. falciparum malaria parasites, reporting mostly synergistic drug-drug interactions [16]. The study focused on only the longer lasting ACT partner drugs since it was assumed that ART derivatives did not stay in the blood stream long enough to greatly impact the drug-drug interactions.

Here, for the first time, the in vitro interactions between TFQ and six ACT schizonticides, as well as chloroquine in asexual and sexual stage P. falciparum are investigated.

Using a SYBR Green I method for asexual stage and flow cytometry based methods for gametocytes, fractional inhibitory concentrations and isobolograms derived from pharmacologically relevant fixed ratios are used to determine decreased (antagonistic) or enhanced (synergistic) drug efficacy of ACT-partner drugs in the presence of TFQ. Only the long lasting ACT-partner drugs are used here, namely the 4-aminoquinolines amodiaquine (AMQ) and naphthoquine (NQ), the bisquinoline piperaquine (PPQ), aryl amino-alcohols lumefantrine (LMF) and mefloquine (MFQ) and the mannich base, pyronaridine (PND) because TFQ and ART derivatives have durations to reach plasma concentrations (Tmax) of 15 hours and <1.8hours respectively, therefore are less likely to interact in vivo [17–19]. These correspond to current

24 ACTs namely artesunate- amodiaquine, artemisinin- naphthoquine, dihydroartemisinin- piperaquine, artemether- lumefantrine, artesunate- mefloquine and artesunate- pyronaridine, all of which are used in different malaria endemic regions (see Table 2-2, Table 2-3) [1, 20]. The 4- aminoquinoline chloroquine (CQ) is included as well because in areas where P. vivax has not yet developed, CQ is still the first- line treatment prior to a PMQ dosage [2]. Drug-drug combination ratios were chosen to reflect both peak blood plasma concentrations (Cmax) of TFQ and ACT- partner drugs, as well as the in vitro median inhibitory drug concentrations (IC50s) of each individual drug to ensure that proper drug-response curves were obtained (see Tables 2-3 and 2-

4). Parasites with different genetic backgrounds and differential drug susceptibilities to both CQ

(Pfcrt K76T mutation) and the ART metabolite dihydroartemisinin (PfKelch13 R539T mutation) are used to assess whether genetic factors might affect the observed asexual stage drug-drug interactions. 3D7αtubIIGFP parasites expressing GFP under the gametocyte-specific α - tubulin II gene promoter are used for the gametocyte drug interaction assays [21]. Differential TFQ drug interactions between asexual and sexual stage parasites are reported.

Overall, our data sheds some light upon TFQ interactions with ACT partner drugs in vitro for P. falciparum and re-emphasizes the need for future interaction studies to include gametocyte stages to test pharmacologically-relevant drug concentrations in new antimalarial combinations.

Follow-up in vivo experiments, either using humanized mice/humans infected with P. vivax/ P. falciparum with varying G6PD activity will be necessary to verify these results.

25 Table 2-1. Structural classification of antimalarial drugs used in study. Canonical SMILES were obtained from PubChem, and structures drawn using ChemDraw 3D Pro14.0 software.

Antimalarial Classification Chemical structure

Chloroquine 4-aminoquinoline

Amodiaquine 4-aminoquinolines

Naphthoquine

Piperaquine Bis-4-

aminoquinoline

Mefloquine Aryl amino-

alcohols

Lumefantrine/

Benflumetol

Pyronaridine Aza-acridine

mannich base

26

Tafenoquine 8-aminoquinoline

Table 2-2 Summary of available artemisinin combination therapies (ACTs). List is a compilation from [1, 3, 20, 22]. N/A represents drugs manufactured in China but data on world usage is missing. EM, WP, SEA and LA stand for Eastern Mediterranean, Western Pacific, S. East Asia and Latin America respectively. Artemisinin/derivative- Brand names World Region in Use partner antimalarial co- formulations (ACT) Artesunate- amodiaquine Coarsucam®, ASAQ-Winthrop®, Co- Africa and EM Artusan®, MalmedFD® Artemisinin- naphthoquine Arco® Africa and WP Artemether- lumefantrine Coartem®, Riamet®, Faverid®, Amatem®, Africa, EM, WP, Lonart®, AL®, Artemine®, Fantem®, LA,SEA Artefan®, Lumartem®, Lumet® Artesunate-mefloquine ASMQ®, Artequin®, Mefliam Plus® Africa, WP, LA, SEA Artesunate- pyronaridine Pyramax® N/A (China) Dihydroartemisinin- Duocotecxin®, Eurartekin®, Eurartesim®, Africa, SEA,WP piperaquine Combimal®, P-Alaxin® Artemisinin- piperaquine Artequick® N/A (China)

27 2.2 Materials and methods

Chemical reagents

Parasite culture medium starters RPMI 1640 and Albumax II were purchased from Gibco

Life Technologies (Grand Island, NY, USA). Antimalarials for drug susceptibility assays namely chloroquine diphosphate (CQ) [Molecular Weight (MW): 515.86], amodiaquine dihydrochloride dihydrate (AMQ) [MW: 464.81], mefloquine hydrochloride (MFQ) [MW: 414.77] and tafenoquine succinate (TFQ) [MW: 581.58] were purchased from Sigma-Aldrich (St. Louis,

MO). Piperaquine tetraphosphate (PPQ) [MW: 999.55] was kindly provided by Chongqing

Kangle Pharmaceuticals (Chongqing, China). Naphthoquine phosphate (NQ) [MW: 605.94], pyronaridine tetraphosphate (PND) [MW: 910.03] and lumefantrine (LMF) [MW: 528.94] were kindly provided by Kunming Pharmaceuticals (Yunnan, China). PND, AMQ, NQ and CQ were dissolved in distilled water to 20mM stock concentrations. PPQ was dissolved in 90% methanol +

10% 1M HCl to 10mM [23]. TFQ, MFQ, and LMF were dissolved in 100% dimethyl sulfoxide

(DMSO: Alfa-Aesar, Ward Hill, MA) to stock concentrations of 40mM, 20mM and 40mM respectively. Cellulose acetate or nylon 0.2um membrane syringe filters (VWR International,

Radnor, PA) were used to sterilize water and DMSO dissolved drugs respectively. All drug stocks were stored at -80°C until ready for use. Chloroquine stocks were protected from light. Working drug concentrations ranging from 20mM to 100nM were freshly prepared in malaria complete medium (MCM: see In vitro parasite culture below) the same day of drug inhibition assay set up.

SYBR Green I PCR Master Mix for asexual parasite growth inhibition assays was purchased from Invitrogen (Eugene, OR, USA). Giemsa azure eosin blue for parasite staining was purchased from Fluka Chemical Corp. (Ronkonkoma, NY). Percoll for density gradient centrifugation was purchased from Sigma.

28 Parasite culture

P. falciparum laboratory- adapted strains of varying genetic backgrounds 3D7 (Africa:

CQ sensitive), HB3 (Honduras: CQ sensitive), 7G8 (Brazil: CQ resistant Pfcrt K76T), DD2

(Indo-China: CQ resistant Pfcrt K76T) and IPC5202 (Cambodia: ART resistant Pfkelch13:

R539T) were obtained from MR4 (Manassas, VA) and maintained at 37°C under an atmosphere of 90% N2, 5%O2, and 5% CO2 in asexual malaria complete medium (MCM) containing RPMI

1640, 25 mM NaHCO3, 25 mM HEPES (pH 7.4), 11 mM glucose, 0.367 mM hypoxanthine and

5 µg/L gentamycin supplemented with 0.5% Albumax II [24, 25]. Asexual MCM was changed daily and percentage parasitemia maintained below 6.5%, at 2.5% hematocrit in O+ human whole blood (Biological Specialty, Colmar, PA).

A transgenic 3D7αtubIIGFP parasite strain with green fluorescent protein expression under the α- tubulin II promoter was kindly provided by Jun Miao, and sexual stage gametocytes induced from it as previously described in [26] with a few alterations. Heparin sodium salt

(Sigma-Aldrich, St. Louis, MO) was used instead of n-Acetyl Glucosamine to inhibit asexual parasite proliferation in gametocyte cultures and late stage II gametocytes were purified by a

75%/35% percoll gradient (pH 7.4) on day 4 post-gametocyte induction [27]. Gametocytes were maintained in gametocyte MCM that unlike asexual MCM contained RPMI 1640 supplemented with 0.25% Albumax II + 5% heat-inactivated AB human serum (Interstate Blood Bank,

Memphis, TN).

Asexual parasite SYBR Green I drug inhibition assay

Parasites were synchronized by sterile pre-warmed 5% D-sorbitol (wt/vol) (J.T. Baker,

Center Valley, PA) treatment for 9 minutes to enrich for ring stage parasites four days following

29 thawing from liquid nitrogen [28]. Prior to drug assay set-up, percentage parasitemia of cell culture was determined by microscopy using 10% Giemsa in distilled water at pH 6.8. In preparation for drug assay set up, a total of 1000 cells was counted and parasites diluted to 0.5% parasitemia and 2% hematocrit with addition of 50% freshly washed red blood cells in incomplete medium (MCM minus Albumax II or serum). Cells were pelleted prior to dilution from their complete medium by centrifugation with a Heraus Megafuge 16R centrifuge (Thermo Scientific,

Waltham, MA) at 900xg for 5 minutes. The prepared parasite sample was loaded into pre-loaded black 96-well plates with 2X 100ul antimalarial drug working solutions diluted 2-fold to a final volume of 200ul to generate a final hematocrit of 1%, 0.5% parasitemia and 1X drug concentration per well. Negative control wells without drug with either MCM, DMSO or 10%

1M HCl/90% methanol dissolved in MCM corresponding to total amounts in working drug solutions were set up in parallel for drugs dissolved in solvents other than water. Where possible,

DMSO concentrations were kept below 0.4%. Plates were incubated for 72 hours in at 37°C in a

5% humidified incubator followed by transfer to -20°C for at least 16 hours to facilitate cell lysis.

Lysis buffer (100ul) consisting of 20mM Tris ( pH 7.4), 5mM EDTA, 0.008% wt/vol saponin and 0.08%; vol/vol Triton X-100 with 0.2ul of SYBR Green I was added to each 96-well plate sample and mixed gently [29]. Plates were the then incubated at room temperature for at least 1 hour and SYBR Green I fluorescence corresponding to parasite density was determined using a

FluoStar Optima plate reader (BMG Labtech, Cary, NC). Median inhibitory concentrations

(IC50s) for each drug alone or in combination with TFQ were determined by three-parametric non-linear regression analysis of the resultant drug response growth curves using GraphPad Prism

5 software (La Jolla, CA).

30 Sexual parasite flow cytometry-based drug inhibition assay

Stages II-V, corresponding to days 4-12 post gametocyte induction) and stage IV gametocytes, corresponding to days 8 & 9 of the 3D7αtubIIGFP strain at 0.04% gametocytemia and

1% hematocrit were incubated in 2-fold serially diluted pre-loaded drug plates for 48 hours to determine stage-specific TFQ inhibitory effects and TFQ-ACT partner drug (drug X) interactions.

Starting drug concentrations for gametocytes ranged from 2mM to 10mM depending on the drug.

Following incubation, parasite samples per well were diluted in pre-warmed 1X Hank’s balanced salt solution (pH 7.4) containing 10mM D-Glucose, 10mM HEPES, 270mM KCl, 270mM NaCl,

1.5mM Na2HPO4 (Cold Spring Harbor Protocol 2008; doi: 10.1101/pdb.rec11561) to minimize auto-fluorescence of MCM to 0.4% hematocrit. Resulting green fluorescence from live parasites that survived antimalarial drug exposure were detected by flow cytometry; 25,000 events per sample were collected on a Guava EasyCyte HT flow cytometer (EMD Millipore Corp., Billerica,

MA). Fluorescence intensity (FI) calculated by FI= normalized events x mean green fluorescence using Flow Jo version 10 software plotted against drug concentration generated drug dose-effect curves from which IC50s for individual and TFQ-drug X combinations were calculated using

Graph Pad Prism 5 software .

Drug-drug combination assays

For asexual parasites, TFQ was combined with ACT partner drugs (drug X: LMF, PPQ,

MFQ, AMQ, NQ, PND) and CQ at fixed ratios reminiscent of both peak plasma concentrations in humans (Cmax) and in vitro IC50s (Tables 2 and 3). IC50s of individual drugs, and drugs in combination with TFQ against 5 laboratory adapted stains namely 3D7, 7G8, DD2, IPC5202 and

HB3 were determined. For sexual stage gametocytes, TFQ was combined in fixed ratios of 3:1,

31 1:1 and 1:3 in hundreds of µM concentrations as described elsewhere [16]. Three biological replicates, each in duplicate at a consistent 0.04% gametocytemia and 0.5% asexual parasitemia to minimize inoculum effects were performed [30]. Apparent IC50s of combinations were used to determine fractional inhibitory concentration indices (FICindex) using the formula (FICTFQ=

Apparent IC50 of drug X in combination with TFQ/IC50 of TFQ alone) + (FICX= Apparent IC50 of drug X in combination with TFQ/IC50 of drug X alone) where drug X represents the ACT-partner drugs under investigation or CQ. An average of FIC indices per fixed ratio represents an additive drug-drug interaction if = 1, a synergistic interaction if <1 and an antagonistic interaction if >1 corresponding to a straight diagonal line, concave curve (left of the diagonal) or convex curve

(right of the diagonal) isobologram of FICTFQ against FICx respectively [13].

Statistical analysis

One-way analysis of variance (ANOVA) and Tukey HSD tests were used to determine the differences between IC50s in different gametocyte stages as well as in IC50s for the different strains to the eight drugs under investigation using SAS University Edition Software (Cary, NC).

A Shapiro-Wilk test was used for testing the extent of data normality and a Levene’s test for homogeneity of weight variance used to determine equality of variances. A p-value of <0.05 was considered significant. A Tukey HSD test was used for multiple comparisons. Least square mean

FICindex was computed along with 95% confidence intervals to determine the extent of drug interaction deviation from additivity. A one sample t-test was performed to determine whether

FICs were equal to or unequal to 1 in either direction to determine synergism (significantly less than one) and antagonism (significantly greater than one).

32 Table 2-3 Pharmacokinetics of drugs used in this study. ACT-partner drugs, as well as CQ and

TFQ and their corresponding elimination half-lives (T1/2), peak plasma concentrations following a single dose (Cmax) and time to peak (Tmax). Amodiaquine’s active metabolite, monodesethyl amodiaquine (mdA) in vivo has a longer elimination half-life compared to its parent drug, AMQ. Data sources include [1] and the individual cited papers in the table. Piperaquine pharmacokinetics vary between starved and fed state, the value cited here was from a fed state.

Artemisinin and derivatives have Tmax values <1.8hr therefore are not used in this experiment

[17, 18]. Cmax micromolar (µM) values are computed from ng/ml or µg/l data reported in cited sources for easier translation to in vitro drug inhibitory concentrations. *TFQ and CQ are used in this study but are not ACT partner drugs.

Partner Drug Elimination T1/2 Cmax (µM) Tmax(h) References mdA/ Amodiaquine (AMQ) 3hr/9-18 days 0.06 2.0 [31]

Naphthoquine (NQ) 12 days 0.02-0.04 3.5 [32, 33]

Lumefantrine (LMF) 4-5 days 13-18 6.0 [34, 35]

Mefloquine (MFQ) 14-21 days 4-7 45-52 [36, 37]

Pyronaridine (PND) 10 days 0.08-0.54 0.5-3.2 [38]

Piperaquine (PPQ) 35 days 0.2 4 [39]

Tafenoquine (TFQ)* (300mg) 14days 0.32-0.7 15 [4, 19]

Chloroquine (CQ)* (600mg) 9 days 0.7-2 6.6 [40–42]

33 Table 2-4 Ratios used for asexual TFQ- ACT partner drug interactions. ACT-partner drugs were combined, in 2-fold decreasing sequence ranging from 4 - 0.05 µM, to an increasing sequence of

TFQ ranging from 0.5 – 4 µM in fixed micromolar concentrations taking into account the Cmax values (table 2-3) but maintaining concentrations encompassing the in vitro IC50 to asexual parasites. 5-fold less of LMF Cmax was used so as not to leave out data points on the exponential and plateau part of the dose-effect curve. Ratio of TFQ (µM): 0.5 : 1: 2: 4: ACT partner ACT-Partner (µM) Amodiaquine (AMQ) 0.4 0.2 0.1 0.05 Naphthoquine (NQ) 0.4 0.2 0.1 0.05 Lumefantrine (LMF) 3.5 1.75 0.88 0.44 Mefloquine (MFQ) 4 2 1 0.5 Pyronaridine (PND) 0.64 0.32 0.16 0.08 Piperaquine (PPQ) 0.8 0.4 0.2 0.1 Chloroquine (CQ) 2.4 1.2 0.6 0.3

2.3 Results

ACT-partner drugs differentially inhibit asexual and sexual parasites

Prior to drug interaction studies, IC50s of each of the drugs under investigation were tested against 5 laboratory adapted asexual parasite strains that are either chloroquine sensitive

(CQS: HB3, 3D7), chloroquine resistant (CQR: 7G8, DD2 ) or artemisinin resistant (ART-R:

IPC5202) to determine whether genetic background significantly affects drug susceptibility. Drug susceptibilities were similar across all five strains for most drugs except for chloroquine as expected of the previously known resistant strains, as well as for IPC5202, the ART-R strain from

Cambodia. The 7G8 and IPC5202 strains also exhibited significantly lower and higher IC50

34 values to MFQ (ANOVA p<0.0001) (Table 2-5) respectively. TFQ drug susceptibilities across strains were not significant.

Drug susceptibilities for sexual stage transgenic parasite strain 3D7αtubIIGFP gametocytes were determined using flow cytometry methods to each of the eight antimalarial drugs under investigation. Additionally, TFQ inhibition of stages II-V gametocytes was tested to determine if gametocyte developmental stage affects TFQ efficacy. There was no significant difference

(ANOVA p>0.05) between the IC50s of TFQ in each of these stages (Table 2-6). As expected, median inhibitory drug concentrations were in the micromolar range for gametocytes, compared to asexual parasites that were in the nanomolar range for all drugs except TFQ (Table 2-5). LMF and PPQ had very low gametocyte inhibition compared to TFQ and AMQ. The rest of the drugs had moderate gametocytocidal activity in the micromolar (µM) range.

35 Table 2-5 Drug susceptibilities of asexual parasites (Mean ±SEM). Lab adapted strains were tested against ACT- partner drugs, as well as chloroquine (CQ), and tafenoquine (TFQ) using the SYBR Green I method. Parts of the world where the strains originate are shown in parentheses. CQS, CQR and ART-R stand for chloroquine sensitive, chloroquine resistant and the artemisinin metabolite, dihydroartemisinin resistant respectively. * indicates significantly different drug susceptibilities to a drug (ANOVA p<0.001). Drug HB3 (Honduras 3D7 (Africa 7G8 (Brazil DD2 (Indo- IPC5202 IC50s(nM)/ CQS) CQS) CQR) China CQR) (Cambodia STRAIN ART-R) AMQ 9.9±1.8 4.8±0.7 19.0±3.8 9.8±1.0 10.3±0.4

NQ 12.1±2.0 4.1±0.9 3.9±0.1 10.9±1.3 5.9±2.3

LMF 8.9±1.9 11.6±0.7 6.9±1.0 11.9±1.2 38.7±5.0

MFQ 16.5±1.6 13.0±0.4 1.6±0.6* 11.6±1.3 28.8±2.8*

PND 3.3±0.5 3.8±0.8 4.8±0.9 4.9±0.6 2.7±0.1

PPQ 17.8±1.8 23.6±0.6 31.6±2.6 15.3±1.5 19.1±0.7

CQ 18.5±1.5 24.5±1.4 318.0±14.4* 308.7±24.5* 416.3±27.7*

TFQ 1875.2±418.9 2132.7±369.3 2380.5±472.3 1129.8±201.0 2072.3±88.2

Table 2-6 Median TFQ inhibitory concentrations by gametocyte stage. Gametocyte developmental days post-induction corresponding to stage identification are as follows; day 4 (stage II), 6 (stage III), 8 or 9 (stage IV) and 11 or 12(stage V) as specified [43, 44]. Gametocyte Stage II III IV V

TFQ IC50 (µM ± SEM) 8.8±2.4 8.1±0.5 9.8±1.4 11.1±1.7

36 Table 2-7 Drug susceptibilities of 3D7αtubIIGFP gametocytes. Median inhibitory drug concentrations of stage IV 3D7αtubIIGFP gametocytes were obtained by flow cytometry followed by dose-effect curve IC50 determination.

αtubIIGFP Drug IC50s (µM) ±SEM of 3D7 gametocytes AMQ 5.9±1.26 NQ 142.5±15.75 LMF 723.6±79.2 MFQ 16.5±11.65 PND 25.1±7.9 PPQ 197.7±12.6 CQ 27.6±0.3 TFQ 9.8±1.4

TFQ synergizes ACT partner drugs in asexual parasites

In order to determine the drug-drug combinations between TFQ and ACT- partner drugs in asexual parasites, fractional inhibitory concentrations (FICs) were used to construct isobolograms and to elucidate whether the combinations are antagonistic (Mean sum of FIC >1), synergistic (Mean sum of FIC <1) or additive (Mean sum of FICs =1) suggesting decreased, enhanced or no effect drug interactions respectively. TFQ synergized the inhibition of ACT- partners in most strains, except in the 3D7 LMF- TFQ and DD2 NQ-TFQ at ratios of high drug X

(LMF or NQ): low TFQ where it showed an antagonistic relationship but a synergistic relationship to all other fixed ratios (Table 2-8, Appendix B).

37 ACT-partner + TFQ combinations differentially affects sexual stage parasites

In order to determine any enhancement or reduction in drug potency of ACT- partner drugs in combination with TFQ, TFQ and ACT- partner drugs were combined in fixed ratios of

1:3, 1:1 and 3:1. FICs and resultant isobolograms were used to determine the interaction effects.

Only piperaquine (PPQ) and pyronaridine (PND) appeared to interact with TFQ synergistically

(Table 2-8, Appendix B).

Table 2-8 Summary of TFQ-ACT-partner drug interactions. Asexual and sexual parasites are shown. Chloroquine or Artemisinin sensitive and resistant parasites, as well as 3D7αtubIIGFP gametocyte drug-drug interactions were investigated by combining TFQ and each drug in the ratios shown in Table 2-4 for asexual parasites and 1: 1, 1:3 and 3:1 in gametocytes. The sum of fractional inhibitory concentrations of each of the 4 ratios per drug were used to construct isobolograms (Appendix B) and the mean Sum of FICs used to interpret synergistic (Syng <1), antagonistic (Antg >1) and additive (Addt =1) interactions. Interactions that deviate from synergism in asexual and antagonism in sexual parasites are indicated in bold. * indicates that at least one ratio showed a different interaction from the overall result. DRUG + TFQ HB3 3D7 7G8 DD2 IPC5202 3D7αtubIIGFP Amodiaquine (AMQ) Syng Syng Syng Syng Syng Antg

Naphthoquine (NQ) Syng Syng Syng Syng* Syng Antg

Lumefantrine (LMF) Syng Syng* Syng Syng Syng Antg

Mefloquine (MFQ) Syng Syng Syng Syng Syng Antg

Pyronaridine (PND) Syng Syng Syng Syng Syng Syng

Piperaquine (PPQ) Syng Syng Syng Syng Syng Syng

Chloroquine (CQ) Syng Syng Syng Syng Syng Antg

38 2.4 Discussion

Resistance of P. vivax to CQ is spreading and a shift from CQ to ACT followed by PMQ to kill liver stage parasites in areas of emerging P. vivax CQ resistance is recommended [45].

Currently, PMQ is the only drug licensed for the radical cure of P. vivax. Its derivative, TFQ, is in late stage clinical development for a similar role, given as a single 300mg dose after a 3-day CQ regimen owing to its inhibitory properties against dormant liver stage parasites [5]. Since ACTs are already the first- line treatment for P. falciparum, this study aimed to investigate TFQ-ACT interactions in areas co-endemic for P. falciparum and P. vivax since mixed infections as well as mixed diagnoses are common [46, 47]. If deployed for P. vivax, TFQ will inevitably come into contact with either CQ or ACT, therefore assessing whether its presence is beneficial for drug potency is necessary for both P. falciparum and P. vivax parasites. As this was an in vitro study,

P. vivax was not used because of the difficulty of maintaining it in continuous culture and obtaining enough of it to use in drug susceptibility assays [11].

Using CQ and ART resistant or sensitive asexual and sexual P. falciparum parasites, it was determined that TFQ had overall mostly synergistic interactions with asexual parasites when combined with ACT-partner drugs (Table 2-8). This is desirable in the field because an augmentation of ACT drug inhibition in the presence of TFQ will likely result in faster cure rates and less asexual parasites surviving to later differentiate into infectious gametocytes. This synergism could be due to TFQ and the ACT-partner drugs having different mechanisms of action thereby evading competitive inhibition of the same drug binding site. TFQ is reported to exhibit low levels of inhibition of hematin polymerization which is likely to supplement the action of the quinolines in this study by further inhibiting hemoglobin digestion in the asexual parasites, but its action in the gametocyte stages remains speculative [48, 49]. In Leishmania donovani and Trypanosoma brucei, TFQ has been reported to interfere with mitochondrial

39 function leading to apoptosis [8, 9] However, in accordance with other findings, TFQ had low drug inhibition in asexual parasites, compared to the ACT-partner drugs in this study suggesting that its effects relating to hemoglobin digestion are negligible (Table 2-5) [13, 14, 16]. The TFQ-

LMF interaction of the CQ and ART sensitive 3D7 parasite was antagonistic at high LMF concentrations, but not at the high TFQ: low LMF concentrations. It is unclear why because LMF was never used in Africa, and this particular strain has single copy number and a wild-type allele for Pfmdr1that is associated with LMF tolerance. This observation might just be a factor of the highest drug concentrations used skewing the results towards antagonism. A similar effect was observed when a high LMF and low primaquine ratio were combined [16]. The CQ-R strain,

DD2 was also antagonistic at high NQ: low NQ ratios and synergistic in all others, suggesting either a drug concentration effect as seen in 3D7 LMF-TFQ or a possible role of its high Pfmdr1 copy number and Pfcrt mutation in reduced susceptibility to the closely related NQ to CQ, but no studies have reported this link to date since NQ, although used in China in the 1970s has received very little attention to date [50].

Few studies have investigated TFQ interactions in vitro, reporting synergistic interactions with CQ and ART respectively [14, 15]. Another study reported, instead, an antagonistic relationship of TFQ to CQ, however unlike the former CQ-TFQ study, ratios closer to in vitro

IC50s, rather than Cmax were used, resulting in pharmacologically relevant concentrations for CQ but not TFQ, because in order to fulfil a 1:3 ratio with a TFQ IC50 of approximately 2µM, a starting concentration of >20µM would have to be used, compared to the 0.5-4µM concentration range used here [13]. Taken together, TFQ appeared to have a positive inhibitory effect on asexual P. falciparum when combined to ACT-partner drugs, regardless of genetic background and CQ/ART susceptibility at fixed ratios taking the Cmax into consideration. It should be noted, however, that only the long- lasting ACT-partner drugs were used here because the artemisinin

40 components will have long been eliminated before the peak TFQ concentration is reached as shown in Table 2-3.

Compared to asexual parasites, gametocyte IC50 values were in the micromolar range, consistent with other studies [16, 51, 52]. Since many antimalarial drugs are active against gametocytes up to stage III, presumably coinciding with a halt in hemoglobin digestion, the drug susceptibilities of gametocytes by developmental stage was investigated [21]. Drug inhibition assays were performed on day 4 (late stage II), day 6 (stage III), day 8 or 9 (stage IV) and day 11 or 12 (stage V). Results showed no significant change in gametocyte susceptibility to TFQ over time (Table 2-6).

With the exception of PND and PPQ, all TFQ-ACT-partner interactions were antagonistic at the stage IV gametocyte stage, unlike at the asexual stage where synergism was the norm. For gametocytes, however, fixed- ratios for gametocytes used were highly divergent from Cmax values to account for the high (µM range) of in vitro gametocyte drug inhibition; pharmacologically relevant concentrations would not have resulted in dose-effect curves necessary to calculate IC50s , this could have affected the outcome, like in asexual studies using similar ratios as discussed earlier [16]. It is hard to deduce by genotype alone why gametocyte interactions are different from the asexual parasites in this study, because only one transgenic strain was used, that is otherwise susceptible to all drugs tested at the asexual stage. Bell, in a review of antimalarial drug interactions cites some probable causes for synergism namely that 1) both drug A and B being studied might bind the same protein such that a conformational change increases the binding of drug B 2) drug A might be binding to a transporter causing increased uptake of drug B 3) drugs A and B likely form a more toxic complex structure and 4) drug B activity might be enhanced by interacting with a more active metabolized form of drug A, although explanation 4) can be ruled out since this study was not carried out in the presence of liver microsomes that could metabolize any of these drugs [48]. All that can be deduced here is

41 that the 4-aminoquinolines and the aryl amino-alcohols have antagonistic interactions with TFQ in gametocytes. Interestingly, the definitions for synergy or antagonism, however, are arbitrary and it may very well be that had the threshold mean sum of fractional inhibitory combinations been set at 2 or 4 instead of 1, mostly synergistic or additive interactions with TFQ would have been observed [48]. It is worth noting that some of the drugs like LMF that in this in vitro assay displayed very high IC50s for gametocytes have been reported to have sporontocidal activity [53].

TFQ itself has some sporontocidal activity, at least in P. berghei and P. vivax therefore a compound effect of the synergism observed at the asexual stage following the rapid killing action of ART or its derivatives not tested here, as well as the increased comparative susceptibility of certain drugs in the early vs late gametocyte stages plus the sporontocidal effect at the mosquito transmission stages should severely suppress a P .falciparum infection, even though P. vivax is the primary target of TFQ development [54–56].

Limitations to this study were the omission of P. vivax in vitro culture, since it might act differently in the presence of ACT and TFQ. Only one transgenic parasite line was available for this study; another GFP expressing parasite strain under the direction of the αtubII [PlasmoDB:

PF3D7_042230] gene promoter with a background other than 3D7 might exhibit different drug- drug interactions. There is currently no female-gametocyte specific marker, so gametocyte drug susceptibilities were tested for both male and female gametocytes, and not individually. Studies have reported that there is differential tolerance to drugs between the sexually dimorphic sexual stages with females being more tolerant [57–59]. As discussed earlier, the fixed-ratios for gametocytes were higher than Cmax, possibly skewing the results. Lastly, AMQ was used here instead of its metabolite mono desethyl- amodiaquine that is more likely to interact with TFQ in vivo. It is suggested that a ratio of the parent drug: the metabolite be employed in vitro for more realistic results [60].

42 Significant contributions of this study however, include the investigation, for the first time of TFQ-ACT-partner drug-drug interactions on parasites with different genetic background at pharmacologically relevant drug concentrations and the use of gametocytes, in addition to asexual stage parasites in the interaction studies; only one other study has included these to investigate primaquine-ACT interactions [16]. In vivo studies will need to be carried out to validate these results as many host factors come into play. For example, some drugs like CQ and

MFQ are known to increase the number of gametocytes, which might be problematic assuming antagonism is reproduced in vivo [61, 62]. Other drugs like TFQ, MFQ, LMF and PPQ have a higher bioavailability when taken with food [4, 22, 39] . With regard to mixed infections, P. falciparum and P. vivax mixed infections have been reported to show differences in severity, onset of one or the other, the number of gametocytes etc compared to single infections [47, 63].

In addition, TFQ Cmax will also be variable depending on cytochrome P450 metabolism and gender, with women displaying higher plasma concentrations than males [64, 65].

Taken together, TFQ’s mostly synergistic effect on at least the asexual stage parasites is good news because although it is being targeted for P. vivax, it has some benefit to P. falciparum infection in the case of mixed infection. Hopefully if the presence of TFQ with ACT enhances the asexual inhibitory effect, less asexual parasites will commit to sexual stage gametocytes that are infectious to mosquitoes. Although being developed for P. vivax, it is important to note that earlier studies of TFQ against Plasmodium falciparum showed chemo-prophylactic activity [6,

66]. Care will need to be taken when choosing an ACT to take prior to TFQ dosage as the least desirable ones are those that show a consensus of antagonism in similar in vitro and in vivo studies. A study testing the dihydroartemisinin-piperaquine and artemether-lumefantrime ACT interactions with TFQ has been completed elsewhere (Unpublished Clinicaltrials.gov identifier:

NCT02184637). In the field, flow cytometry methods using transgenic parasites will not be possible but the sensitive real-time quantitative nucleic acid sequence-based amplification (QT-

43 NASBA) can perhaps be used to determine the duration of gametocyte clearance with the different combinations [67] . TFQ does however cause severe hemolysis in individuals with reduced G6PD activity so in vivo studies in humanized mice and humans with both normal and reduced G6PD activity studies will also need to be undertaken [68]. In summary, the future of malaria chemotherapy lies in the identification of combination therapies so as to maximize inhibitory effects to parasites and to slow down the evolution of resistance; drug-drug interactions such as this, with varied parasite strains and drug tolerance will therefore be imperative to test the different drug candidates prior to clinical trials.

2.5 Conclusions

Although TFQ is in late stage clinical trials for P. vivax, it appears to synergize ACTs against P. falciparum in vitro; this might be beneficial in mixed infections of world regions where

P. falciparum and P. vivax are co-endemic. Here we show that drug interactions between asexual and sexual stage parasites differ, prompting future drug interaction studies to include gametocytes instead of just the asexual stage as has been the norm. The flow cytometry based method used here, is one reproducible way to do this in vitro, although different methods may have to be applied in field isolates.

Cmax values vary from drug to drug, so fixed ratios for isobologram analysis should be chosen carefully to reflect in vivo pharmacokinetics, whilst maintaining in vitro inhibition efficacy. In vivo TFQ-ACT/CQ drug interaction work in humanized mice of varying G6PD activity as well as human clinical trials are a necessary follow-up to results reported here since host factors such as gender, immunity, drug metabolism as well as diet, among others, might influence the results obtained.

44 References

1. Eastman RT, Fidock D a: Artemisinin-based combination therapies: a vital tool in efforts to eliminate malaria. Nat Rev Microbiol 2009, 7:864–874.

2. White NJ: Primaquine to prevent transmission of falciparum malaria. Lancet Infect Dis 2013, 13:181.

3. WHO: World Malaria Report 2014. Volume WHO/HTM/GM; 2014.

4. Crockett M, Kain KC: Tafenoquine: a promising new antimalarial agent. Expert Opin Investig Drugs 2007, 16:705–715.

5. Li Q, O’Neil M, Xie L, Caridha D, Zeng Q, Zhang J, Pybus B, Hickman M, Melendez V: Assessment of the prophylactic activity and pharmacokinetic profile of oral tafenoquine compared to primaquine for inhibition of liver stage malaria infections. Malar J 2014, 13:141.

6. Shanks GD, Kain KC, Keystone JS: Malaria chemoprophylaxis in the age of drug resistance. II. Drugs that may be available in the future. Clin Infect Dis 2001, 33:381–385.

7. Llanos-Cuentas A, Lacerda M V., Rueangweerayut R, Krudsood S, Gupta SK, Kochar SK, Arthur P, Chuenchom N, Möhrle JJ, Duparc S, Ugwuegbulam C, Kleim JP, Carter N, Green J a., Kellam L: Tafenoquine plus chloroquine for the treatment and relapse prevention of Plasmodium vivax malaria (DETECTIVE): A multicentre, double-blind, randomised, phase 2b dose-selection study. Lancet 2014, 383:1049–1058.

8. Carvalho L, Martínez-García M, Pérez-Victoria I, Manzano JI, Yardley V, Gamarro F, Pérez-Victoria JM: The Oral Antimalarial Drug Tafenoquine Shows Activity against Trypanosoma brucei. Antimicrob Agents Chemother 2015, 59:6151–6160.

9. Carvalho L, Luque-Ortega JR, Manzano JI, Castanys S, Rivas L, Gamarro F: Tafenoquine, an antiplasmodial 8-aminoquinoline, targets Leishmania respiratory complex III and induces apoptosis. Antimicrob Agents Chemother 2010, 54:5344–5351.

10. Baird JK, Dewi M, Subekti D, Elyazar I, Satyagraha AW: Noninferiority of glucose- 6-phosphate dehydrogenase deficiency diagnosis by a point-of-care rapid test vs the laboratory fluorescent spot test demonstrated by copper inhibition in normal human red blood cells. Transl Res 2015, 165:677–688.

11. Roobsoong W, Tharinjaroen CS, Rachaphaew N, Chobson P, Schofield L, Cui L, Adams JH, Sattabongkot J: Improvement of culture conditions for long-term in vitro culture of Plasmodium vivax. Malar J 2015, 14:297.

12. Udomsangpetch R, Somsri S, Panichakul T, Chotivanich K, Sirichaisinthop J, Yang Z, Cui L, Sattabongkot J: Short-term in vitro culture of field isolates of Plasmodium vivax using umbilical cord blood. Parasitol Int 2007, 56:65–9.

45 13. Gorka AP, Jacobs LM, Roepe PD: Cytostatic versus cytocidal profiling of quinoline drug combinations via modified fixed-ratio isobologram analysis. Malar J 2013, 12:332.

14. Bray PG, Deed S, Fox E, Kalkanidis M, Mungthin M, Deady LW, Tilley L: Primaquine synergises the activity of chloroquine against chloroquine-resistant P. falciparum. Biochem Pharmacol 2005, 70:1158–1166.

15. Ramharter M, Noedl H, Thimasarn K, Wiedermann G, Wernsdorfer G, Wernsdorfer WH: In vitro activity of tafenoquine alone and in combination with artemisinin against Plasmodium falciparum. Am J Trop Med Hyg 2002, 67:39–43.

16. Cabrera M, Cui L: In Vitro Activities of Primaquine-Schizonticide Combinations on Asexual Blood Stages and Gametocytes of Plasmodium falciparum. Antimicrob Agents Chemother 2015(September):AAC.01948–15.

17. Ali S, Najmi MH, Tarning J, Lindegardh N: Pharmacokinetics of artemether and dihydroartemisinin in healthy Pakistani male volunteers treated with artemether- lumefantrine. Malar J 2010, 9:275.

18. Morris C a, Duparc S, Borghini-Fuhrer I, Jung D, Shin C-S, Fleckenstein L: Review of the clinical pharmacokinetics of artesunate and its active metabolite dihydroartemisinin following intravenous, intramuscular, oral or rectal administration. Malar J 2011, 10:263.

19. Green J a., Patel AK, Patel BR, Hussaini A, Harrell EJ, McDonald MJ, Carter N, Mohamed K, Duparc S, Miller AK: Tafenoquine at therapeutic concentrations does not prolong fridericia-corrected QT interval in healthy subjects. J Clin Pharmacol 2014, 54:995– 1005.

20. Benjamin J, Moore B, Lee ST, Senn M, Griffin S, Lautu D, Salman S, Siba P, Mueller I, Davis TME: Artemisinin-Naphthoquine Combination Therapy for Uncomplicated Pediatric Malaria: a Tolerability, Safety, and Preliminary Efficacy Study. Antimicrob Agents Chemother 2012, 56:2465–2471.

21. Wang Z, Wang Y, Cabrera M, Zhang Y, Gupta B, Wu Y, Kemirembe K, Hu Y: Artemisinin Resistance at the China-Myanmar Border and Association with Mutations in the K13 Propeller Gene. 2015, 59:6952–6959.

22. Varios: Milestones In Drug Therapy: Aromatase Inhibitors. 2008.

23. Muangnoicharoen S, Johnson DJ, Looareesuwan S, Krudsood S, Ward SA: Role of known molecular markers of resistance in the antimalarial potency of piperaquine and dihydroartemisinin in vitro. Antimicrob Agents Chemother 2009, 53:1362–6.

24. Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois A-C, Khim N, Kim S, Duru V, Bouchier C, Ma L, Lim P, Leang R, Duong S, Sreng S, Suon S, Chuor CM, Bout DM, Ménard S, Rogers WO, Genton B, Fandeur T, Miotto O, Ringwald P, Le Bras J, Berry A, Barale

46 J-C, Fairhurst RM, Benoit-Vical F, Mercereau-Puijalon O, Ménard D: A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature 2014, 505:50–5.

25. Trager W, Jensen JB: Human malaria parasites in continuous culture. Science 1976, 193:673–675.

26. Fivelman QL, McRobert L, Sharp S, Taylor CJ, Saeed M, Swales CA, Sutherland CJ, Baker DA: Improved synchronous production of Plasmodium falciparum gametocytes in vitro. Mol Biochem Parasitol 2007, 154:119–123.

27. Miao J, Wang Z, Liu M, Parker D, Li X, Chen X, Cui L: Plasmodium falciparum: generation of pure gametocyte culture by heparin treatment. Exp Parasitol 2013, 135:541–5.

28. Lambros C, Vanderberg JP: Synchronization of Plasmodium falciparum erythrocytic stages in culture. J Parasitol 1979, 65:418–420.

29. Smilkstein M, Sriwilaijaroen N, Kelly JX, Wilairat P, Riscoe M: Simple and inexpensive fluorescence-based technique for high-throughput antimalarial drug screening. Antimicrob Agents Chemother 2004, 48:1803–6.

30. Hawley SR, Bray PG, Mungthin M, Atkinson JD, O’Neill PM, Ward S a.: Relationship between antimalarial drug activity, accumulation, and inhibition of heme polymerization in Plasmodium falciparum in vitro. Antimicrob Agents Chemother 1998, 42:682–686.

31. Rijken MJ, McGready R, Jullien V, Tarning J, Lindegardh N, Phyo AP, Win AK, Hsi P, Cammas M, Singhasivanon P, White NJ, Nosten F: Pharmacokinetics of amodiaquine and desethylamodiaquine in pregnant and postpartum women with Plasmodium vivax malaria. Antimicrob Agents Chemother 2011, 55:4338–42.

32. Qu HY, Gao HZ, Hao GT, Li YY, Li HY, Hu JC, Wang XF, Liu WL, Liu ZY: Single-dose safety, pharmacokinetics, and food effects studies of compound naphthoquine phosphate tablets in healthy volunteers. J Clin Pharmacol 2010, 50:1310–8.

33. Batty KT, Salman S, Moore BR, Benjamin J, Lee ST, Page-Sharp M, Pitus N, Ilett KF, Mueller I, Hombhanje FW, Siba P, Davis TME: Artemisinin-naphthoquine combination therapy for uncomplicated pediatric malaria: a pharmacokinetic study. Antimicrob Agents Chemother 2012, 56:2472–84.

34. Djimdé AA, Tekete M, Abdulla S, Lyimo J, Bassat Q, Mandomando I, Lefèvre G, Borrmann S: Pharmacokinetic and pharmacodynamic characteristics of a new pediatric formulation of artemether-lumefantrine in African children with uncomplicated Plasmodium falciparum malaria. Antimicrob Agents Chemother 2011, 55:3994–9.

35. Lefèvre G, Bhad P, Jain JP, Kalluri S, Cheng Y, Dave H, Stein DS: Evaluation of two novel tablet formulations of artemether-lumefantrine (Coartem) for bioequivalence in a randomized, open-label, two-period study. Malar J 2013, 12:312.

47 36. Price R, Simpson J a., Teja-Isavatharm P, Than MM, Luxemburger C, Heppner DG, Chongsuphajaisiddhi T, Nosten F, White NJ: Pharmacokinetics of mefloquine combined with artesunate in children with acute falciparum malaria. Antimicrob Agents Chemother 1999, 43:341–346.

37. Gutman J, Green M, Durand S, Rojas OV, Ganguly B, Quezada WM, Utz GC, Slutsker L, Ruebush TK, Bacon DJ: Mefloquine pharmacokinetics and mefloquine-artesunate effectiveness in Peruvian patients with uncomplicated Plasmodium falciparum malaria. Malar J 2009, 8:58.

38. Croft SL, Duparc S, Arbe-Barnes SJ, Craft JC, Shin C-S, Fleckenstein L, Borghini- Fuhrer I, Rim H-J: Review of pyronaridine anti-malarial properties and product characteristics. Malar J 2012, 11:270.

39. Nguyen TC, Nguyen NQ, Nguyen XT, Bui D, Travers T, Edstein MD: Pharmacokinetics of the antimalarial drug piperaquine in healthy Vietnamese subjects. Am J Trop Med Hyg 2008, 79:620–3.

40. Karunajeewa H a., Salman S, Mueller I, Baiwog F, Gomorrai S, Law I, Page-Sharp M, Rogerson S, Siba P, Ilett KF, Davis TME: Pharmacokinetics of chloroquine and monodesethylchloroquine in pregnancy. Antimicrob Agents Chemother 2010, 54:1186–1192.

41. Gbotosho GO, Happi CT, Sijuade a, Ogundahunsi O a T, Sowunmi a, Oduola a MJ: Comparative study of interactions between chloroquine and chlorpheniramine or promethazine in healthy volunteers: a potential combination-therapy phenomenon for resuscitating chloroquine for malaria treatment in Africa. Ann Trop Med Parasitol 2008, 102:3–9.

42. Cook J a, Randinitis EJ, Bramson CR, Wesche DL: Lack of a pharmacokinetic interaction between azithromycin and chloroquine. Am J Trop Med Hyg 2006, 74:407–12.

43. Carter R, Miller LH: Evidence for environmental modulation of gametocytogenesis in Plasmodium falciparum in continuous culture. Bull World Health Organ 1979, 57(Suppl. 1):37–52.

44. Reader J, Botha M, Theron A, Lauterbach SB, Rossouw C, Engelbrecht D, Wepener M, Smit A, Leroy D, Mancama D, Coetzer TL, Birkholtz L-M: Nowhere to hide: interrogating different metabolic parameters of Plasmodium falciparum gametocytes in a transmission blocking drug discovery pipeline towards malaria elimination. Malar J 2015, 14:213.

45. Price RN, von Seidlein L, Valecha N, Nosten F, Baird JK, White NJ: Global extent of chloroquine-resistant Plasmodium vivax: a systematic review and meta-analysis. Lancet Infect Dis 2014, 14:982–91.

46. Smithuis F, Kyaw MK, Phe O, Win T, Aung PP, Oo APP, Naing AL, Nyo MY, Myint NZH, Imwong M, Ashley E, Lee SJ, White NJ: Effectiveness of five artemisinin

48 combination regimens with or without primaquine in uncomplicated falciparum malaria: An open-label randomised trial. Lancet Infect Dis 2010, 10:673–681.

47. Snounou G, White NJ: The co-existence of Plasmodium: Sidelights from falciparum and vivax malaria in Thailand. Trends Parasitol 2004, 20:333–339.

48. Bell A: Antimalarial drug synergism and antagonism: mechanistic and clinical significance. FEMS Microbiol Lett 2005, 253:171–84.

49. Vennerstrom JL, Nuzum EO, Miller RE, Dorn A, Gerena L, Dande P a., Ellis WY, Ridley RG, Milhous WK: 8-Aminoquinolines active against blood stage Plasmodium falciparum in vitro inhibit hematin polymerization. Antimicrob Agents Chemother 1999, 43:598–602.

50. Wang JY, Cao WC, Shan CQ, Zhang M, Li GF, Ding D Ben, Shi YL, Wu BA: Naphthoquine phosphate and its combination with artemisinine. Acta Trop 2004, 89:375– 381.

51. Duffy S, Avery VM: Identification of inhibitors of Plasmodium falciparum gametocyte development. Malar J 2013, 12:408.

52. Peatey CL, Leroy D, Gardiner DL, Trenholme KR: Anti-malarial drugs: how effective are they against Plasmodium falciparum gametocytes? Malar J 2012, 11:34.

53. Adjalley SH, Johnston GL, Li T, Eastman RT, Ekland EH, Eappen a. G, Richman a., Sim BKL, Lee MCS, Hoffman SL, Fidock D a.: PNAS Plus: Quantitative assessment of Plasmodium falciparum sexual development reveals potent transmission-blocking activity by methylene blue. Proc Natl Acad Sci 2011, 108:E1214–E1223.

54. Lucantoni L, Duffy S, Adjalley SH, Fidock D a., Avery VM: Identification of MMV malaria box inhibitors of Plasmodium falciparum early-stage gametocytes using a luciferase-based high-throughput assay. Antimicrob Agents Chemother 2013, 57:6050–6062.

55. Ponsa N, Sattabongkot J, Kittayapong P, Eikarat N, Coleman RE: Transmission- blocking activity of tafenoquine (WR-238605) and artelinic acid against naturally circulating strains of Plasmodium vivax in Thailand. Am J Trop Med Hyg 2003, 69:542–7.

56. Coleman RE, Clavin a M, Milhous WK: Gametocytocidal and sporontocidal activity of antimalarials against Plasmodium berghei ANKA in ICR Mice and Anopheles stephensi mosquitoes. Am J Trop Med Hyg 1992, 46:169–82.

57. White NJ, Ashley E a, Recht J, Delves MJ, Ruecker a, Smithuis FM, Eziefula a C, Bousema T, Drakeley C, Chotivanich K, Imwong M, Pukrittayakamee S, Prachumsri J, Chu C, Andolina C, Bancone G, Hien TT, Mayxay M, Taylor WR, von Seidlein L, Price RN, Barnes KI, Djimde a, Ter Kuile F, Gosling R, Chen I, Dhorda MJ, Stepniewska K, Guerin P, Woodrow CJ, et al.: Assessment of therapeutic responses to gametocytocidal drugs in Plasmodium falciparum malaria. Malar J 2014, 13:483.

49 58. Ruecker a, Mathias DK, Straschil U, Churcher TS, Dinglasan RR, Leroy D, Sinden RE, Delves MJ: A male and female gametocyte functional viability assay to identify biologically relevant malaria transmission-blocking drugs. Antimicrob Agents Chemother 2014, 58:7292–302.

59. Delves MJ, Ruecker A, Straschil U, Lelièvre J, Marques S, López-Barragán MJ, Herreros E, Sinden RE: Male and female Plasmodium falciparum mature gametocytes show different responses to antimalarial drugs. Antimicrob Agents Chemother 2013, 57:3268–3274.

60. Mariga ST, Gil JP, Sisowath C, Wernsdorfer WH, Bjorkman A: Synergism between amodiaquine and its major metabolite, desethylamodiaquine, against Plasmodium falciparum in vitro. Antimicrob Agents Chemother 2004, 48:4089–4096.

61. Buckling a, Ranford-Cartwright LC, Miles a, Read a F: Chloroquine increases Plasmodium falciparum gametocytogenesis in vitro. Parasitology 1999, 118 ( Pt 4:339–346.

62. Price R., Nosten F, Luxemburger C, ter Kuile F., Paiphun L, Chongsuphajaisiddhi T, White N.: Effects of artemisinin derivatives on malaria transmissibility. Lancet 1996, 347:1654–1658.

63. Price R, Nosten F, Simpson J a., Luxemburger C, Phaipun L, Kuile F Ter, Van Vugt M, Chongsuphajaisiddhi T, White NJ: Risk factors for gametocyte carriage in uncomplicated falciparum malaria. Am J Trop Med Hyg 1999, 60:1019–1023.

64. Vuong C, Xie LH, Potter BMJ, Zhang J, Zhang P, Duan D, Nolan CK, Sciotti RJ, Zottig VE, Dhammika Nanayakkara N., Tekwani BL, Walker L a., Smith PL, Paris RM, Read LT, Li Q, Pybus BS, Sousa JC, Reichard G a., Smith B, Marcsisin SR: Differential CYP 2D Metabolism Alters Tafenoquine Pharmacokinetics. Antimicrob Agents Chemother 2015(April):AAC.00343–15.

65. Edstein MD, Nasveld PE, Kocisko D a., Kitchener SJ, Gatton ML, Rieckmann KH: Gender differences in gastrointestinal disturbances and plasma concentrations of tafenoquine in healthy volunteers after tafenoquine administration for post-exposure vivax malaria prophylaxis. Trans R Soc Trop Med Hyg 2007, 101:226–230.

66. Brueckner RP, Coster T, Wesche DL, Shmuklarsky M, Schuster BG: Prophylaxis of Plasmodium falciparum infection in a human challenge model with WR 238605, a new 8- aminoquinoline antimalarial. Antimicrob Agents Chemother 1998, 42:1293–4.

67. Schneider P, Wolters L, Schoone G, Schallig H, Sillekens P, Hermsen R, Sauerwein R: Real-time nucleic acid sequence-based amplification is more convenient than real-time PCR for quantification of Plasmodium falciparum. J Clin Microbiol 2005, 43:402–5.

68. Rochford R, Ohrt C, Baresel PC, Campo B, Sampath A, Magill AJ, Tekwani BL, Walker L a: Humanized mouse model of glucose 6-phosphate dehydrogenase deficiency for in vivo assessment of hemolytic toxicity. Proc Natl Acad Sci U S A 2013, 110:17486–91.

50 Chapter 3

Conclusions and future directions

3.1 Summary of findings and relevance

This thesis investigates drug-drug interactions between tafenoquine (TFQ), an 8- aminoquinoline drug in late clinical drug development for P. vivax and six long-lasting ACT- partner drugs in Plasmodium falciparum asexual and sexual stage parasites. TFQ is being developed for the radical cure of P. vivax following a 3-day course of CQ, but an ongoing shift from CQ to ACT antimalarials prompted the assessment of ACT-TFQ interactions (Llanos-

Cuentas et al., 2013). Based on results obtained showing mostly a synergistic effect, at least in the asexual stages, it appears that TFQ dosage for P. vivax malaria following either a three-day drug regimen of CQ or ACT may have potential off-target benefits in the treatment of P. falciparum for individuals with a mixed infection. Care must be taken, if TFQ is deployed in the near future for P. vivax that the ACT of choice used does not have an antagonistic effect on either P. falciparum or P. vivax gametocyte carriage, as that might hinder efforts to control transmission to mosquitoes.

In vivo studies in model organisms or in humans in malaria-endemic areas will have to be undertaken to verify drug-drug interactions since host factors such as immunity or drug metabolism might come into play. A study to investigate the drug-drug interactions of TFQ treatment following treatment with two ACTs, dihydroartemisinin- piperaquine and artemether- lumefantrine has been completed, but is yet to be published; this should yield some in vivo data on beneficial/adverse effects of TFQ when combined with ACT, rather than the still widely used

CQ for P. vivax due to CQ resistance in P. vivax not being as widespread as it is in P. falciparum

(ClinicalTrials.gov identification: NCT02184637; Green et al., 2015: ASTMH abstract 1209). In

51 addition, GlaxoSmithKline, the pharmaceutical company developing TFQ, is carrying out a clinical trial to compare PMQ and TFQ toxicity in people with and without a G6PD deficiency since both drugs cause severe hemolysis in these people, and TFQ having a longer elimination half-life than PMQ, might cause additional complications (ClinicalTrials.gov identifier:

NCT01376167). A hemolytic effect equal to or lower than that of PMQ is desirable.

To my knowledge, this is the first study in vitro that tests the drug interactions between

TFQ and ACT-partner drugs using both sexual and asexual stage parasites at ratios reflective of the pharmacokinetics of each individual drug in the human body. Most studies use arbitrary ratios of 1:1, 1:3 and 3: 1 based on in vitro IC50s, but these may not necessarily reflect drug peak concentrations in vivo, so the conclusions cannot easily be translated to the clinic (Gorka, et al.,

2013; Akoachere et al., 2005).

Lastly, it is my hope that with the rise of drug-drug interaction studies in coming years for antimalarial combination drugs, of which there are many in clinical trials, drug interaction assays will become more standardized to better reflect in vivo drug exposure, since as shown here, the experimental conditions as well as drug concentrations used can give differing results, and that more stringent definitions will be developed to define synergy and antagonism, because these vary, with some papers citing that synergy is any drug combination of <1, and others using

0.5 as a cut- off point. Antagonism has been reported at thresholds ranging from an FIC index of above 1 to above 4; suggesting that anyone can use whichever guideline they would like to avoid their drug combinations of interest being classified as antagonistic (Fivelman et al., 2004; Bell,

2005; Wells et al., 2015).

52 3.2 Perspectives on malaria control

Recently published work on the 8-aminoquinoline PMQ, used for both gametocytocidal purposes in P. falciparum and for elimination of the liver stage in P. vivax, has shown that when taken concomitantly with ACT partner drugs, the interactions are predominantly additive or synergistic, except in the case of lumefantrine (LMF) at certain concentrations (Cabrera and Cui,

2015). This is great news considering that the PMQ used in vitro is less effective compared to its metabolite, carboxyprimaquine, in vivo, suggesting that the combinations are likely even more synergistic in vivo (Ganesan et al., 2009). This study comes in 9 years after ACTs were officially recommended by the WHO as the first line treatment for malaria, and PMQ dosage being recommended in addition to either CQ or ACT antimalarials in areas of emerging ART resistance to suppress transmission of infectious gametocytes from humans to mosquito vectors and to kill dormant liver parasites in P. vivax, ovale and malariae (Wells et al., 2015; WHO 2014).

Considering how long these drugs have been co-administered, it is astonishing that only a handful of studies have studied in vitro drug-drug interactions to currently used antimalarials, moreover most have been performed with only asexual and not sexual stage parasites because if the goal is to increase efficacy of gametocyte killing, then a suitable synergistic combination of drugs is desired (Gorka et al., 2013, Bray et al., 2005). Also, with the anticipated evolution of resistance to every monotherapy drug developed, fixed-dose combination drugs like ACTs are likely to remain the preferred drug regimens as they are presumed to lower the chances of resistance developing rapidly (White, 2004). Therefore, in vitro and in vivo drug-drug combination studies, in my opinion, will likely become even more routine because they will help guide the process of choosing which drugs to co-formulate.

Malaria deaths have certainly decreased in the last decade, owing to the effectiveness of

ACT, and increased funding for vector control through insecticide treated nets, site-specific

53 elimination through mass drug administration, and more research funding on basic research of the malaria parasite and the mosquito vector (WHO, 2014). In 2013 alone, about $2.7 billion was spent worldwide on malaria control efforts. There has also been significant progress towards the development of a protective vaccine for malaria, although currently the front-runner RTS,S vaccine has only limited protective efficacy of no greater than 55% and only for a few months

(Wells et al., 2015). Antimalarial chemotherapy, therefore still plays a large role in the control of the spread of malaria.

It is known that gametocytes are the sole link between mosquito and human malaria transmission, so a focus on developing drugs that target both asexual parasites that cause malaria symptoms and sexual stage parasites that facilitate mosquito transmission is key. A few candidates in the pipeline include a previously used chemotherapeutic dye, and two new classes of drugs, the spiroindolones and imidazolopiperazines etc to mention but a few (Leong et al.,

2014; van Pelt-Koops et al., 2012; Coulibaly et al., 2015). Currently, the 8-aminoquinoline, PMQ is the only transmission suppressing drug approved for human use on the market, although bulaquine/ elubaquine, an 8-aminoquinoline that is metabolized to PMQ is being used in India for transmission blocking, as well as for the prevention of P. vivax relapses of malaria but has yet to be approved by the Stringent Regulatory Authority of the European Medicine s Agency for broader usage (White, 2013; Krudsood et al., 2006).

Currently, the problem with PMQ and TFQ is that they cause severe hemolysis in people with a G6PD deficiency. The classic test for G6PD has been the fluorescent spot test but it has a few shortcomings, namely that it requires a UV light, cannot accurately measure G6PD activity in heterozygous females because it is an X chromosome recessive condition, is dependent on temperature and requires a series of cold chain experiments. (Beutler, 2008; Baird et al., 2015;

WHO, 2015). The recently developed CareSTART (AccessBio Inc) has the potential to improve

G6PD screening because it is cheap, fast and is not dependent on temperature, and has been used

54 successfully in the field with 96% sensitivity (von Fricken et al., 2014; Adu-Gyasi et al., 2015)

The use of humanized mice will also aid in the initial investigation of effects of experimental drugs in vivo, before experiments are translated to monkeys/ humans (Rochford et al., 2013).

Another complication with transmission blocking drugs is the differential effects in male and female gametocytes, as some studies have suggested that females are more tolerant to many antimalarial drugs and are likely to persist in circulation following treatment (White et al., 2014).

Several assays have been developed to investigate sex differences to antimalarial therapy, but these are conflicting, with some drugs showing gametocytocidal activity in one study, but not in others (Delves et al., 2013; Adjalley et al., 2011; Reader et al., 2015; Ruecker et al., 2014). One difficulty is that so far there is no reported female specific marker that could aid in drug screening efforts targeting female gametocytes by flow cytometry (Schwank et al., 2010).

Taken together, malaria control is continuing to make steady progress in the right direction, but for this trend to persist there is a need for continuous funding, as well as co- operation between academics and pharmaceutical companies (Wells et al., 2015). The fact that a lot of pharmaceutical companies developing antimalarials are sharing their data publicly, and are willing to perform this research despite a lack of a foreseeable profit since their target market is in mostly low-income countries, is commendable. Of course the downstream benefits will be felt by everybody when economies of malaria-inflicted countries perform better and require less aid from the developed world, and when expatriates, exchange students, tourists and soldiers can safely travel to malaria-endemic countries. Here at The Pennsylvania State University, there is noteworthy research concerning the use of viruses, bacteria, transgenic mosquitoes and fungi for mosquito control and suppression of mosquito stage parasite growth, all of which will be valuable additions to the multi-target effort to develop safe, resistance-proof and relatively cheap methods in the fight against malaria (Heinig et al., 2015; Suzuki et al., 2015; Murdock et al., 2014).

55 3.3 References

Adjalley, Sophie H., Geoffrey L. Johnston, Tao Li, Richard T. Eastman, Eric H. Ekland,

Abraham G. Eappen, Adam Richman et al. "Quantitative assessment of Plasmodium falciparum sexual development reveals potent transmission-blocking activity by methylene blue." Proceedings of the National Academy of Sciences 108, no. 47 (2011): E1214-E1223.

Adu-Gyasi, Dennis, Kwaku Poku Asante, Sam Newton, David Dosoo, Sabastina

Amoako, George Adjei, Nicholas Amoako et al. "Evaluation of the Diagnostic Accuracy of

CareStart G6PD Deficiency Rapid Diagnostic Test (RDT) in a Malaria Endemic Area in Ghana,

Africa." (2015): e0125796.

Akoachere, Monique, Kathrin Buchholz, Elisabeth Fischer, Jürgen Burhenne, Walter E.

Haefeli, R. Heiner Schirmer, and Katja Becker. "In vitro assessment of methylene blue on chloroquine-sensitive and-resistant Plasmodium falciparum strains reveals synergistic action with ." Antimicrobial agents and chemotherapy 49, no. 11 (2005): 4592-4597.

Baird, J. Kevin, Mewahyu Dewi, Decy Subekti, Iqbal Elyazar, and Ari W. Satyagraha.

"Noninferiority of glucose-6-phosphate dehydrogenase deficiency diagnosis by a point-of-care rapid test vs the laboratory fluorescent spot test demonstrated by copper inhibition in normal human red blood cells." Translational Research 165, no. 6 (2015): 677-688.

Bell, Angus. "Antimalarial drug synergism and antagonism: mechanistic and clinical significance." FEMS microbiology letters 253, no. 2 (2005): 171-184.

Beutler, Ernest. "Glucose-6-phosphate dehydrogenase deficiency: a historical perspective." Blood 111, no. 1 (2008): 16-24.

Bray, Patrick G., Samantha Deed, Emma Fox, Martha Kalkanidis, Mathirut Mungthin,

Leslie W. Deady, and Leann Tilley. "Primaquine synergises the activity of chloroquine against chloroquine-resistant P. falciparum." Biochemical pharmacology 70, no. 8 (2005): 1158-1166.

56 Cabrera, Mynthia, and Liwang Cui. "In Vitro Activities of Primaquine-Schizonticide

Combinations on Asexual Blood Stages and Gametocytes of Plasmodium falciparum." Antimicrobial agents and chemotherapy (2015): AAC-01948.

Coulibaly, Boubacar, Michael Pritsch, Mamadou Bountogo, Peter E. Meissner, Eric

Nebié, Christina Klose, Meinhard Kieser et al. "Efficacy and safety of triple combination therapy with artesunate-amodiaquine-methylene blue for falciparum malaria in children: a randomised controlled trial in Burkina Faso." Journal of Infectious Diseases (2014): jiu540.

Gorka, Alexander P., Lauren M. Jacobs, and Paul D. Roepe. "Cytostatic versus cytocidal profiling of quinoline drug combinations via modified fixed-ratio isobologram analysis." Malaria

Journal 12 (2013): 332.

Fivelman, Quinton L., Ipemida S. Adagu, and David C. Warhurst. "Modified fixed-ratio isobologram method for studying in vitro interactions between atovaquone and proguanil or dihydroartemisinin against drug-resistant strains of Plasmodium falciparum." Antimicrobial agents and chemotherapy 48, no. 11 (2004): 4097-4102.

Ganesan, Shobana, Babu L. Tekwani, Rajnish Sahu, Lalit M. Tripathi, and Larry A.

Walker. "Cytochrome P 450-dependent toxic effects of primaquine on human erythrocytes." Toxicology and applied pharmacology 241, no. 1 (2009): 14-22.

Glaxosmithkline. “A multi-centre, double-blind, randomised, parallel-group, active controlled study to evaluate the efficacy, safety and tolerability of tafenoquine (SB-252263,

WR238605) in subjects with Plasmodium vivax malaria”. Study ID: 112582. < http://www.gsk- clinicalstudyregister.com/study/112582#ps?> Accessed Oct 27th, 2015.

Green, J.A et al., “Drug-drug interaction study of tafenoquine and the ACTs dihydroartemisinin-piperaquine (DHA-PQP) and artemether-lumefantrine (AL)”. ASTMH abstract 1209.< http://www.abstractsonline.com/Plan/ViewAbstract.aspx?sKey=6aca7ab3-b3a0-

4466-9a91-5bc8dfdc8ff0&cKey=f0ea8b4f-c1a5-46ae-8892-

57 2b5101eac6bf&mKey=%7bAB652FDF-0111-45C7-A5E5-0BA9D4AF5E12%7d> Accessed Oct

28th, 2015.

Heinig, Rebecca L., Krijn P. Paaijmans, Penelope A. Hancock, and Matthew B. Thomas.

"The potential for fungal biopesticides to reduce malaria transmission under diverse environmental conditions." Journal of Applied Ecology (2015).

Krudsood, Srivicha, Polrat Wilairatana, Noppadon Tangpukdee, Kobsiri Chalermrut,

Siripun Srivilairit, Vipa Thanachartwet, Sant Muangnoicharoen, Natthanej Luplertlop, Gary M.

Brittenham, and Sornchai Looareesuwan. "Safety and tolerability of elubaquine (bulaquine, CDRI

80/53) for treatment of Plasmodium vivax malaria in Thailand." The Korean journal of parasitology 44, no. 3 (2006): 221-228.

Leong, Joel, Ruobing Li, Jay Prakash Jain, Gilbert Lefèvre, Baldur Magnusson, Thierry

Diagana, and Peter Pertel. "A first-in-human randomized, double-blind, placebo-controlled, single-and multiple-ascending oral dose study of novel spiroindolone KAE609, to assess the safety, tolerability and pharmacokinetics in healthy adult volunteers." Malaria Journal 13, no.

Suppl 1 (2014): O37.

Llanos-Cuentas, Alejandro, Marcus V. Lacerda, Ronnatrai Rueangweerayut, Srivicha

Krudsood, Sandeep K. Gupta, Sanjay K. Kochar, Preetam Arthur et al. "Tafenoquine plus chloroquine for the treatment and relapse prevention of Plasmodium vivax malaria

(DETECTIVE): a multicentre, double-blind, randomised, phase 2b dose-selection study." The

Lancet 383, no. 9922 (2014): 1049-1058.

Murdock, Courtney C., Simon Blanford, Grant L. Hughes, Jason L. Rasgon, and Matthew

B. Thomas. "Temperature alters Plasmodium blocking by Wolbachia." Scientific reports 4

(2014).

van Pelt-Koops, J. C., H. E. Pett, W. Graumans, M. van der Vegte-Bolmer, G. J. van

Gemert, M. Rottmann, B. K. S. Yeung, T. T. Diagana, and R. W. Sauerwein. "The spiroindolone

58 drug candidate NITD609 potently inhibits gametocytogenesis and blocks Plasmodium falciparum transmission to anopheles mosquito vector." Antimicrobial agents and chemotherapy 56, no. 7

(2012): 3544-3548.

Reader, Janette, Mariëtte Botha, Anjo Theron, Sonja B. Lauterbach, Claire Rossouw,

Dewaldt Engelbrecht, Melanie Wepener et al. "Nowhere to hide: interrogating different metabolic parameters of Plasmodium falciparum gametocytes in a transmission blocking drug discovery pipeline towards malaria elimination." Malaria Journal 14, no. 1 (2015): 213.

Rochford, Rosemary, Colin Ohrt, Paul C. Baresel, Brice Campo, Aruna Sampath, Alan J.

Magill, Babu L. Tekwani, and Larry A. Walker. "Humanized mouse model of glucose 6- phosphate dehydrogenase deficiency for in vivo assessment of hemolytic toxicity." Proceedings of the National Academy of Sciences 110, no. 43 (2013): 17486-17491.

Ruecker, A., D. K. Mathias, U. Straschil, T. S. Churcher, R. R. Dinglasan, D. Leroy, R.

E. Sinden, and M. J. Delves. "A male and female gametocyte functional viability assay to identify biologically relevant malaria transmission-blocking drugs." Antimicrobial agents and chemotherapy 58, no. 12 (2014): 7292-7302.

Schwank, Samana, Colin J. Sutherland, and Chris J. Drakeley. "Promiscuous expression of α-tubulin II in maturing male and female Plasmodium falciparum gametocytes." PloS one

(2010): e14470.

Suzuki, Yasutsugu, Tapan K. Barik, Rebecca M. Johnson, and Jason L. Rasgon. "In vitro and in vivo host range of Anopheles gambiae densovirus (AgDNV)." Scientific reports 5 (2015).

von Fricken, Michael E., Thomas A. Weppelmann, Will T. Eaton, Roseline Masse,

Madsen VE Beau de Rochars, and Bernard A. Okech. "Performance of the CareStart glucose-6- phosphate dehydrogenase (G6PD) rapid diagnostic test in Gressier, Haiti." The American journal of tropical medicine and hygiene 91, no. 1 (2014): 77-80.

59 Wells, Timothy NC, Rob Hooft van Huijsduijnen, and Wesley C. Van Voorhis. "Malaria medicines: a glass half full?." Nature Reviews Drug Discovery (2015).

White, Nicholas J., Elizabeth A. Ashley, Judith Recht, Michael J. Delves, Andrea

Ruecker, Frank M. Smithuis, Alice C. Eziefula et al. "Assessment of therapeutic responses to gametocytocidal drugs in Plasmodium falciparum malaria." Malaria Journal 13, no. 1 (2014):

483.

World Health Organization. "World Malaria Report 2014 (2014)." WHO: Geneva (2014).

World Health Organization. "Control and elimination of plasmodium vivax malaria: a technical brief." (2015).

60 Appendix A

A1: Gametocyte induction

A1.1 Equipment and Materials

- Heraeus Megafuge 16R Centrifuge (Thermo Sci)

- Eclipse 50i Microscope 100x (Nikon)

- Humidified 5% CO2 incubator

- Blue vented cap culture flasks (Corning)

A1.2 Reagents

- Parasite thawing solutions

Purpose: Recovery of previously cryopreserved parasites in liquid nitrogen (-196°C).

12% NaCl in distilled water

1.8% NaCl in distilled water

0.9% NaCl/ 0.2% Glucose in distilled water

Critical steps: All three solutions should be filter sterilized by passing through a 0.2µm membrane and pre-warmed at 37°C before use.

- Heparin sodium salt from porcine intestinal mucosa (Sigma)

Purpose: To inhibit merozoite invasion hence eliminating asexual stage proliferation in gametocyte culture (Miao et al., 2013).

Critical steps: Dissolve 50mg/ml in distilled water and filter sterilize using 0.2µm membrane and syringe.

61 - Percoll (Sigma)

Purpose: For high density centrifugation to separate blood, asexual stage and sexual stage parasites.

Critical steps: Make up to 90% with incomplete medium (pH 7.4) and filter sterilize, store at 4°C.

Pre- warm to 37°C before gametocyte culture centrifugation.

- Malaria Complete Medium (MCM). RPMI 1640 + Albumax II (Lipid rich bovine serum).

Purpose: Provide necessary nutrients for read blood cell culture

Recipe below makes 4 liters of medium pH 7.4.

RPMI 1640 (Gibco) 41.6g

HEPES (Fisher Sci) 23.8g

Hypoxanthine (Calbiochem) 0.2g

NaHCO3 8.4g

Gentamycin antibiotic (Gibco) 300uL of 50mg/ml

MilliQ water (Millipore) Make up to 4L

Albumax II (Gibco) : Asexual culture MCM 20g

Critical steps: RPMI 1640 and Albumax II powder are stored at 4°C. Stir for at least 4 hours after adding hypoxanthine before adding the rest of the ingredients (list is written in chronological order of ingredient addition). Filter- sterilize using 0.2µm membrane (VWR). For gametocyte cultures, add 5% O+ heat inactivated human serum to 0.25% Albumax II (gametocyte MCM).

- Incomplete medium: Same as above, without Albumax II or 5% Human serum

Purpose: For washing whole blood to remove buffy coat and for temporary cell suspensions and washes.

- Whole human Blood: O+ from male donor (Biological Specialty Corp.)

Purpose: Asexual P. falciparum malaria parasites infect human red blood cells (RBCs) and digest host cell hemoglobin for nutrients.

62 Critical steps: Wash 3 times to remove buffy coat that contains leukocytes and complement system at 3500rpm for 5 minutes. Store in 50% incomplete medium at 4°C and use within 10 days, making sure to re-wash and re-suspend every 72 hrs.

- 5% Sorbitol (J.T Baker)

Purpose: Used for mixed stage culture synchronization to enrich ring stage parasites because red blood cells containing mature parasites will be preferentially lysed (Lambros and Vanderberg,

1979)

- 10% Giemsa Azure Eosin methylene Blue pH 6.8 (Fluka)

Purpose: For differentially staining infected and uninfected red blood cells

Critical steps: Dissolve in distilled water and use within 24 hrs. For better resolution, use phosphate buffered giemsa, pH 7.4

A1.3 Procedure (Modified from Fivelman et al., 2007; Lucantoni et al., 2013)

1. Thaw out parasite strains from liquid nitrogen using 12% NaCl, 1.8% and

0.9%NaCl/0.2% Glucose solutions in succession, incubating for 5 minutes and shaking

between addition steps. Keep culture medium in small T25 blue vented cap flasks in

humidified 37°C incubator.

Note: Gametocytogenesis in my hands is better with recently thawed out parasites (no

older than 2 week-old culture).

2. Maintain culture at 2.5% hematocrit in 50ml MCM (with Albumax II) in 37ºC incubator.

3. At about 5% ring parasitemia determined by 10% giemsa staining, perform sorbitol

synchronization (5% D-Sorbitol) for 9 minutes to enrich for ring stage parasites. Split the

culture into 3 flasks and make up to 30ml total MCM (with 0.25% Albumax II and 5%

heat inactivated human serum) at 2.5% hematocrit.

63 4. Grow parasites for an additional 48 hr cycle and perform a 2nd sorbitol synchronization

for 9 minutes. This is termed day -4. Make cell culture up to 50ml at 1.5% hematocrit.

5. Day -3: Count 2.5-3% trophozoite parasites and make up to 50ml with gametocyte MCM.

6. Day -2: Count ring parasitemia in order to determine how much conditioned medium

(CM) to leave and how much to take out and add fresh media. This nutritional stress step

is essential for stress- induced gametocytogenesis. If 10% parasitemia, aspirate 20ml of

CM, leaving 30ml add 20 ml fresh gametocyte MCM to the flask, if 12%, leave 25 ml

CM and add 25ml of fresh gametocyte MCM.

7. Day -1: At about 11am, check for parasite stress, and add 5-10ml of gametocyte MCM to

boost growth and prevent excessive cell death, but still maintain stressful conditions.

8. At about 3.30pm, still on day -1, transfer all stressed culture to a large T225 culture

vented flask with freshly washed blood to a total of 150ml gametocyte MCM (including

the 50ml CM that was used to stress the parasites at 3% hematocrit.

9. Replace gametocyte MCM every day with 100ml fresh MCM making sure not to aspirate

too much CM since gametocytes become lighter as they mature and could be accidentally

aspirated.

10. On day 1, add heparin to prevent asexual proliferation in the gametocyte induced culture.

Make sure culture stays at 37°C because gametocytes are very sensitive to temperature

change.

11. Culture as in 9 for an additional 12 days, adding heparin up to day 5 of gametocyte

culture.

Note: If gametocytes are needed earlier than day 12, percoll purify them using carefully

pre-layered 75%/35%/ percoll, centrifuged at 4000rpm for 15 minutes with a 0

deceleration in 50ml conical flasks with equal volumes of percoll layers (75% layer at the

bottom, 35% in the middle, 3.5x diluted parasitized red blood cell culture at the top) and

64 parasite culture (diluted 3.5x). Collect the interface layer and re-suspend in fresh RBCs

and fresh gametocyte MCM. 11mls per layer for each 50ml conical flask for a total of

33ml/tube works well. Keep the pipet tip just above the liquid over a slightly tilted tube

when applying percoll layers. Be sure to balance the centrifuge with tubes opposite the

sample with the same volume of incomplete media. The top most floating layer will be

dead cells because they are the lightest, followed by gametocytes at the interface layer,

and then RBCs will be at the bottom layer. Check the smear via microscope for each

layer to verify that you have the right parasites you are looking for. Wash out the percoll

in pre-warmed incomplete medium three times before resuspension in culture.

A.2 Flow cytometry method to determine gametocyte drug inhibition

A2.1 Materials and Equipment

- Eppendorf centrifuge 5415D

- Integra ViaFlo 96 multi-channel pipettor

- Guava EasyCyte HT flow cytometer

Purpose: Flow cytometry separates cells by size and complexity as a diluted liquid sample flows through a capillary. In this case the cells desired are fluorescent green, and are separated from the background red blood cells by a green laser.

- Purified transgenic parasites tagged with GFP or fluorescent dye

- Non- sterile clear round bottomed 96-well plate (Falcon)

Purpose: Following parasite drug- incubation of 48 hours, surviving parasites must be counted using a flow cytometer. The sample is prepared by diluting the parasite sample in Hanks Buffer

Saline to maintain osmotic potential of the parasite and to minimize auto- fluorescence of the

65 MCM. These plates must be round-bottomed, not flat; to prevent damage to the capillary as it reads samples.

- Sterile Black 96-well plates (Costar)

Purpose: Drug dilutions are prepared in these and parasite culture added to them for a 48 hour incubation. Unlike the round bottomed plates, these must be sterile.

Critical steps: Pre-warm plates at 37°C to prevent moisture formation and to maintain a stable temperature for the parasite culture when it is transferred to the plate.

A2.2 Reagents

- 2X HBS

Purpose: To maintain osmotic pressure of cells when re-suspended for flow cytometry.

Diluting the sample in MCM is not recommended as RPMI 1640 contains phenol red that is

likely auto- fluorescent.

To make 250ml, use the following amounts.

D-Glucose 0.5g

HEPES 2.5g

KCl 0.18

NaCl 4g

Na2HPO4 0.05g

Critical steps: Dissolve up to 250ml of distilled water and make up to pH 7.4 with NaOH.

Filter-sterilize with 0.2µm membrane and store at 4°C. Make up to 1X HBS with sterile water

and pre-warm at 37°C before use.

- 100% DMSO

Purpose: To dissolve non-water soluble antimalarial drugs in preparation for drug assays.

66 Critical steps: Filter DMSO over a nylon 0.2µm membrane prior to adding it to drug powders

to remove any particulates. You do not need to re-filter the drug stocks after preparing them

because virtually no contaminants can grow in DMSO, as it is toxic to cells.

A2.3 Procedure (Modified from Wang et al., 2014)

1. Perform a guava clean on the flow cytometer in preparation for sample reading 15

minutes before preparing a sample.

Note: Make sure to have defined your gates for your cell sample that will clearly show a

separation between your desired cell population, in this case, green fluorescent parasites,

and the background red blood cells. You will use the same gates for multiple cell samples

of the same experiment for consistency.

2. On either day 8 or 9 of gametocyte induction (from Procedure A1), take out a 110µl

aliquot of culture at 2.5% hematocrit and spin it down for 1.5 minutes using the

Eppendorf Microfuge at 3.6rpm in a 1.5ml microfuge tube to pellet out the red blood

cells from the gametocyte MCM.

Note: Prior to drug treatment of 3 replicate experiments, one must maintain the same

hematocrit and number of initial gametocytes to avoid fluctuations in data due to an

inoculum effect (whereby an increase/decrease in parasite-load or hematocrit results in

differential drug inhibition). This step is used to measure the existing number of

gametocytes in the initial culture in order to be eventually diluted to a chosen constant

starting gametocytemia of 0.04% in all drug exposure my experiments.

3. In a new 1.5ml tube, add 600ul of pre-warmed 1x HBS (pH 7.4) and re-suspend 2.4µl of

the pelleted infected red blood cells to a total hematocrit of 0.4%.

67 4. Run the sample on the Guava EasyCyte flow cytometer using Guavasoft 2.7 software at

defined parameters. For 3D7αtubIIGFP parasites, use a very low flow rate (0.12µl/s) to

collect 150,000 events at a Forward scatter of 1.61, Side scatter of 2.48, Green laser gain

control of 2.18, Yellow gain at 8 and a Red gain of 22.6.

Note: These parameters worked well in separating out the red blood cells (clustering on

the left edge of my red vs green fluorescence dot plot) and the desired green fluorescent

parasites (clustering to the right of the red blood cells).

Fig 4-1. Dot plots of control and fluorescent parasites. Flow cytometer dot plots from left

to right of red blood cells, wild type non-fluorescent 3D7 gametocyte parasites and

3D7αtubIIGFP parasites showing differential distribution within defined low and high GFP

gates. 150,000 events were collected per sample at a very low flow rate of 0.12µl/s.

5. Record the percent number of gametocytes and dilute up to 0.04% gametocytemia with

freshly washed RBCs at a hematocrit of 2% to prepare for drug screening in sterile black

plates.

Note: 14 ml of parasitized culture at 2200 rpm for 5 minutes for all my drug assays for

consistency was centrifuged each time, as the packed cell volume can vary at higher or

lower speeds resulting in addition of more or less cells than desired to my dilution to

0.04%.

68 6. Prepare the desired concentration of antimalarial drug stocks in either water/ DMSO

depending on solubility; vortex to mix thoroughly and syringe- filter them through a

0.2µm cellulose acetate membrane for water, and 0.2µm nylon membrane for DMSO.

Depending on the Molecular weight of the drugs, 10 mM- 100mM stocks are prepared

and store them at -80°C to minimize degradation.

Note: DMSO will dissolve a cellulose acetate filter, so a recommended filter like nylon

should be used. Read the MSDS of the drugs before use because some are light sensitive

e.g chloroquine or dihydroartemisinin and must be covered with aluminum foil or

prepared in an amber light proof 1.5ml microfuge tube instead.

7. Make 4x working drug concentrations of the highest inhibitory concentration of choice of

your drugs under investigation. This is because the desired final liquid volume per well of

a 96-well plate is 200µl. We start off by adding 100µl of gametocyte MCM per well.

8. 100 µl of the 4x drug is then added to all wells in column A to make it up to 2x of the

desired concentration. 2- fold serial dilutions are then carried out using a ViaFlo

multichannel pipetting machine by aspirating 100µl from each well on the left and

dispensing it in an adjacent well on the right, mixing thoroughly, and taking out 100µl of

the resulting drug medium and dispensing it in the following adjacent well on the right

until the second to last well. The final well in column H is a negative control well with no

drug, just the drug solvent + MCM. Total DMSO percentage should be maintained below

0.4% for drugs that are dissolved in this solvent.

9. Gently mix the previously prepared 0.04% gametocyte culture in 2% red blood cells and

add 100µl to each well in the 96-well plate for a total hematocrit of 1%.

10. Store the completed drug dosed parasite plates at 37°C in a humidified CO2 incubator for

48 hours without agitation. Note the time because 48 hours later, the plates will be read

by flow cytometry.

69 11. After 47 hours, start preparing the sample for flow cytometry to detect surviving

parasites. 1X HBS and clear round bottomed 96-well plates for flow cytometry analysis

should be warmed to 37°C at this point. The flow cytometer should be cleaned at least

fifteen minutes prior to sample analysis.

12. At the 48- hour time point, obtain a pre-warmed clear round bottom 96-well plate from

the 37°C incubator. Add 120µl of pre-warmed 1X HBS to each well in the 96-well plate.

Mix gently and add 80µl of each sample per well for a total of 200ul cell sample for a

final hematocrit of 0.4%.

Note: Check the upper limit of cell concentration of your flow cytometer. This particular

flow cytometer clogs up if the cell concentration goes above 3500 cells/µl, so a

hematocrit of ~0.25 and 0.45% is ideal for sample analysis.

13. Read the sample using the flow cytometer making sure to collect at least 25,000 cells at a

high flow rate.

Note: One 96-well plate takes exactly 1 hour to read if 25,000 cells are collected.

Medium or low flow rates may be chosen for increased precision if one has fewer

samples, however, waiting for >2 hours/plate may not be ideal if one has more than 3

plates to sample. Make sure to save the sample files as .FCS for further analysis using

Flow Jo software.

14. Flow Jo V10 software is used to analyze the dot plots obtained by plotting the red

fluorescence versus green fluorescence and defining a gate for the desired GFP cell

population.

Note: In order to determine inhibitory drug concentrations, data are normalized by the

formula Normalized gating count= (Number of events in defined gate/Total events)*

25,000. The fluorescence intensity (FI) is then obtained by FI= Normalized gating count

* Mean green fluorescence. FI on the y axis is then plotted against the drug concentration

70 to obtain sigmoid curves that are analyzed by non-linear regression (GraphPad Prism) in order to obtain the median inhibitory drug concentrations per drug on the fluorescent gametocytes tested.

71 A2.4 References

Fivelman, Quinton L., Louisa McRobert, Sarah Sharp, Cathy J. Taylor, Maha Saeed,

Claire A. Swales, Colin J. Sutherland, and David A. Baker. "Improved synchronous production of

Plasmodium falciparum gametocytes in vitro."Molecular and biochemical parasitology 154, no. 1

(2007): 119-123.

Lambros, Chris, and Jerome P. Vanderberg. "Synchronization of Plasmodium falciparum erythrocytic stages in culture." The Journal of parasitology (1979): 418-420.

Lucantoni, Leonardo, Sandra Duffy, Sophie H. Adjalley, David A. Fidock, and Vicky M.

Avery. "Identification of MMV malaria box inhibitors of Plasmodium falciparum early-stage gametocytes using a luciferase-based high-throughput assay." Antimicrobial agents and chemotherapy 57, no. 12 (2013): 6050-6062.

Miao, Jun, Zenglei Wang, Min Liu, Daniel Parker, Xiaolian Li, Xiaoguang Chen, and

Liwang Cui. "Plasmodium falciparum: Generation of pure gametocyte culture by heparin treatment." Experimental parasitology 135, no. 3 (2013): 541-545.

Smilkstein, Martin, Nongluk Sriwilaijaroen, Jane Xu Kelly, Prapon Wilairat, and Michael

Riscoe. "Simple and inexpensive fluorescence-based technique for high-throughput antimalarial drug screening." Antimicrobial agents and chemotherapy 48, no. 5 (2004): 1803-1806.

Wang, Zenglei, Min Liu, Xiaoying Liang, Salil Siriwat, Xiaolian Li, Xiaoguang Chen,

Daniel M. Parker, Jun Miao, and Liwang Cui. "A flow cytometry-based quantitative drug sensitivity assay for all Plasmodium falciparum gametocyte stages." PloS one 9, no. 4 (2014): e93825.

72 Appendix B: Drug-Drug interaction isobolograms for each lab strain

1.2 MFQ 1 HB3 PPQ 0.8 AMQ 0.6 LMF

FIC TFQ FIC 0.4 NQ PND 0.2 CQ 0 0 0.5 1 1.5 FIC X

1.2 DD2 MFQ 1 PPQ

0.8 AMQ 0.6 LMF

FIC TFQ FIC 0.4 NQ PND 0.2 CQ 0 0 0.5 1 1.5 FIC X

1.2 IPC5202 1 MFQ PPQ

0.8 AMQ 0.6 LMF FIC TFQ FIC 0.4 NQ PND 0.2 CQ 0 0 0.5 1 1.5 FIC X

73

1.2 7G8 MFQ 1 PPQ AMQ 0.8 LMF 0.6

NQ FIC TFQ FIC 0.4 PND 0.2 CQ 0 0 0.5 1 1.5 FIC X

1.2 3D7 1 MFQ PPQ

0.8 AMQ 0.6 LMF FIC TFQ FIC 0.4 NQ PND 0.2 CQ 0 0 0.5 1 1.5 2 2.5 FIC X

3 3D7αtubIIGFP 2.5 MFQ

2 PPQ

1.5 AMQ FIC TFQ FIC 1 LMF NQ 0.5 PND 0 0 1 2 3 4 5 CQ FIC X

Fig 5-1 Isobolograms of asexual and sexual parasite strains. Mean fractional inhibitory concentrations per fixed ratio of tafenoquine are plotted against those for the ACT-partner drug

(FICTFQ vs FICX).