31.08.2013 Views

Experimental infection and protection against ... - TI Pharma

Experimental infection and protection against ... - TI Pharma

Experimental infection and protection against ... - TI Pharma

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>Experimental</strong> <strong>infection</strong> <strong>and</strong> <strong>protection</strong><br />

<strong>against</strong> Plasmodium falciparum malaria in<br />

humans<br />

Meta Roestenberg


ISBN 978-90-9027235-1<br />

Cover photo by Iris Spaink, www.irisspaink.nl<br />

Printed by Ridderprint<br />

Copyright 2013© Meta Roestenberg<br />

All rights reserved. No part of this book may be reproduced or transmitted in<br />

any form or by any means, electronic of mechanical, including photocopying,<br />

recording, or by any information storage <strong>and</strong> retrieval system without express<br />

written permission from the autor, or where appropriate, the publisher of the<br />

articles


<strong>Experimental</strong> <strong>infection</strong> <strong>and</strong> <strong>protection</strong><br />

<strong>against</strong> Plasmodium falciparum malaria in<br />

humans<br />

Proefschrift<br />

ter verkrijging van de graad van doctor<br />

aan de Radboud Universiteit Nijmegen<br />

op gezag van de rector magnificus prof. mr. S.C.J.J.Kortman,<br />

volgens besluit van het college van Decanen<br />

In het openbaar te verdedigen op ma<strong>and</strong>ag 14 januari 2013<br />

om 15.30 uur precies<br />

Door<br />

Meta Roestenberg<br />

Geboren op 24 januari 1981<br />

te De Bilt


Promotoren:<br />

Prof. dr. R.W. Sauerwein<br />

Prof. dr. A.J.A.M. van der Ven<br />

Manuscriptcommissie:<br />

Prof. dr. P.A.B.M. Smits<br />

Prof. dr. C.G. Figdor<br />

Prof. dr. J.T. van Dissel, Leiden University


Contents<br />

Contents 5<br />

Chapter 1 - Introduction 7<br />

Section 1 - Apical Membrane Antigen 1 29<br />

Chapter 2 - Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium<br />

falciparum AMA1 Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide<br />

ISA 720 or AS02. 31<br />

Chapter 3 - Humoral immune responses to a single allele PfAMA1 vaccine in<br />

healthy malaria-naïve adults. 57<br />

Chapter 4 - A saturation of antibody avidity <strong>and</strong> concentration induced by<br />

malaria vaccine c<strong>and</strong>idate Apical Membrane Antigen 1 81<br />

Section 2 - Controlled human malaria <strong>infection</strong> model 95<br />

Chapter 5 - Comparison of clinical <strong>and</strong> parasitological data from experimental<br />

human malaria challenge trials 97<br />

Chapter 6 - Efficacy of pre-erythrocytic <strong>and</strong> blood-stage malaria vaccines can be<br />

assessed in small sporozoite challenge trials in human volunteers 117<br />

Chapter 7 - NF135.C10: a new Plasmodium falciparum clone for controlled<br />

human malaria <strong>infection</strong>s 129<br />

Chapter 8 - Induction of malaria in volunteers by intradermal injection of<br />

cryopreserved Plasmodium falciparum sporozoites 151<br />

Section 3 - Whole parasite inoculation 171<br />

Chapter 9 - Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 173<br />

Chapter 10 - Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite<br />

inoculation 193<br />

Chapter 11 - General Discussion 215<br />

Chapter 12 - Summary, Samenvatting, List of publications, Dankwoord, C.V. 243


Chapter 1<br />

Introduction


Introduction 9<br />

Malaria<br />

Malaria is one of the world's most common infectious diseases with an annual<br />

225 million cases leading to an estimated 655 000 deaths in 2010, mainly among<br />

young children. Malaria morbidity <strong>and</strong> mortality is particularly severe in Sub-<br />

Saharan Africa, where 91% of deaths occur [1](Figure 1).<br />

Figure 1. Malaria, countries or areas at risk for transmission, 2010.<br />

www.who.int/malaria<br />

Malaria is responsible for an enormous economic burden due to medical<br />

expenses, missed education <strong>and</strong> lost productivity. It has been estimated that the<br />

GDP of countries endemic for malaria grows at 1.3% per year slower than<br />

countries which are malaria-free, even after other factors such as initial income<br />

level, overall health <strong>and</strong> tropical location are taken into account [2]. Moreover,<br />

in highly endemic settings up to 50% of low birth weight (LBW) deliveries can be<br />

attributed to malaria in pregnancy, leading to approximately 100 000 infant<br />

deaths annually [3].<br />

Plasmodium falciparum (Pf) is the malaria parasite species responsible for<br />

almost all global malaria deaths. It is one of five human malaria parasite species<br />

transmitted by the bites of infected Anopheles mosquitoes. A clinical Pf <strong>infection</strong><br />

in humans is characterised by non-specific symptoms such as fever, headache,<br />

myalgia, fatigue <strong>and</strong> abdominal discomfort. Neurological deficits such as coma<br />

or convulsions <strong>and</strong> severe anemia are signs of severe disease <strong>and</strong> have high<br />

mortality rates [4]. Plasmodium vivax, ovale, malariae <strong>and</strong> knowlesi cause a


10 Chapter 1<br />

generally milder disease that is rarely fatal, but, in the case of vivax <strong>and</strong> ovale<br />

malaria, that may remain in a dormant form for years.<br />

Malaria control receives high priority from the international community, which<br />

has been established in the United Nation's Millennium Development Goal 6<br />

aiming at the reduction of the incidence of malaria <strong>and</strong> other major diseases by<br />

2015. Malaria control is also central to Millennium Development Goal 4,<br />

targeting a two-third reduction in the mortality rate among children under the<br />

age of five [5].<br />

Current control strategies are based on early diagnosis <strong>and</strong> treatment of malaria<br />

<strong>infection</strong>s combined with preventive measures aimed at mosquito control. The<br />

development of widespread resistance to the anti-malarial drugs chloroquine<br />

<strong>and</strong> sulfadoxine-pyrimethamine has re-directed treatment strategies to<br />

artemisinine-based combination therapies, such as artemether plus<br />

lumefantrine, artesunate plus amodiaquine, artesunate plus mefloquine or<br />

artesunate plus sulfadoxine-pyrimethamine, which are now the WHOrecommended<br />

treatment regimens. Unfortunately, Pf resistance has been<br />

observed to many of the currently used antimalarial drugs (amodiaquine,<br />

chloroquine, mefloquine, quinine <strong>and</strong> sulfadoxine-pyrimethamine) <strong>and</strong>, more<br />

recently, also to artemisinine derivatives [6]. To date, there are no new<br />

medicines in advanced stages of development to replace the artimisinins [7].<br />

Vector control is the primary intervention for reducing malaria transmission at<br />

the community level. When universal vector control coverage is achieved by<br />

impregnating bednets <strong>and</strong> spraying indoor surfaces of houses with insecticides,<br />

transmission can be reduced to close to zero. However, the increasing resistance<br />

of mosquitoes to insecticides including dichlorodiphenyltrichloroethane (DDT)<br />

<strong>and</strong> pyrethroids, particularly in Africa, poses challenges to current prevention<br />

policies [8].<br />

The current control strategies permitted the interruption of malaria<br />

transmission in low transmission countries, particularly in those with a robust<br />

institutional infrastructure <strong>and</strong> well-functioning health systems, <strong>and</strong> in those<br />

neighbouring malaria-free areas [8]. However, the lack of new effective antimalarial<br />

drugs <strong>and</strong> insecticides places malaria control <strong>and</strong> elimination efforts at<br />

considerable risk. In order to reach the ultimate goal of malaria eradication,<br />

much greater gains could be achieved with currently available tools, including<br />

elimination from a number of countries <strong>and</strong> regions, but even with maximal<br />

effort we will fall short of elimination in many areas <strong>and</strong> of global eradication


Introduction 11<br />

Preclinical Clinical<br />

Phase 1A Phase 1B Phase 2A Phase 2B Phase 3<br />

Aim Safety, dose, efficacy, long-term Safety & Safety & Efficacy & Efficacy & Adverse<br />

effects<br />

dose dose side effects side effects reactions<br />

<strong>and</strong> longterm<br />

Population Rodent <strong>and</strong>/or simian Non- Endemic Non- Endemic Endemic<br />

endemicendemic<br />

Duration 3-6 years 3-12 3-12 3-24 3-24 3 years<br />

months months months months<br />

Typical size of<br />

~10 ~10 ~10 100-300 10,000test<br />

group<br />

RTS,S/AS01E<br />

AdCh63/MVA ME-TRAP<br />

PfSPZ: metabolically active, nonreplicating<br />

malaria sporozoite vaccine<br />

Adenovirus (Ad35) vectored CS<br />

polyepitope DNA EP1300<br />

CSVAC<br />

Pf CelTOS FMP012<br />

Ad35.CS.01/Ad26.CS.01<br />

(Heterologous adenovirus primeboost<br />

strategy)<br />

Two-component paediatric malaria<br />

vaccine (CS)<br />

Pf GAP p52-/p32- (Genetically<br />

Attenuated Sporozoites)<br />

RTS,S/AS02A<br />

DNA/MVA prime-boost Multi-Epitope<br />

(ME) string + TRAP<br />

FP9 MVA prime-boost ME-TRAP<br />

FP9 CSP + LSA-1 epitope/ MVA-CSP +<br />

LSA-1 epitope<br />

MuStDO5 (Multi-Stage DNA vaccine<br />

Operation, 5 antigens)<br />

NMRC-MV-Ad-PfC<br />

CSP long synthetic peptide<br />

LSA-3 (inactive)<br />

FMP011/AS01B (LSA-1 E. coliexpressed<br />

in AS01B adjuvant)<br />

FMP011/AS02A (LSA-1 E. coliexpressed<br />

in AS02A adjuvant)<br />

DNA/MVA CSP<br />

HepB Core-Ag CSP VLP<br />

RTS,S/AS02 <strong>and</strong> SSP2/TRAP<br />

RTS,S/AS02 <strong>and</strong> MVA CSP<br />

RTS,S/AS02 <strong>and</strong> DNA CSP<br />

CSP DNA immunization<br />

50,000<br />

Pre-erythrocytic vaccine c<strong>and</strong>idates<br />

Figure 2: Malaria vaccines in (pre-)clinical development. Vaccines in lighter shades have<br />

stopped or halted in development. [16]<br />

[7]. Current efforts should thus be supplemented with a structured research<br />

agenda taking a multidisciplinary global approach [9]. An effective malaria<br />

vaccine is a key tool of critical research that is needed to support malaria control<br />

<strong>and</strong> eradication [9]. Ambitious goals in this regard have been set by the Malaria<br />

Vaccine Technology Roadmap, which aims to develop <strong>and</strong> license a malaria<br />

vaccine with more than 80% protective efficacy <strong>against</strong> clinical disease that lasts<br />

longer than four years, by the year 2025 [10]. However, despite extensive


12 Chapter 1<br />

Preclinical Clinical<br />

Phase 1A Phase 1B Phase 2A Phase 2B Phase 3<br />

Aim Safety, dose, efficacy, long-term effects Safety & dose Safety & dose Efficacy & side Efficacy & side Adverse<br />

effects effects reactions <strong>and</strong><br />

long-term<br />

Population Rodent <strong>and</strong>/or simian Non-endemic Endemic Non-endemic Endemic Endemic<br />

Duration 3-6 years 3-12 months 3-12 months 3-24 months 3-24 months 3 years<br />

Typical size of<br />

test group<br />

~10 ~10 ~10 100-300 10,000-50,000<br />

FMP2.1/AS02A (AMA-1 3D7 E. coli-expressed in<br />

Erythrocytic vaccine c<strong>and</strong>idates<br />

Combined vaccine<br />

c<strong>and</strong>idates<br />

Transmissio<br />

n blocking<br />

vaccine<br />

c<strong>and</strong>idates<br />

GMZ2<br />

MSP3 [181-276]<br />

AMA1-C1/Alhydrogel ® + CPG 7909<br />

AdCh63/MVA MSP1<br />

FMP2.1/AS01B (AMA-1 3D7 E. coli expressed<br />

in ASO1B adjuvant)<br />

AdCh63 AMA1/MVA AMA1<br />

EBA175 RII<br />

FMP010/AS01B (MSP-1 42 FVO E. coliexpressed<br />

in AS01B adjuvant)<br />

SE36<br />

BSAM-2/Alhydrogel®+CPG 7909<br />

JAIVAC (MSP1 19/EBA175)<br />

P27A<br />

pfAMA1 DiCo<br />

PAM VAR2CSA<br />

Var2-CSA DBL2/3-X<br />

MSP1 full length<br />

FMP1/AS02A (MSP-1 42 3D7 E. coli-expressed<br />

in AS02A adjuvant)<br />

MSP2-C1/ISA720<br />

AMA1-FVO [25-545]<br />

PfCP2.9 (MSP-1 19/AMA-1 chimera)<br />

GLURP [85-213]<br />

MSP1-C1/AlOH/AlOH + CpG<br />

AMA1-C1/ ISA720<br />

AMA-1 (Australia)<br />

NMRC-M3V-Ad-PfCA<br />

CSP, AMA1 virosomes (PEV 301,302)<br />

SR11.1<br />

NMRC-M3V-Ad-Pf5<br />

RESA, MSP1, MSP2 (combination B)<br />

FP9/MVA Polyprotein<br />

RTS,S/AS02 <strong>and</strong> FMP1/AS02<br />

NMRC-M3V-Ad-PfCA in Adjuvant (7DW8-5)<br />

Pf10C-MBP<br />

Pfs25-rEPA/Alhydrogel<br />

Pfs25-Pfs25 conjugate vaccine<br />

Pfs25<br />

Figure 2 (continued): Malaria vaccines in (pre-)clinical development. Vaccines in lighter<br />

shades have stopped or halted in development. [16]<br />

research efforts <strong>and</strong> a significant increase in the number of vaccine c<strong>and</strong>idates<br />

being developed, there is no licensed malaria vaccine yet.<br />

Malaria vaccine development<br />

Developments in molecular techniques have advanced the development of<br />

malaria vaccine c<strong>and</strong>idates to a current number of approximately 36 Pf<br />

c<strong>and</strong>idate (sub-unit) malaria vaccines in clinical development [11], an overview is<br />

provided in Figure 2. Only a fraction of lead vaccine c<strong>and</strong>idates entering the


Introduction 13<br />

pipeline of clinical development will ultimately be licensed for clinical use.<br />

Clinical development is time-consuming <strong>and</strong> costly <strong>and</strong> starts with safety <strong>and</strong><br />

immunogenicity trials. If satisfactory, clinical trials aimed at obtaining efficacy<br />

profiles will be undertaken. Whereas the first two criteria can generally be<br />

assessed in a small initial Phase I trial, vaccine efficacy in the field can only be<br />

assessed in larger study cohorts with at least a few hundred volunteers in<br />

malaria-endemic areas (Phase IIb trials). With only a limited number of<br />

competent field trial sites <strong>and</strong> a downward trend in malaria incidence in several<br />

endemic areas, the size <strong>and</strong> costs of Phase IIb trials are rising [12], underpinning<br />

the need to develop tools for downstream selection of vaccine c<strong>and</strong>idates.<br />

Conventional vaccine technologies rely on the production of a single<br />

recombinant or synthetic subunit protein or peptide vaccine in a yeast or<br />

bacterial vector or administer (attenuated) whole organisms [13]. Proteins may<br />

also be combined together to enhance <strong>protection</strong> or increase the breath of the<br />

immune response. Alternatively, DNA vaccines have been tried, but they proved<br />

to be less immunogenic [14]. In order to increase or modify the vaccine-elicited<br />

immune response, vaccines are often administered in conjunction with an<br />

adjuvant. Adjuvants can consist of many different materials that are either<br />

mixed with or linked to the vaccine. Moreover, delivery platforms can deliver a<br />

vaccine to a certain site, such as (Adeno-)viral vectors or liposomes that can take<br />

care of intracellular delivery of the vaccine. In such cases, the DNA-sequence<br />

vaccine is expressed by the host cell. Particularly combinations of a viralvectored<br />

DNA prime <strong>and</strong> protein booster injections have proven to be successful<br />

[15].<br />

Malaria vaccine c<strong>and</strong>idates<br />

The vaccine c<strong>and</strong>idates currently under pre-clinical or clinical development are<br />

categorized according to the life cycle stage of antigen expression (Figure 3). The<br />

pre-erythrocytic stage begins when a female Anopheles mosquito inoculates Pf<br />

sporozoites into the human skin. These sporozoites travel to the human liver<br />

<strong>and</strong> invade hepatocytes, where they develop into thous<strong>and</strong>s of asexual parasites<br />

called merozoites. This process takes approximately one week <strong>and</strong> is clinically<br />

silent. Pre-erythrocytic stage vaccines aim to prevent the passage of parasites<br />

through the human liver <strong>and</strong> subsequent blood-stage <strong>infection</strong>. Ideally, such<br />

vaccines should be 100% effective to prevent disease, since any merozoite<br />

released from the liver is potentially capable of exponential growth by invasion


14 Chapter 1<br />

Pre-erythrocytic stage<br />

Asexual<br />

erythrocytic stage<br />

Merozoites<br />

invade<br />

erythrocytes<br />

Asexual<br />

blood stages<br />

Sexual <strong>and</strong><br />

mosquito stage<br />

Blood vessel<br />

Exo-erythrocytic<br />

schizont<br />

Sporozoites released<br />

into bloodstream<br />

Sporozoite<br />

Sporozoites<br />

enter liver cells<br />

Some merozoites<br />

develop into<br />

gametocytes<br />

Lymph<br />

node<br />

Merozome<br />

Liver cells <strong>and</strong><br />

merosomes rupture<br />

<strong>and</strong> merozoites<br />

released into<br />

bloodstream<br />

Merozoite<br />

Erythrocytic<br />

schizont<br />

Mosquito<br />

Salivary<br />

gl<strong>and</strong><br />

Gut<br />

Zygote<br />

Ookinete<br />

Gametes<br />

Gametocytes taken<br />

up by mosquito in<br />

bloodmeal<br />

Mosquito<br />

Sporozoites<br />

released <strong>and</strong><br />

travel to<br />

salivary gl<strong>and</strong>s<br />

Sporozoite<br />

Oocyst<br />

Male <strong>and</strong> female<br />

gametes form<br />

zygotes <strong>and</strong><br />

oocysts develop<br />

Figure 3. Plasmodium falciparum life cycle showing the three developmental stages of<br />

the parasite that are targeted by malaria vaccine c<strong>and</strong>idates [12].<br />

of red blood cells (erythrocytic or blood stage). This developmental stage is<br />

responsible for the clinical signs <strong>and</strong> symptoms of malaria <strong>infection</strong>. Erythrocytic<br />

stage vaccines focus on inhibiting parasite multiplication, thereby decreasing the<br />

probability of complications <strong>and</strong> (severe) disease. In contrast to pre-erythrocytic<br />

vaccines, an erythrocytic vaccine may therefore be only partially efficacious <strong>and</strong><br />

can still protect <strong>against</strong> severe disease. Following asexual multiplication in<br />

erythrocytes, a small percentage of merozoites convert to sexual forms, the<br />

gametocytes. Gametocytes are infectious to Anopheles mosquitoes that bite the<br />

infected host. Transmission blocking vaccines contain sexual blood stage or<br />

mosquito stage antigens that prevent <strong>infection</strong> of mosquitoes <strong>and</strong> thus further<br />

transmission of the parasite, reducing the spread of malaria in the population.


Introduction 15<br />

The division of antigen expression according to parasite life-cycle stages,<br />

although useful, may be a simplification of reality since many antigens are<br />

shared between stages [17-19].<br />

Pre-erythrocytic vaccine c<strong>and</strong>idates<br />

The development of pre-erythrocytic vaccines started with the observation by<br />

Ruth Nussenzweig that vaccination of mice with irradiated sporozoites results in<br />

<strong>protection</strong> from sporozoite challenge. [20]. Following this observation, the<br />

exploration of sporozoite antigens lead to the identification of the<br />

circumsporozoite protein (CS), which was capable of inducing immunity in mice<br />

[21]. Further development of this antigen, particularly by the Walter Reed Army<br />

Institute of Research in the United States, has resulted in the most advanced<br />

vaccine c<strong>and</strong>idate so far: GlaxoSmithKline's RTS,S vaccine. This c<strong>and</strong>idate<br />

consists of a Hepatitis B surface antigen fused to the Pf circumsporozoite<br />

protein, adjuvanted by GlaxoSmithKline's proprietary adjuvant AS01 containing<br />

the immunostimulants MPL <strong>and</strong> QS21 in liposomes. It is currently in phase III<br />

clinical development <strong>and</strong> has consistently shown 30-60% clinical <strong>protection</strong> [22-<br />

27].<br />

Other promising pre-erythrocytic antigens include thrombospondin-related<br />

adhesion protein (TRAP) <strong>and</strong> liver stage antigen 1 <strong>and</strong> 3 (LSA). Cytokine<br />

responses to LSA-1 were associated with parasitemia <strong>and</strong> clinical malaria in<br />

endemic human populations <strong>and</strong> the antigen is safe <strong>and</strong> immunogenic in<br />

primates <strong>and</strong> humans [28-30]. Unfortunately, a phase IIa efficacy trial with the<br />

AS01/AS02 adjuvanted recombinant LSA1 product did not show any <strong>protection</strong><br />

or delay in prepatent period [31]. The vaccine c<strong>and</strong>idate LSA-3 was safe <strong>and</strong><br />

immunogenic in humans, but did not protect malaria naïve volunteers in a<br />

controlled human malaria <strong>infection</strong> trial (Nieman et al. manuscript in<br />

preparation). A vaccine based on multiple T- <strong>and</strong> B-cell epitopes, amongst which<br />

LSA-1 <strong>and</strong> LSA-3, fused to the TRAP antigen protected malaria-naïve subjects<br />

<strong>against</strong> subsequent challenge with infectious mosquitoes, when administered as<br />

a heterologous prime-boost schedule whereby the plasmid DNA vaccine is<br />

boosted by a viral-vectored vaccine [32], but did not show any efficacy in field<br />

trials in both adult <strong>and</strong> children [33, 34].<br />

The most successful immunization strategy so far, however, was directly derived<br />

from the irradiated sporozoites used in the murine models of Ruth Nussenzweig.<br />

Infected, laboratory-reared Anopheles stephensi mosquitoes were irradiated <strong>and</strong>


16 Chapter 1<br />

allowed to bite on human volunteers. Irradiation attenuated the sporozoites<br />

located in the mosquito salivary gl<strong>and</strong>. It was shown that more than a 1000 of<br />

such bites could protect humans from subsequent challenge [35]. The technical<br />

challenge of whole-sporozoite immunization is the manufacturing process, since<br />

the sporozoite parasite forms cannot be cultured outside their mosquito host.<br />

The production of whole-sporozoite vaccines that comply with current vaccine<br />

regulations for registration thus requires the st<strong>and</strong>ardized isolation, purification<br />

<strong>and</strong> preservation of parasites from mosquito salivary gl<strong>and</strong>s. This procedure<br />

meets numerous technical hurdles [36].<br />

Blood stage vaccine c<strong>and</strong>idates<br />

Despite the fact that blood stage parasite multiplication is the only parasite<br />

developmental stage that causes disease, relatively few blood-stage antigens are<br />

in clinical development as vaccines thus far. Nevertheless, erythrocytic stage<br />

vaccines are considered valuable, because they may not only be used as single<br />

antigen vaccines but also protect <strong>against</strong> break-through <strong>infection</strong>s in a multistage<br />

vaccine with pre-erythrocytic vaccine components [37]. Erythrocytic stage<br />

malaria vaccine c<strong>and</strong>idates include Apical Membrane Antigen 1, (AMA1),<br />

Erythrocyte-Binding Antigen-175 (EBA-175), Glutamate-Rich Protein (GLURP),<br />

Merozoite Surface Protein 1 (MSP1), MSP2, MSP3 <strong>and</strong> Serine-Repeat Antigen 5<br />

(SERA5), all of which are highly expressed on the merozoite surface [11]. To<br />

date, none of these c<strong>and</strong>idates have demonstrated <strong>protection</strong> <strong>against</strong> clinical<br />

outcomes [37]. Efficacy of these c<strong>and</strong>idates may be hampered by the genetic<br />

diversity of the parasite surface proteins, which is likely due to the selective<br />

pressure exerted by the human immune response [37].<br />

AMA1 is one of the most advanced blood stage malaria vaccine c<strong>and</strong>idates.<br />

Animal data consistently show that AMA1 antibodies alone can protect mice <strong>and</strong><br />

monkeys <strong>against</strong> subsequent challenge, raising high hopes for the vaccine [38-<br />

40]. The protein is maximally expressed in late schizony of erythrocytic<br />

development <strong>and</strong> relocates from the parasite microneme to the merozoite<br />

surface, where it plays a role in erythrocyte invasion [41]. Possibly, AMA1 is also<br />

involved in liver cell invasion [42], making the antigen an attractive target<br />

particularly for antibodies. Unfortunately, AMA1 is also highly polymorphic with<br />

hundreds of haplotypes identified [43]. Another potential target for blood-stage<br />

vaccines is the Pf erythrocyte membrane protein 1 (PfEMP1) family, a highly<br />

polymorphic protein encoded by a great number of genes. The PfEMP1s are


Introduction 17<br />

Figure 4. Population indices of immunity to malaria in Kilifi, Kenya. [53]<br />

expressed on the surface of infected erythrocytes <strong>and</strong> are essential for<br />

adherence of the parasite to vascular endothelium to avoid destruction in the<br />

spleen [11, 44]. As one parasite clone expressing one PfEMP1 is detected by the<br />

immune system, another parasite clone expressing another PfEMP1 takes over.<br />

Despite its diversity, efforts are undertaken to identify conserved domains or<br />

induce antibodies <strong>against</strong> PfEMP1s, since these antigens are responsible for<br />

specific disease manifestations such as placental or cerebral malaria [45, 46].<br />

Placental malaria is mediated by parasites expressing a specific variant surface<br />

antigen, var2CSA [47]. Acquired immunity to placental malaria in multigravid<br />

women corresponds with acquisition of var2CSA-specific antibodies [48].<br />

Transmission blocking vaccine c<strong>and</strong>idates<br />

Transmission Blocking Vaccines consist of molecules eliciting the production of<br />

antibodies that will interfere with the parasite development in the mosquito.<br />

Four Pf antigens have attracted most attention as potential components of<br />

mosquito stage vaccines: gametocyte/gamete antigens Pfs230 <strong>and</strong> Pfs48/45 <strong>and</strong><br />

zygote/ookinete antigens Pfs25 <strong>and</strong> Pfs28. Naturally occurring antibodies<br />

directed <strong>against</strong> these proteins can be found in people living in endemic areas<br />

[49, 50]. Nonetheless, these antigen targets show very limited sequence<br />

diversity compared to blood stage antigens. A major practical difficulty has<br />

been in expressing the proteins in the correct conformation to achieve<br />

immunogenicity [51]. A phase I trial of Pfs25 in healthy human volunteers was<br />

halted prematurely because of systemic reactogenicity [52].


18 Chapter 1<br />

Malaria immunity<br />

Central to the hampering vaccine development is our limited underst<strong>and</strong>ing of<br />

precisely what constitutes immunity to Pf malaria. In areas of natural exposure,<br />

clinical immunity to malaria slowly develops in repeatedly exposed humans. In<br />

such populations, immunity to severe disease develops in early childhood,<br />

whereas immunity to mild disease is not typically acquired until late adolescence<br />

[53, 54] (Figure 4). Immunity is never sterile <strong>and</strong> the cumulative incidence of<br />

parasitemia may approach 100% in adults [55]. Instead, natural acquired<br />

immunity appears to be directed at the control of parasite replication <strong>and</strong><br />

limited parasite clearance.<br />

Both humoral <strong>and</strong> cellular mechanisms contribute to the immunological<br />

responses to Pf parasites, depending on the stage of the life cycle. Although<br />

antibodies can inhibit the invasion of parasites into hepatocytes [56], rodent<br />

studies suggest that pre-erythrocytic immunity is dependent mainly upon<br />

cascades of cellular <strong>and</strong> cytokine interactions [57, 58]. CD8+ T cells have been<br />

implicated as the principal effector cells, <strong>and</strong> IFN-γ as a critical effector molecule<br />

[59, 60].<br />

Given the absence of antigen processing in erythrocytes, immunity to blood<br />

stage malaria parasites is primarily conferred by humoral immune responses<br />

[61], although T-cells may be able to also limit blood stage parasite growth by<br />

enhancing phagocytosis by macrophages [62]. The importance of antibodies in<br />

blood stage parasite inhibition was originally demonstrated by passive transfer<br />

experiments [63, 64], whereby the transfer of antibodies from exposed<br />

volunteers suppressed parasitemia in Pf patients. Antibodies may act<br />

independently in inhibiting parasite growth, as shown by in vitro parasite growth<br />

inhibition assays (GIA) [65], or they may assist the cellular killing of parasites, an<br />

in vitro phenomenon called antibody-dependent cell-mediated cytotoxicity [66].<br />

Ideally, immunological assays should be able to predict the efficacy of malaria<br />

vaccines. Unfortunately, immune correlates of <strong>protection</strong> are lacking <strong>and</strong> thus<br />

malaria vaccine development depends on a trial-<strong>and</strong>-error approach [12].<br />

Controlled malaria <strong>infection</strong>s<br />

A controlled malaria <strong>infection</strong> provides the opportunity of obtaining preliminary<br />

efficacy data for a vaccine c<strong>and</strong>idate in a selected small group of malaria naïve<br />

volunteers <strong>and</strong> act as the key tool for downstream selection of vaccine


Introduction 19<br />

c<strong>and</strong>idates. Initially developed as a therapy for neurosyphilis in the 1920’s [67],<br />

controlled malaria <strong>infection</strong>s were established in their present form in the<br />

1980s, when techniques to culture parasites <strong>and</strong> obtain laboratory-reared<br />

infectious mosquitoes were available [68-70]. In controlled <strong>infection</strong> trials,<br />

human (malaria-naïve) volunteers are deliberately exposed to bites of a predefined<br />

number of infectious mosquitoes. Subjects are subsequently followed,<br />

mostly on an out-patient basis, for clinical <strong>and</strong> parasitological endpoints. As soon<br />

as parasites are detected in the circulation by microscopy, volunteers are<br />

treated with anti-malarials for obvious safety reasons. As such, parasitemia is<br />

generally kept below 0.0004% (~10-20 Pf/µl) <strong>and</strong> severe malaria does not occur<br />

[71-73]. By comparing the proportion of infected subjects between vaccine <strong>and</strong><br />

control groups, preliminary efficacy data can be obtained. If clinical <strong>protection</strong> is<br />

not achieved, the pre-patent period may be used as a surrogate marker<br />

preceding clinical <strong>protection</strong>. The recent development of molecular techniques<br />

for the detection <strong>and</strong> quantification of parasites [74] has further refined the<br />

parasitological endpoints to include a detailed analysis of kinetics of blood stage<br />

parasite growth. In addition, statistical models have become available to analyse<br />

these data [75, 76]. Such analysis may be helpful to identify the mechanism of<br />

immunity induced by the vaccine c<strong>and</strong>idate [77, 78].<br />

Controlled malaria <strong>infection</strong> studies have halted the development of c<strong>and</strong>idate<br />

vaccines LSA1 <strong>and</strong> PfCS102 [31, 79], whereas a ~30-50% <strong>protection</strong> induced by<br />

the vaccine c<strong>and</strong>idate RTS,S in an initial controlled <strong>infection</strong> study was translated<br />

into a similar protectivity <strong>against</strong> clinical malaria in the field [27, 80-83].<br />

Controlled malaria <strong>infection</strong> trials are thought particularly suitable for evaluation<br />

of pre-erythrocytic malaria vaccine c<strong>and</strong>idates, since they employ the natural<br />

route of <strong>infection</strong> (mosquito bite) <strong>and</strong> allow full pre-erythrocytic stage<br />

development. For asexual erythrocytic stage vaccines, the use of controlled<br />

malaria <strong>infection</strong>s is controversial, because only low levels of blood stage<br />

parasitemia are induced until the trial is interrupted by treatment [12]. Vaccines<br />

would thus have to exert significant inhibition in a very small time window.<br />

Because controlled human malaria <strong>infection</strong> trials require the combined<br />

technical ability to rear Pf infected mosquitoes <strong>and</strong> the clinical facility to closely<br />

monitor healthy human volunteers, such trials are currently routinely carried out<br />

in only five institutions worldwide: the US Military Malaria Vaccine Program; the<br />

University of Maryl<strong>and</strong>, USA; the Radboud University Nijmegen Medical Centre,


20 Chapter 1<br />

The Netherl<strong>and</strong>s; The University of Oxford, UK; <strong>and</strong>, more recently, Seattle<br />

Biomed, USA [84].<br />

Thesis outline<br />

The current thesis aims at exploring the induction of <strong>protection</strong> <strong>against</strong><br />

Plasmodium falciparum malaria in humans with specific attention for three<br />

research questions:<br />

What is the safety <strong>and</strong> immunogenicity of malaria vaccine c<strong>and</strong>idate Apical<br />

Membrane Antigen 1 (AMA1) in humans?<br />

Apical Membrane Antigen 1 is a promising vaccine c<strong>and</strong>idate. It has been<br />

produced as a recombinant protein, covering amino acids 25-545, in Pichia<br />

pastoris <strong>and</strong> has shown to be immunogenic with satisfactory<br />

pharmacotoxicology in animal models. As with most asexual erythrocytic stage<br />

antigens, AMA1 is highly polymorphic <strong>and</strong> relies on the induction of antibodies<br />

for conveying <strong>protection</strong>. We will address the following objectives in its first<br />

phase of clinical development:<br />

- to determine the safety profile of PfAMA1 FVO (25-545) when<br />

administered to humans in two different doses with three different<br />

adjuvants (Chapter 2)<br />

- to determine the quantity <strong>and</strong> quality of antibodies induced by PfAMA1<br />

FVO (25-545) in humans (Chapter 2,3,4)<br />

How can we optimize controlled human malaria <strong>infection</strong>s in order to<br />

maximize safety <strong>and</strong> scientific information?<br />

Controlled human malaria <strong>infection</strong>s (CHMI) are currently used to assess<br />

preliminary efficacy of pre-erythrocytic vaccine c<strong>and</strong>idates in a limited number<br />

of institutions worldwide. Because controlled <strong>infection</strong>s seem an efficient tool<br />

for investigating malaria immunology <strong>and</strong> c<strong>and</strong>idate vaccine efficacy, we will<br />

investigate how to optimize its protocol. We will explore the comparability <strong>and</strong><br />

power of the current model with the following specific objectives:<br />

- to identify parameters influencing the comparability of CHMI safety <strong>and</strong><br />

parasitological outcome in different institutions (Chapter 5)


Introduction 21<br />

- to identify parameters influencing the power of CHMI to detect vaccine<br />

efficacy of pre-erythrocytic <strong>and</strong> erythrocytic malaria vaccines (Chapter<br />

6)<br />

We next will address two important aspects of improvement to the CHMI model:<br />

- to increase the portfolio of Pf strains that can be used for <strong>infection</strong><br />

(Chapter 7)<br />

- to investigate the administration of parasites by needle (Chapter 8)<br />

Can <strong>protection</strong> to malaria in humans be efficiently induced by inoculation of<br />

live whole sporozoites?<br />

The inoculation of irradiated sporozoites by mosquito bite has shown to induce<br />

full <strong>protection</strong> in humans, albeit inefficiently <strong>and</strong> with limited longevity. We will<br />

explore whether viable intact sporozoites induce <strong>protection</strong> more efficiently,<br />

eventually aiming to create a model for malaria immunity. The administration of<br />

chloroquine prophylaxis will prevent clinical disease whilst the exposure to<br />

viable sporozoites allows exposure of parasite antigen. We address the following<br />

specific objectives:<br />

- to induce <strong>protection</strong> to Pf malaria in malaria naïve volunteers by<br />

exposure to parasites whilst taking chloroquine prophylaxis (Chapter 9)<br />

- to investigate the longevity of induced <strong>protection</strong> (Chapter 10)


22 Chapter 1<br />

References<br />

1. World Malaria Report, 2011. (http://www.who.int/malaria)<br />

2. Gallup JL, Sachs JD. The economic burden of malaria. Am J Trop Med Hyg 2001;<br />

64:85-96.<br />

3. Desai M, ter Kuile FO, Nosten F et al. Epidemiology <strong>and</strong> burden of malaria in<br />

pregnancy. Lancet Infect Dis 2007; 7:93-104.<br />

4. Fauci AS, Braunwald E, Kasper DL et al. Harrison's Internal Medicine. 17 ed.<br />

2008.<br />

5. Millennium Development Goals, 2012. (www.un.org/millenniumgoals)<br />

6. WHO guidelines for the treatment of malaria, 2011.<br />

(http://www.who.int/malaria/publications/atoz/9789241547925/en/index.htm<br />

l)<br />

7. Mendis K, Rietveld A, Warsame M, Bosman A, Greenwood B, Wernsdorfer WH.<br />

From malaria control to eradication: The WHO perspective. Trop Med Int Health<br />

2009; 14:802-809.<br />

8. World Malaria Report, 2011. (http://www.who.int/malaria)<br />

9. Alonso PL, Brown G, Arevalo-Herrera M et al. A research agenda to underpin<br />

malaria eradication. PLoS Med 2011; 8:e1000406.<br />

10. Malaria Vaccine Technology Roadmap, 2006.<br />

(http://www.malariavaccine.org/files/Malaria_Vaccine_TRM_Final_000.pdf)<br />

11. Crompton PD, Pierce SK, Miller LH. Advances <strong>and</strong> challenges in malaria vaccine<br />

development. J Clin Invest 2010; 120:4168-4178.<br />

12. Sauerwein RW, Roestenberg M, Moorthy VS. <strong>Experimental</strong> human challenge<br />

<strong>infection</strong>s can accelerate clinical malaria vaccine development. Nat Rev<br />

Immunol 2011; 11:57-64.<br />

13. Greenwood BM, Targett GA. Malaria vaccines <strong>and</strong> the new malaria agenda. Clin<br />

Microbiol Infect 2011; 17:1600-1607.<br />

14. Wang R, Doolan DL, Le TP et al. Induction of antigen-specific cytotoxic T<br />

lymphocytes in humans by a malaria DNA vaccine. Science 1998; 282:476-480.<br />

15. Hill AV, Reyes-S<strong>and</strong>oval A, O'Hara G et al. Prime-boost vectored malaria<br />

vaccines: progress <strong>and</strong> prospects. Hum Vaccin 2010; 6:78-83.<br />

16. WHO Malaria Vaccine Rainbow Table, 2011.<br />

(http://www.who.int/vaccine_research/links/Rainbow/en/index.html)<br />

17. Mazier D, Goma J, Pied S et al. Hepatic phase of malaria: a crucial role as "gobetween"<br />

with other stages. Bull World Health Organ 1990; 68 Suppl:126-131.<br />

18. Krzych U, Lyon JA, Jareed T et al. T lymphocytes from volunteers immunized<br />

with irradiated Plasmodium falciparum sporozoites recognize liver <strong>and</strong> blood<br />

stage malaria antigens. J Immunol 1995; 155:4072-4077.<br />

19. Belnoue E, Voza T, Costa FT et al. Vaccination with live Plasmodium yoelii blood<br />

stage parasites under chloroquine cover induces cross-stage immunity <strong>against</strong><br />

malaria liver stage. J Immunol 2008; 181:8552-8558.


Introduction 23<br />

20. Nussenzweig RS, V<strong>and</strong>erberg J, Most H, Orton C. Protective immunity produced<br />

by the injection of x-irradiated sporozoites of plasmodium berghei. Nature<br />

1967; 216:160-162.<br />

21. Nussenzweig V, Nussenzweig RS. Development of a sporozoite malaria vaccine.<br />

Am J Trop Med Hyg 1986; 35:678-688.<br />

22. Alonso PL, Sacarlal J, Aponte JJ et al. Efficacy of the RTS,S/AS02A vaccine <strong>against</strong><br />

Plasmodium falciparum <strong>infection</strong> <strong>and</strong> disease in young African children:<br />

r<strong>and</strong>omised controlled trial. Lancet 2004; 364:1411-1420.<br />

23. Alonso PL, Sacarlal J, Aponte JJ et al. Duration of <strong>protection</strong> with RTS,S/AS02A<br />

malaria vaccine in prevention of Plasmodium falciparum disease in Mozambican<br />

children: single-blind extended follow-up of a r<strong>and</strong>omised controlled trial.<br />

Lancet 2005; 366:2012-2018.<br />

24. Sacarlal J, Aide P, Aponte JJ et al. Long-term safety <strong>and</strong> efficacy of the<br />

RTS,S/AS02A malaria vaccine in Mozambican children. J Infect Dis 2009;<br />

200:329-336.<br />

25. Bejon P, Lusingu J, Olotu A et al. Efficacy of RTS,S/AS01E vaccine <strong>against</strong> malaria<br />

in children 5 to 17 months of age. N Engl J Med 2008; 359:2521-2532.<br />

26. Abdulla S, Oberholzer R, Juma O et al. Safety <strong>and</strong> immunogenicity of<br />

RTS,S/AS02D malaria vaccine in infants. N Engl J Med 2008; 359:2533-2544.<br />

27. Agn<strong>and</strong>ji ST, Lell B, Soulanoudjingar SS et al. First results of phase 3 trial of<br />

RTS,S/AS01 malaria vaccine in African children. N Engl J Med 2011; 365:1863-<br />

1875.<br />

28. Luty AJ, Lell B, Schmidt-Ott R et al. Interferon-gamma responses are associated<br />

with resistance to re<strong>infection</strong> with Plasmodium falciparum in young African<br />

children. J Infect Dis 1999; 179:980-988.<br />

29. John CC, T<strong>and</strong>e AJ, Moormann AM et al. Antibodies to pre-erythrocytic<br />

Plasmodium falciparum antigens <strong>and</strong> risk of clinical malaria in Kenyan children. J<br />

Infect Dis 2008; 197:519-526.<br />

30. Pichyangkul S, Kum-Arb U, Yongvanitchit K et al. Preclinical evaluation of the<br />

safety <strong>and</strong> immunogenicity of a vaccine consisting of Plasmodium falciparum<br />

liver-stage antigen 1 with adjuvant AS01B administered alone or concurrently<br />

with the RTS,S/AS01B vaccine in rhesus primates. Infect Immun 2008; 76:229-<br />

238.<br />

31. Cummings JF, Spring MD, Schwenk RJ et al. Recombinant Liver Stage Antigen-1<br />

(LSA-1) formulated with AS01 or AS02 is safe, elicits high titer antibody <strong>and</strong><br />

induces IFN-gamma/IL-2 CD4+ T cells but does not protect <strong>against</strong> experimental<br />

Plasmodium falciparum <strong>infection</strong>. Vaccine 2010; 28:5135-5144.<br />

32. McConkey SJ, Reece WH, Moorthy VS et al. Enhanced T-cell immunogenicity of<br />

plasmid DNA vaccines boosted by recombinant modified vaccinia virus Ankara<br />

in humans. Nat Med 2003; 9:729-735.<br />

33. Moorthy VS, Imoukhuede EB, Milligan P et al. A r<strong>and</strong>omised, double-blind,<br />

controlled vaccine efficacy trial of DNA/MVA ME-TRAP <strong>against</strong> malaria <strong>infection</strong><br />

in Gambian adults. PLoS Med 2004; 1:e33.


24 Chapter 1<br />

34. Bejon P, Mwacharo J, Kai O et al. A phase 2b r<strong>and</strong>omised trial of the c<strong>and</strong>idate<br />

malaria vaccines FP9 ME-TRAP <strong>and</strong> MVA ME-TRAP among children in Kenya.<br />

PLoS Clin Trials 2006; 1:e29.<br />

35. Hoffman SL, Goh LM, Luke TC et al. Protection of humans <strong>against</strong> malaria by<br />

immunization with radiation-attenuated Plasmodium falciparum sporozoites. J<br />

Infect Dis 2002; 185:1155-1164.<br />

36. Hoffman SL, Billingsley PF, James E et al. Development of a metabolically active,<br />

non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria.<br />

Hum Vaccin 2010; 6:97-106.<br />

37. Ellis RD, Sagara I, Doumbo O, Wu Y. Blood stage vaccines for Plasmodium<br />

falciparum: current status <strong>and</strong> the way forward. Hum Vaccin 2010; 6:627-634.<br />

38. Narum DL, Ogun SA, Thomas AW, Holder AA. Immunization with parasitederived<br />

apical membrane antigen 1 or passive immunization with a specific<br />

monoclonal antibody protects BALB/c mice <strong>against</strong> lethal Plasmodium yoelii<br />

yoelii YM blood-stage <strong>infection</strong>. Infect Immun 2000; 68:2899-2906.<br />

39. Dutta S, Sullivan JS, Grady KK et al. High antibody titer <strong>against</strong> apical membrane<br />

antigen-1 is required to protect <strong>against</strong> malaria in the Aotus model. PLoS One<br />

2009; 4:e8138.<br />

40. Crewther PE, Matthew ML, Flegg RH, Anders RF. Protective immune responses<br />

to apical membrane antigen 1 of Plasmodium chabaudi involve recognition of<br />

strain-specific epitopes. Infect Immun 1996; 64:3310-3317.<br />

41. Remarque EJ, Faber BW, Kocken CH, Thomas AW. Apical membrane antigen 1: a<br />

malaria vaccine c<strong>and</strong>idate in review. Trends Parasitol 2008; 24:74-84.<br />

42. Silvie O, Franetich JF, Charrin S et al. A role for apical membrane antigen 1<br />

during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol<br />

Chem 2004; 279:9490-9496.<br />

43. Takala SL, Coulibaly D, Thera MA et al. Extreme polymorphism in a vaccine<br />

antigen <strong>and</strong> risk of clinical malaria: implications for vaccine development. Sci<br />

Transl Med 2009; 1:2ra5.<br />

44. Scherf A, Lopez-Rubio JJ, Riviere L. Antigenic variation in Plasmodium<br />

falciparum. Annu Rev Microbiol 2008; 62:445-470.<br />

45. Jensen AT, Magistrado P, Sharp S et al. Plasmodium falciparum associated with<br />

severe childhood malaria preferentially expresses PfEMP1 encoded by group A<br />

var genes. J Exp Med 2004; 199:1179-1190.<br />

46. Kraemer SM, Smith JD. Evidence for the importance of genetic structuring to<br />

the structural <strong>and</strong> functional specialization of the Plasmodium falciparum var<br />

gene family. Mol Microbiol 2003; 50:1527-1538.<br />

47. Umbers AJ, Aitken EH, Rogerson SJ. Malaria in pregnancy: small babies, big<br />

problem. Trends Parasitol 2011; 27:168-175.<br />

48. Hviid L. The role of Plasmodium falciparum variant surface antigens in<br />

protective immunity <strong>and</strong> vaccine development. Hum Vaccin 2010; 6:84-89.<br />

49. Bousema T, Roeffen W, Meijerink H et al. The dynamics of naturally acquired<br />

immune responses to Plasmodium falciparum sexual stage antigens Pfs230 &<br />

Pfs48/45 in a low endemic area in Tanzania. PLoS One 2010; 5:e14114.


Introduction 25<br />

50. Ouedraogo AL, Roeffen W, Luty AJ et al. Naturally acquired immune responses<br />

to Plasmodium falciparum sexual stage antigens Pfs48/45 <strong>and</strong> Pfs230 in an area<br />

of seasonal transmission. Infect Immun 2011; 79:4957-4964.<br />

51. Saul A. Mosquito stage, transmission blocking vaccines for malaria. Curr Opin<br />

Infect Dis 2007; 20:476-481.<br />

52. Wu Y, Ellis RD, Shaffer D et al. Phase 1 trial of malaria transmission blocking<br />

vaccine c<strong>and</strong>idates Pfs25 <strong>and</strong> Pvs25 formulated with montanide ISA 51. PLoS<br />

One 2008; 3:e2636.<br />

53. Marsh K, Kinyanjui S. Immune effector mechanisms in malaria. Parasite<br />

Immunol 2006; 28:51-60.<br />

54. Gupta S, Snow RW, Donnelly CA, Marsh K, Newbold C. Immunity to noncerebral<br />

severe malaria is acquired after one or two <strong>infection</strong>s. Nat Med 1999;<br />

5:340-343.<br />

55. Doolan DL, Dobano C, Baird JK. Acquired immunity to malaria. Clin Microbiol<br />

Rev 2009; 22:13-36, Table.<br />

56. Nussenzweig RS, Nussenzweig V. Antisporozoite vaccine for malaria:<br />

experimental basis <strong>and</strong> current status. Rev Infect Dis 1989; 11 Suppl 3:S579-<br />

S585.<br />

57. Tsuji M, Zavala F. T cells as mediators of protective immunity <strong>against</strong> liver<br />

stages of Plasmodium. Trends Parasitol 2003; 19:88-93.<br />

58. Doolan DL, Hoffman SL. The complexity of protective immunity <strong>against</strong> liverstage<br />

malaria. J Immunol 2000; 165:1453-1462.<br />

59. McCall MB, Sauerwein RW. Interferon-gamma--central mediator of protective<br />

immune responses <strong>against</strong> the pre-erythrocytic <strong>and</strong> blood stage of malaria. J<br />

Leukoc Biol 2010; 88:1131-1143.<br />

60. Doolan DL, Martinez-Alier N. Immune response to pre-erythrocytic stages of<br />

malaria parasites. Curr Mol Med 2006; 6:169-185.<br />

61. Yazdani SS, Mukherjee P, Chauhan VS, Chitnis CE. Immune responses to asexual<br />

blood-stages of malaria parasites. Curr Mol Med 2006; 6:187-203.<br />

62. Good MF, Xu H, Wykes M, Engwerda CR. Development <strong>and</strong> regulation of cellmediated<br />

immune responses to the blood stages of malaria: implications for<br />

vaccine research. Annu Rev Immunol 2005; 23:69-99.<br />

63. Cohen S, McGregor IA, Carrington S. Gamma-globulin <strong>and</strong> acquired immunity to<br />

human malaria. Nature 1961; 192:733-737.<br />

64. Sabchareon A, Burnouf T, Ouattara D et al. Parasitologic <strong>and</strong> clinical human<br />

response to immunoglobulin administration in falciparum malaria. Am J Trop<br />

Med Hyg 1991; 45:297-308.<br />

65. Kennedy MC, Wang J, Zhang Y et al. In vitro studies with recombinant<br />

Plasmodium falciparum apical membrane antigen 1 (AMA1): production <strong>and</strong><br />

activity of an AMA1 vaccine <strong>and</strong> generation of a multiallelic response. Infect<br />

Immun 2002; 70:6948-6960.<br />

66. Bouharoun-Tayoun H, Attanath P, Sabchareon A, Chongsuphajaisiddhi T,<br />

Druilhe P. Antibodies that protect humans <strong>against</strong> Plasmodium falciparum<br />

blood stages do not on their own inhibit parasite growth <strong>and</strong> invasion in vitro,<br />

but act in cooperation with monocytes. J Exp Med 1990; 172:1633-1641.


26 Chapter 1<br />

67. Covell G, Nicol WD. Clinical, chemotherapeutic <strong>and</strong> immunological studies on<br />

induced malaria. Br Med Bull 1951; 8:51-55.<br />

68. Campbell CC, Collins WE, Nguyen-Dinh P, Barber A, Broderson JR. Plasmodium<br />

falciparum gametocytes from culture in vitro develop to sporozoites that are<br />

infectious to primates. Science 1982; 217:1048-1050.<br />

69. Ponnudurai T, Lensen AH, van Gemert GJ, Bensink MP, Bolmer M, Meuwissen<br />

JH. Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes.<br />

Parasitology 1989; 98 Pt 2:165-173.<br />

70. Ifediba T, V<strong>and</strong>erberg JP. Complete in vitro maturation of Plasmodium<br />

falciparum gametocytes. Nature 1981; 294:364-366.<br />

71. Church LW, Le TP, Bryan JP et al. Clinical manifestations of Plasmodium<br />

falciparum malaria experimentally induced by mosquito challenge. J Infect Dis<br />

1997; 175:915-920.<br />

72. Epstein JE, Rao S, Williams F et al. Safety <strong>and</strong> clinical outcome of experimental<br />

challenge of human volunteers with Plasmodium falciparum-infected<br />

mosquitoes: an update. J Infect Dis 2007; 196:145-154.<br />

73. Verhage DF, Telgt DS, Bousema JT et al. Clinical outcome of experimental<br />

human malaria induced by Plasmodium falciparum-infected mosquitoes. Neth J<br />

Med 2005; 63:52-58.<br />

74. Hermsen CC, Telgt DS, Linders EH et al. Detection of Plasmodium falciparum<br />

malaria parasites in vivo by real-time quantitative PCR. Mol Biochem Parasitol<br />

2001; 118:247-251.<br />

75. Hermsen CC, de Vlas SJ, van Gemert GJ, Telgt DS, Verhage DF, Sauerwein RW.<br />

Testing vaccines in human experimental malaria: statistical analysis of<br />

parasitemia measured by a quantitative real-time polymerase chain reaction.<br />

Am J Trop Med Hyg 2004; 71:196-201.<br />

76. Bejon P, Andrews L, Andersen RF et al. Calculation of liver-to-blood inocula,<br />

parasite growth rates, <strong>and</strong> preerythrocytic vaccine efficacy, from serial<br />

quantitative polymerase chain reaction studies of volunteers challenged with<br />

malaria sporozoites. J Infect Dis 2005; 191:619-626.<br />

77. Spring MD, Cummings JF, Ockenhouse CF et al. Phase 1/2a study of the malaria<br />

vaccine c<strong>and</strong>idate apical membrane antigen-1 (AMA-1) administered in<br />

adjuvant system AS01B or AS02A. PLoS One 2009; 4:e5254.<br />

78. Thompson FM, Porter DW, Okitsu SL et al. Evidence of blood stage efficacy with<br />

a virosomal malaria vaccine in a phase IIa clinical trial. PLoS One 2008; 3:e1493.<br />

79. Genton B, D'Acremont V, Lurati-Ruiz F et al. R<strong>and</strong>omized double-blind<br />

controlled Phase I/IIa trial to assess the efficacy of malaria vaccine PfCS102 to<br />

protect <strong>against</strong> challenge with P. falciparum. Vaccine 2010; 28:6573-6580.<br />

80. Kester KE, McKinney DA, Tornieporth N et al. Efficacy of recombinant<br />

circumsporozoite protein vaccine regimens <strong>against</strong> experimental Plasmodium<br />

falciparum malaria. J Infect Dis 2001; 183:640-647.<br />

81. Casares S, Brumeanu TD, Richie TL. The RTS,S malaria vaccine. Vaccine 2010;<br />

28:4880-4894.<br />

82. Kester KE, Cummings JF, Ockenhouse CF et al. Phase 2a trial of 0, 1, <strong>and</strong> 3<br />

month <strong>and</strong> 0, 7, <strong>and</strong> 28 day immunization schedules of malaria vaccine


Introduction 27<br />

RTS,S/AS02 in malaria-naive adults at the Walter Reed Army Institute of<br />

Research. Vaccine 2008; 26:2191-2202.<br />

83. Bojang KA, Milligan PJ, Pinder M et al. Efficacy of RTS,S/AS02 malaria vaccine<br />

<strong>against</strong> Plasmodium falciparum <strong>infection</strong> in semi-immune adult men in The<br />

Gambia: a r<strong>and</strong>omised trial. Lancet 2001; 358:1927-1934.<br />

84. Moorthy VS, Diggs C, Ferro S et al. Report of a consultation on the optimization<br />

of clinical challenge trials for evaluation of c<strong>and</strong>idate blood stage malaria<br />

vaccines, 18-19 March 2009, Bethesda, MD, USA. Vaccine 2009; 27:5719-5725.


Section 1<br />

Apical Membrane Antigen 1


Chapter 2<br />

Safety <strong>and</strong> Immunogenicity of a<br />

Recombinant Plasmodium falciparum<br />

AMA1 Malaria vaccine Adjuvanted with<br />

Alhydrogel TM , Montanide ISA 720 or AS02.<br />

Meta Roestenberg* 1 , Ed Remarque 2 , Erik de Jonge 1 , Rob Hermsen 1 , Hildur<br />

Blythman 3 , Odile Leroy 3 , Egeruan Imoukhuede 3 , Soren Jepsen 3 , Opokua Ofori-<br />

Anyinam 4 , Bart Faber 2 , Clemens H. M. Kocken 2 , Mir<strong>and</strong>a Arnold 2 , Vanessa<br />

Walraven 2 , Karina Teelen 1 , Will Roeffen 1 , Quirijn de Mast 1 , W. Ripley Ballou 4,5 ,<br />

Joe Cohen 4 , Marie Claude Dubois 4 , Stéphane Ascarateil 6 , Andre van der Ven 1 ,<br />

Alan Thomas 2 , Robert Sauerwein 1<br />

1 Radboud University Nijmegen Medical Centre, Nijmegen, The Netherl<strong>and</strong>s<br />

2 Biomedical Primate Research Centre, Rijswijk, The Netherl<strong>and</strong>s<br />

3 European Malaria Vaccine Initiative, Copenhagen, Denmark<br />

4 GlaxoSmithKline Biologicals, Rixensart, Belgium<br />

5 Present address: Bill <strong>and</strong> Melinda Gates Foundation, Seattle, USA<br />

6 SEPPIC, Paris, France<br />

PLoS ONE 2008; 3:e3960


Chapter 2 32<br />

Abstract<br />

Plasmodium falciparum Apical Membrane Antigen 1 (PfAMA1) is a c<strong>and</strong>idate<br />

vaccine antigen expressed by merozoites <strong>and</strong> sporozoites. It plays a key role in<br />

red blood cell <strong>and</strong> hepatocyte invasion that can be blocked by antibodies.<br />

We assessed the safety <strong>and</strong> immunogenicity of recombinant PfAMA1 in a doseescalating,<br />

phase Ia trial. PfAMA1 FVO strain, produced in Pichia pastoris, was<br />

reconstituted at 10 µg <strong>and</strong> 50 µg doses with three different adjuvants,<br />

Alhydrogel, Montanide ISA720 <strong>and</strong> AS02 Adjuvant System. Six r<strong>and</strong>omised<br />

groups of healthy male volunteers, 8-10 volunteers each, were scheduled to<br />

receive three immunisations at 4-week intervals. Safety <strong>and</strong> immunogenicity<br />

data were collected over one year.<br />

Transient pain was the predominant injection site reaction (80-100%).<br />

Induration occurred in the Montanide 50 µg group, resulting in a sterile abscess<br />

in two volunteers. Systemic adverse events occurred mainly in the AS02 groups<br />

lasting for 1-2 days. Erythema was observed in 22% of Montanide <strong>and</strong> 59% of<br />

AS02 group volunteers. After the second dose, six volunteers in the AS02 group<br />

<strong>and</strong> one in the Montanide group who reported grade 3 erythema (>50mm) were<br />

withdrawn as they met the stopping criteria. All adverse events resolved. There<br />

were no vaccine-related serious adverse events.<br />

Humoral responses were highest in the AS02 groups. Antibodies showed activity<br />

in an in vitro growth inhibition assay up to 80%. Upon stimulation with the<br />

vaccine, peripheral mononuclear cells from all groups proliferated <strong>and</strong> secreted<br />

IFNγ <strong>and</strong> IL-5 cytokines.<br />

In conclusion, all formulations showed distinct reactogenicity profiles. All<br />

formulations with PfAMA1 were immunogenic <strong>and</strong> induced functional<br />

antibodies.


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

Introduction<br />

In sub-Saharan Africa the burden of death <strong>and</strong> disease from Plasmodium<br />

falciparum malaria is particularly severe. To date, there are no approved<br />

vaccines to help reduce this burden, although a number of c<strong>and</strong>idate vaccines<br />

have been put forward. The majority of the c<strong>and</strong>idates target the preerythrocytic<br />

circumsporozoite protein (CSP) <strong>and</strong> the merozoite proteins<br />

Merozoite Surface Protein 1 (MSP1) <strong>and</strong> Apical Membrane Antigen 1 (AMA1)[1].<br />

The RTS,S c<strong>and</strong>idate vaccine has shown efficacy in infants <strong>and</strong> children [2,3] <strong>and</strong><br />

a phase III clinical trial is planned. The MSP1 <strong>and</strong> AMA1 c<strong>and</strong>idate vaccines are in<br />

early stage clinical development <strong>and</strong> efficacy trials will provide information to<br />

determine whether these antigens are suitable targets, <strong>and</strong> whether they can be<br />

deployed singly or as components of a multivalent malaria vaccine.<br />

Following an infected mosquito bite, P. falciparum sporozoites migrate to<br />

hepatocytes, each developing over a period of a week to release several<br />

thous<strong>and</strong> merozoites. These initiate cyclical asexual blood stage development,<br />

producing merozoites that invade erythrocytes. AMA1 is an integral membrane<br />

protein of merozoites <strong>and</strong> sporozoites <strong>and</strong> has a central role in parasite invasion<br />

of erythrocytes <strong>and</strong> potentially hepatocytes that can be inhibited by anti-AMA1<br />

antibody [4–6]. In merozoites, AMA1 is synthesised as an 83kDa molecule<br />

originally localised to the microneme. Around the time of merozoite release <strong>and</strong><br />

the subsequent rapid erythrocyte invasion, the protein is N-terminally cleaved to<br />

a 66 kDa form. This translocates to the merozoite surface <strong>and</strong> undergoes<br />

secondary proteolytic processing, shedding soluble fragments (44 or 48kDa) [7].<br />

Immunisation with AMA1 can provide <strong>protection</strong> <strong>against</strong> <strong>infection</strong> in<br />

experimental animal models, <strong>and</strong> can induce antibodies that show functionality<br />

in in vitro growth inhibition assays (GIA). However, AMA1 is polymorphic <strong>and</strong><br />

immune responses have varying degrees of strain specificity <strong>and</strong> growth<br />

inhibition [8].<br />

Previous Phase I trials have shown that growth inhibitory antibodies can be<br />

induced by immunisation with PfAMA1 [9,10], but immunogenicity varied<br />

depending on the vaccine formulation. In particular, the choice of adjuvant has a<br />

major effect on the safety, stability, immunogenicity <strong>and</strong>, presumably, eventual<br />

efficacy of a vaccine [11]. Adjuvants can be tools that channel the immune<br />

response to generate high levels of the desired type of long-lived immunity.<br />

33


34 Chapter 2<br />

Alhydrogel, an aluminium salt, is the most widely used adjuvant in licensed<br />

human vaccines <strong>and</strong> is therefore used as a st<strong>and</strong>ard to compare other adjuvants.<br />

Unfortunately, in combination with malaria antigens, it has generally induced<br />

poor responses [10,12–14]. Montanide ISA 720, a squalene based water-in-oil<br />

adjuvant formulation has shown promising results in previous malaria vaccine<br />

trials [15–18], possibly due to the slow-release capacity of the inert water-in-oil<br />

emulsion <strong>and</strong> immune stimulating effects of its components [19]. AS02, a<br />

proprietary Adjuvant System from GlaxoSmithKline Biologicals based on an oilin-water<br />

formulation, contains 50 µg each of the immunostimulants<br />

monophosphoryl lipid A (MPL) <strong>and</strong> Quillaja saponaria 21 (QS21) [20]. It has been<br />

used to adjuvant the RTS,S malaria c<strong>and</strong>idate vaccine that targets CSP. To date,<br />

this c<strong>and</strong>idate is the only malaria vaccine that has induced <strong>protection</strong> in adults,<br />

children <strong>and</strong> infants in natural field trials [20–23]. When combined with<br />

Alhydrogel, RTS,S did not convey <strong>protection</strong> in a combined phase I/IIa trial<br />

[24]. AS02 is capable of eliciting high antibody titres along with strong cellmediated<br />

immunity [25], both of which are believed to contribute to the efficacy<br />

of the RTS,S c<strong>and</strong>idate vaccine [26].<br />

Because of their central role in vaccine formulation, the development of<br />

adjuvants <strong>and</strong> delivery systems have become increasingly important. This study<br />

aims at comparing the safety <strong>and</strong> immunogenicity of PfAMA1 in two dosages<br />

formulated with three different adjuvants in a phase Ia trial.<br />

Materials <strong>and</strong> Methods<br />

Vaccine preparation<br />

Clinical grade PfAMA1-FVO[25-545] was developed [27] <strong>and</strong> produced [28] as<br />

previously reported. In brief, FVO strain PfAMA1 was codon adapted to<br />

expression in the methylotrophic yeast Pichia pastoris. Glycosylation sites were<br />

conservatively mutated, <strong>and</strong> the ectodomain comprising amino acids 25-545 was<br />

expressed. PfAMA1-FVO[25-545] preparation was manufactured <strong>and</strong> lyophilised<br />

according to current good manufacturing practice in multidose vials containing<br />

either 120 µg (44 µg EDTA, 180 µg sucrose <strong>and</strong> 120 µg NaHCO3, lot B) or 62.5 µg<br />

(23 µg EDTA, 25 mg saccharose, 226 µg K2HPO4 <strong>and</strong> 187 µg NaH2PO4, lot C) of<br />

AMA1 that were stored between -18°C <strong>and</strong> -30 °C <strong>and</strong> between +2 <strong>and</strong> +8°C<br />

respectively. Quality control <strong>and</strong> stability data are described by Faber et al. [28].<br />

Reconstitution <strong>and</strong> mixing of vaccine with adjuvant was performed under sterile<br />

conditions under responsibility of the hospital pharmacist.


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

PfAMA1 vaccine at 50 µg (high dose) <strong>and</strong> 10 µg (low dose) PfAMA1 per<br />

injection (0.5 ml) was formulated with three different adjuvants <strong>and</strong>, after<br />

preparation, was kept at a constant temperature of +4°C for a maximum of six<br />

hours until injection. For the Alhydrogel formulation, 1.2 ml aluminium<br />

hydroxide suspension at 2 mg/ml (Statens Serum Institut (SSI), Copenhagen,<br />

Denmark) was added to the 120 µg PfAMA1 vial (lot B) to obtain a high dose (50<br />

µg in 0.5 ml) <strong>and</strong> 6 ml was added to obtain a low dose (10 µg in 0.5 ml)<br />

formulation. The resulting amount of aluminium in each vaccine was 0.5 mg.<br />

Stability studies confirmed adsorption of 99.9% of the antigen to the aluminium.<br />

Montanide formulations were prepared by dissolving the contents of the 120 µg<br />

PfAMA1 vial (lot B) in sterile phosphate buffered saline (145mM NaCl, 5mM<br />

Phosphate, pH7.4), 0.32 ml for the high dose <strong>and</strong> 1.6 ml for the low dose<br />

formulation. Montanide ISA 720 (SEPPIC, Paris, France) was subsequently added,<br />

0.88 ml for the high dose to obtain 1.2 ml of formulation (50 µg PfAMA1 in 0.5<br />

ml) <strong>and</strong> 4.4 ml for the low dose to obtain 6 ml of formulation of which five 10 µg<br />

PfAMA1 in 0.5 ml doses could be prepared. The suspension was prepared by<br />

manually pushing through a 22 gauge syringe coupling piece (3038068 Omnilabo<br />

International, Breda, The Netherl<strong>and</strong>s) at +20°C for twenty up <strong>and</strong> down strokes.<br />

The suspension was confirmed to be homogeneous <strong>and</strong> reached a median<br />

droplet size of approximately 1.5 µm (SD 0.17 µm) by particle size<br />

measurements with the Malvern Mastersizer S by SEPPIC.<br />

For the AS02 formulation, the contents of one vial of lyophilized PfAMA1<br />

containing 62.5 µg of antigen (lot C) was mixed by gentle shaking with AS02<br />

(approximately 0.6 ml) [29]. A 0.5 ml dose contained approximately 50 µg AMA-<br />

1 in 500 µl AS02 (high dose). For low dose preparations (10 µg) five times more<br />

AS02 adjuvant was added to the 62.5 µg vial of AMA1, from which five 0.5 ml<br />

low vaccine doses could be obtained.<br />

Study design<br />

This study was designed as a dose-escalating phase Ia trial to assess the safety<br />

<strong>and</strong> immunogenicity of two dosages of PfAMA1 with three different adjuvants.<br />

Volunteers were thus r<strong>and</strong>omised into six different groups, each of which was<br />

aimed to constitute of a limited number of 10 volunteers for safety reasons.<br />

R<strong>and</strong>omisation was performed by an external statistician in six blocks through a<br />

computer program. Block r<strong>and</strong>omization were used to ensure equal distribution<br />

of adjuvants among the immunisation groups. There was no stratification for sex<br />

<strong>and</strong>/or age. The r<strong>and</strong>omization list was provided to the pharmacy departments.<br />

35


36 Chapter 2<br />

The clinical investigators allocated the next available number on entry into the<br />

trial. The code was revealed to the researchers once recruitment, data<br />

collection, <strong>and</strong> laboratory analyses were complete. The immunisations were<br />

thus performed blind, so neither volunteers, nor investigator or laboratory<br />

personnel were aware of the adjuvant allocation. Because of the dose-escalating<br />

design, the trial could not be blinded for dose.<br />

For logistical reasons, the AS02 adjuvanted groups were immunised nine months<br />

after the Alhydrogel <strong>and</strong> Montanide groups, breaking the blind for this trial<br />

arm. A subsequent bias cannot formally be excluded but seems unlikely, since all<br />

trial procedures were identical. All immunisations were performed<br />

intramuscularly in the deltoid region of alternate arms at 0, 4 <strong>and</strong> 8 weeks.<br />

Participants<br />

We aimed to recruit 60 healthy, malaria naïve male volunteers, aged between<br />

18 to 45 years through advertisements at the Radboud University Nijmegen<br />

Medical Centre. Potential volunteers provided a medical history <strong>and</strong> a physical<br />

examination was conducted with routine laboratory tests consisting of full blood<br />

count, serum biochemistries <strong>and</strong> serologic assays for human immunodeficiency<br />

virus, hepatitis C <strong>and</strong> B virus. Volunteers were excluded from participation if<br />

they had any symptoms, signs or laboratory values suggestive of systemic illness,<br />

including renal, hepatic, cardiovascular, pulmonary, skin, immunodeficiency,<br />

psychiatric <strong>and</strong> other conditions, which could interfere with the interpretation of<br />

the study results or compromise the health of the volunteers, or received<br />

chronic medication, had a history of drug or alcohol abuse interfering with social<br />

function one year prior to enrolment, or a known hypersensitivity to any of the<br />

vaccine components. Additional reasons for exclusion were a history of malaria<br />

or residence in malaria endemic areas within the past six months, previous<br />

participation in a malaria vaccine trial or receiving vaccines other than the study<br />

vaccines. Furthermore, volunteers were not enrolled in any other clinical trial,<br />

<strong>and</strong> agreed to remain available to be closely monitored. All volunteers provided<br />

written informed consent. The study was approved by the Institutional Review<br />

Board (CMO Regio Arnhem-Nijmegen, 2005/015). The study was conducted in<br />

accordance with the Declaration of Helsinki principles for the conduct of clinical<br />

trials <strong>and</strong> the International Committee of Harmonization Good Clinical Practice<br />

Guidelines [30] <strong>and</strong> registered at www.clinicaltrials.gov (NCT00730782).


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

Assessment of safety<br />

Volunteers were observed for 30 minutes <strong>and</strong> evaluated on days 1, 3, 7 <strong>and</strong> 14<br />

after every immunisation. At each visit, local <strong>and</strong> systemic reactogenicity was<br />

assessed by a physician <strong>and</strong> findings recorded <strong>and</strong> scored as follows: grade 1,<br />

mild reaction (easily tolerated), grade 2, moderate reaction (interferes with<br />

normal activity), or grade 3, severe reaction (prevents normal activity). Redness,<br />

swelling <strong>and</strong> induration (according to Brighton collaboration definitions,<br />

www.brightoncollaboration.org) were measured with a ruler, <strong>and</strong> categorised<br />

according to the longest diameter as grade 1: 20 <strong>and</strong> 50mm. Temperature was measured with an oral thermometer; fever<br />

intensity was defined as grade 1 (37.5°C to 38°C), grade 2 (>38°C to 39°C) or<br />

grade 3 (>39°C). The following adverse events were solicited <strong>and</strong> recorded<br />

routinely during the 14 days after immunisation: injection site pain, redness <strong>and</strong><br />

swelling, systemic fatigue, fever, headache, malaise, myalgia, joint pain,<br />

gastrointestinal symptoms <strong>and</strong> contralateral local reactions.<br />

Blood samples<br />

Safety was also determined by serial laboratory evaluations of clinical chemistry<br />

<strong>and</strong> haematology on blood samples collected 7 <strong>and</strong> 28 days after immunisation.<br />

For evaluation of immunogenicity, blood was collected in Vacutainer CPT tubes<br />

(Becton <strong>and</strong> Dickinson) <strong>and</strong> processed within two hours after collection on<br />

immunisation days, one month after each immunisation <strong>and</strong> on Days 140 <strong>and</strong><br />

365. Plasma was collected after centrifugation (2000 g 15’) aliquoted <strong>and</strong> stored<br />

at –20°C for antibody analysis (ELISA, Immuno Fluorescence Assay (IFA) <strong>and</strong><br />

Growth Inhibition Assay (GIA)). Peripheral blood mononuclear cells (PBMC) were<br />

collected, washed in PBS (800 g, 10 min) <strong>and</strong> immediately used for assays<br />

(lymphocyte stimulation assay <strong>and</strong> ELISPOT).<br />

Measurement of anti-AMA1 antibodies by ELISA <strong>and</strong> IFA<br />

Antibody to PfAMA1 was measured using a st<strong>and</strong>ardized ELISA protocol. All<br />

procedures used Phosphate buffered saline (PBS) <strong>and</strong> for washing steps 0.05%<br />

Tween 20 (Sigma-Aldrich). Briefly, wells in 96-well polystyrene plates (NUNC<br />

Maxisorp, Sanbio), were coated overnight (100 µl, 0.5 µg/ml PfAMA1, 4°C),<br />

washed (3x), blocked (60 min, 3% BSA (Sigma-Aldrich)) <strong>and</strong> washed (3x) before<br />

addition of 100 µL from duplicate dilution series (diluted in PBS-Tween BSA, one<br />

hour, +37°C). After washing (3x) goat anti-human IgG alkaline phosphate (Perbio<br />

Science) diluted 1:1250 in 0.5% BSA, 0.05% Tween was added, (one hour, +37°C).<br />

Plates were washed <strong>and</strong> 100 μl of 1 mg/ml para-nitro-phenyl-phosphatase<br />

37


38 Chapter 2<br />

(Fluka, Sigma-Aldrich) substrate was added (30 minutes, room temperature). A<br />

human plasma pool from a malaria endemic area was used as reference positive<br />

control, whereas a plasma pool from eight healthy malaria-naive Dutch<br />

volunteers was used as a negative control. Optical density was measured at<br />

405nm. Variation between duplicates was set to a maximum of 15%.<br />

Measurements with a greater variation were repeated. The st<strong>and</strong>ard curve of<br />

human plasma pool from a malaria endemic area, defined to contain 400<br />

Arbitrary Units, was fitted to a four-parameter hyperbolic function, using the<br />

ADAMSEL program (E. Remarque, unpublished work). Using this st<strong>and</strong>ard curve,<br />

optical density from samples were converted to Arbitrary Units (AU). Test<br />

samples that did not fall within the linear part of the optical density range of the<br />

st<strong>and</strong>ard were tested at alternate dilutions.<br />

IFA was performed on cultured P. falciparum parasitized red blood cells. Ten<br />

well black slides (30-966-A black, Nutacon, The Netherl<strong>and</strong>s) were coated with a<br />

washed parasite suspension of 3 x 10 6 parasites/ml, air dried <strong>and</strong> kept at –80°C<br />

until used. FCR3 parasites, expressing an AMA1 protein with one amino-acid<br />

difference from the FVO parasites, <strong>and</strong> NF54 strain parasites, with 26 amino-acid<br />

difference in AMA1 protein were used to prepare slides.<br />

Based on antibody titres by ELISA on day 84, a representative sample of fifteen<br />

sera was selected for IFA, containing at least two samples from each adjuvant<br />

group <strong>and</strong> at least three samples with low, intermediate or high ELISA titres.<br />

Before use slides were brought to room temperature in an evacuated exicator.<br />

Plasma was diluted in PBS (1:40, 1:80, 1:160, 1:320, 1:640) <strong>and</strong> a final volume of<br />

20 µL was added to the wells <strong>and</strong> incubated for 0.5 hour, at room temperature.<br />

As for the ELISA protocol, the malaria-naïve blood bank donor plasma pool was<br />

used as a negative control <strong>and</strong> human malaria endemic plasma was used as a<br />

positive control. After washing (2x in PBS) <strong>and</strong> air drying, slide samples were<br />

incubated with rabbit anti-human Immunoglobin FITC (F0200, DAKO, Denmark)<br />

in 0.05% w.v Evans Blue (3169, Merck), PBS for 30 minutes at room<br />

temperature. Slides were washed twice <strong>and</strong> incubated for 15 minutes with DAPI<br />

(4’-6-Diamindino-2-phenylindole, 24653, Merck, Darmstadt, Germany), 5 µg/ml<br />

in PBS. After washing (2x in PBS) slides were mounted with Vectashield<br />

Mounting Medium (H-1000, Brunschwig, Amsterdam), covered with a deck-slide<br />

<strong>and</strong> read immediately by two independent blinded examiners. Examiners<br />

identified the highest dilution still showing a staining pattern above the<br />

background of pre-immunisation samples. Differences between examiners were<br />

never greater than one dilution <strong>and</strong> the mean of both dilutions was taken.


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

ELISPOT for IFNy <strong>and</strong> IL-5<br />

ELISPOT was performed according to manufacturer’s instructions (Becton <strong>and</strong><br />

Dickinson Elispot Set Human IFNγ or IL-5). In summary, plates provided in the set<br />

were coated with either IFNγ or IL-5 capture antibody (5 µg/ml, overnight, 4°C).<br />

After blocking with complete medium solution (RPMI 1640 (Invitrogen)<br />

containing 10% Fetal Bovine Serum (FBS Invitrogen, Breda, The Netherl<strong>and</strong>s), 1%<br />

Glutamax (Invitrogen), 1% Penicillin-Streptomycin (GIBCO-BRL, Invitrogen), 1%<br />

MEM) 100 µl of 105 PBMC suspension <strong>and</strong> 100 µl of PfAMA1 containing either<br />

60 µg, 12 µg or 2.4 µg was added per well. Positive controls were stimulated<br />

with Tetanus Toxoid 10 µg/ml (RIVM, Bilthoven, The Netherl<strong>and</strong>s) <strong>and</strong><br />

phytohaemagglutinin 5 µg/ml (PHA-L Sigma-Aldrich) end concentration.<br />

Negative controls were incubated with complete medium solution (mean<br />

SFC/10 5 cells 31±17 for IFNγ <strong>and</strong> 9±7 for IL-5). After incubation (40 hours, 37°C in<br />

humidified 5% CO2), biotinylated anti human IFNγ <strong>and</strong> IL-5 (0.25 µg/ml <strong>and</strong> 2<br />

µg/ml, respectively), containing 10% FBS was added (two hours at room<br />

temperature). Streptavidin-HRP was used as an enzyme conjugate. Detection<br />

was performed with the Becton <strong>and</strong> Dickinson AEC Substrate Reagent Set,<br />

according to manufacturer’s instruction. Spot-forming cell numbers were<br />

counted by ELISPOT reader (4 Microtitre Plate Reader, AELVIS, Sanquin,<br />

Amsterdam) <strong>and</strong> analysed by the ELISPOT Analysis Software Version 4.0<br />

(Sanquin, Amsterdam). All measurements were performed in triplo. Variation<br />

between triplicates was set to a maximum of 20%.<br />

Lymphocyte Stimulation Assay<br />

Lymphocyte stimulation assays were performed as described previously [31].<br />

Peripheral blood mononuclear cell suspension (PBMC) was diluted to 1 x 10 6<br />

PBMC per ml in Dulbecco’s MEM (DMEM) with Glutamax-I, 2 mM pyruvate <strong>and</strong><br />

high Glucose (GIBCO BRL, Invitrogen) supplemented with 10 mM HEPES buffer<br />

(GIBCO BRL, Invitrogen), 100 IU/mL Penicillin-Streptomycin (GIBCO BRL,<br />

Invitrogen), 100 µM non-essential amino acids (GIBCO BRL, Invitrogen) <strong>and</strong> 2.5%<br />

human AB serum (AB) (Bodinco BV, Alkmaar, The Netherl<strong>and</strong>s). 100 µl of PBMC<br />

was added to 100 µl PfAMA1 (30, 6 or 1.2 µg/ml in PBS) in 96 well Nunclon<br />

surface flat plates (Life Technology). Plates were incubated (six days, +37°C,<br />

humidified 0.5%CO2) before labelling (10 µl 3H thymidine, 0.25 uCi per well, 24<br />

hours) <strong>and</strong> harvested onto Wallac filter mats using the Wallac Beta plate<br />

harvester. Incorporated 3H-thimidine was determined using a Wallac Beta Plate<br />

counter. Stimulation indices (SI) were calculated relative to control wells to<br />

which no PfAMA1 had been added. PBMC were tested in parallel for their<br />

39


40 Chapter 2<br />

Figure 1. Study flow chart showing number of volunteers r<strong>and</strong>omised, withdrawn <strong>and</strong><br />

completing follow-up. Coding for adjuvant as follows: Alum = Alhydrogel, Mon =<br />

Montanide. Reasons for withdrawal are given: “rash” = allergic rash unrelated to study<br />

procedure, “erythema” = grade 3 injection site erythema leading to withdrawal, “other<br />

vac.” = concomitant Hepatitis B vaccination leading to exclusion.<br />

ability to be stimulated with Tetanus Toxoid (Purified Tetanus Toxoid 150 Lf/ml,<br />

RIVM, Bilthoven, The Netherl<strong>and</strong>s) <strong>and</strong> phytohaemagglutinin 5 µg/ml (PHA-L,<br />

Sigma-Aldrich).<br />

In vitro parasite growth inhibition<br />

Antibodies to be used for parasite inhibition assays were purified on protein A<br />

columns (Immunopure Plus Pierce, St Louis, MO, USA) using st<strong>and</strong>ard protocols,<br />

exchanged into RPMI 1640 using Amicon Ultra-15 concentrators (30 kDa cut-off,<br />

Millipore, Irel<strong>and</strong>), filter-sterilised <strong>and</strong> stored at -20°C until use. IgG<br />

concentrations were determined using a Nanodrop ND-1000 spectrophotometer<br />

(Nanodrop Technologies, Wilmington, DE, USA).<br />

P. falciparum strain FCR3 was cultured in vitro using st<strong>and</strong>ard P. falciparum<br />

culture techniques in an atmosphere of 5% CO2, 5% O2 <strong>and</strong> 90% N2. FCR3 AMA1<br />

(accession no. M34553) differs by one amino acid in the pro-sequence from FVO<br />

AMA1 (accession no. AJ277646).


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

The effect of purified IgG antibodies on parasite invasion was evaluated with<br />

two IgG concentrations (5 <strong>and</strong> 10 mg/mL, respectively) in triplicate using 96 well<br />

flat-bottomed plates (Greiner) with synchronized cultures of P. falciparum<br />

schizonts at a starting parasitemia of 0.2-0.4%, a haematocrit of 2.0% <strong>and</strong> a final<br />

volume of 100 µL containing 10% control non-immune human serum, 20 µg /ml<br />

gentamicin in RPMI 1640. After 40 to 42 hours, cultures were resuspended, <strong>and</strong><br />

50 µL was transferred into 200 µL ice-cold PBS. The cultures were then<br />

centrifuged, the supernatant removed <strong>and</strong> the plates were frozen. Inhibition of<br />

parasite growth was estimated using the pLDH assay as previously described<br />

[14]. Parasite growth inhibition, reported as a percentage, was calculated as<br />

follows: 100 - ((Odexperimental - Od background)/ (Odcontrol - Odbackground)<br />

x 100). IgG purified from plasma before immunisation was used as a control, <strong>and</strong><br />

culture medium was used to measure the background Od.<br />

Statistical methods<br />

Safety analyses were based on intention to treat data selection (n=56). For<br />

immunology assays, per protocol analyses were used (n=47). Between group<br />

differences were calculated by one-way ANOVA, using post-hoc Bonferroni<br />

when p


42 Chapter 2<br />

Adjuvant Alhydrogel Montanide AS02 Total<br />

PfAMA1<br />

dose<br />

10µg 50µg 10µg 50µg 10µg 50µg<br />

N 10 10 10 9 9 8 56<br />

Total 8<br />

10<br />

9 9<br />

9 8 53<br />

LOCAL<br />

(80.0 %) (100 %) (90.0 %) (100 %) (100 %) (100 %) (94.6 %)<br />

Pain<br />

8<br />

(80.0 %)<br />

10<br />

(100 %)<br />

8<br />

(80.0 %)<br />

9<br />

(100 %)<br />

9<br />

(100 %)<br />

8<br />

(100 %)<br />

52<br />

(92.9 %)<br />

Erythema<br />

- -<br />

2<br />

(20.0 %)<br />

2<br />

(22.2%)<br />

4<br />

(44.4%)<br />

6<br />

(75.0%)<br />

14<br />

(25%)<br />

Swelling<br />

- -<br />

1<br />

(10.0 %)<br />

-<br />

3<br />

(33.3 %)<br />

1<br />

(12.5 %)<br />

5<br />

(8.9 %)<br />

Induration<br />

- -<br />

1<br />

(10.0 %)<br />

2<br />

(22.2 %)<br />

- -<br />

3<br />

(5.4 %)<br />

Sterile<br />

abscess<br />

SYSTEMIC<br />

- - -<br />

2<br />

(22.2 %)<br />

- -<br />

2<br />

(3.6 %)<br />

Headache<br />

1<br />

(10.0 %)<br />

-<br />

2<br />

(20.0 %)<br />

-<br />

6<br />

(66.7 %)<br />

7<br />

(87.5 %)<br />

16<br />

(28.6 %)<br />

Malaise<br />

- - -<br />

1<br />

(11.1 %)<br />

6<br />

(66.7 %)<br />

7<br />

(87.5 %)<br />

14<br />

(25.0 %)<br />

Fever<br />

- - - -<br />

5<br />

(55.6 %)<br />

5<br />

(62.5 %)<br />

10<br />

(17.9 %)<br />

Myalgia<br />

- - - -<br />

4<br />

(44.4 %)<br />

2<br />

(25.0 %)<br />

6<br />

(10.7 %)<br />

Nausea<br />

1<br />

(10.0 %)<br />

- - -<br />

1<br />

(11.1 %)<br />

2<br />

(25.0 %)<br />

4<br />

(7.1 %)<br />

Fatigue<br />

- - - - -<br />

2<br />

(25.0 %)<br />

2<br />

(3.6 %)<br />

Arthralgia<br />

- - - -<br />

1<br />

(11.1 %)<br />

-<br />

1<br />

(1.8 %)<br />

Abdominal<br />

pain<br />

- - - -<br />

1<br />

(11.1 %)<br />

-<br />

1<br />

(1.8 %)<br />

Table 2. Number of volunteers reporting vaccine related adverse events per dose <strong>and</strong><br />

adjuvant group.<br />

be closely monitored for social, geographic or psychological reasons. Table 1<br />

shows the demographics of volunteers per r<strong>and</strong>omised group. The mean age<br />

was 23 years old (range 18 – 42 years) <strong>and</strong> all but one were Caucasian.<br />

Safety <strong>and</strong> reactogenicity<br />

No serious adverse events occurred that were definitely, probably, or possibly<br />

related to immunisation. No clinically relevant changes in vital signs or<br />

laboratory values were reported throughout the study. Forty-seven volunteers<br />

(84%) received all three immunisations; nine were excluded for one or more<br />

immunisations (Figure 1). Two of these were excluded for reasons unrelated to<br />

the trial procedures. One (Alhydrogel 10 µg group) developed a generalised<br />

rash assessed as unrelated to the vaccine between the first <strong>and</strong> second<br />

immunisations <strong>and</strong> one (Montanide 10 µg group) received a concomitant<br />

hepatitis B immunisation. Seven volunteers were excluded because they<br />

developed grade 3 erythema (diameter >50mm) after the second immunisation;


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

one in the Montanide 10 µg group, the other six in the AS02 groups (two in the<br />

10 µg group, four in the 50 µg group).<br />

Volunteers in all groups presented with local injection site reactions, the most<br />

predominant being transient mild to moderate pain (80-100%, table 2).<br />

Erythema was commonly observed (10 of 17 volunteers) in the AS02 adjuvanted<br />

groups, occurring after the second <strong>and</strong> third immunisation. In the Montanide<br />

group 4 of 18 volunteers developed erythema. Seven volunteers reported<br />

grade 3 erythema <strong>and</strong> were withdrawn from further immunisation after dose 2.<br />

The skin in grade 3 (diameter >50 mm) erythema was not painful <strong>and</strong> did not<br />

limit daily activities. Episodes of erythema generally lasted 2-3 days.<br />

Induration at the site of injection occurred in three volunteers in the course of<br />

the study (table 2). One volunteer, in the Montanide 10 µg group, developed<br />

moderate induration 15 days after the first immunisation, lasting for five days.<br />

The second <strong>and</strong> third immunisations in this volunteer were well tolerated;<br />

induration did not re-appear. Another volunteer developed induration starting<br />

nine days after the first immunisation in the left arm with 50 µg PfAMA1 in<br />

Montanide, lasting 25 days. The second immunisation was well tolerated, but<br />

the left arm induration re-appeared one day after the third immunisation,<br />

accompanied by pain <strong>and</strong> induration at the previous immunisation site in the<br />

contralateral (right) arm. Four weeks later, the induration became soft <strong>and</strong><br />

fluctuated, indicating abscess formation. A total of 63 ml of opaque, brown fluid<br />

was aspirated by two subsequent punctures, after which the abscess <strong>and</strong><br />

induration resolved spontaneously <strong>and</strong> disappeared completely at 81 days post<br />

third immunisation. The third volunteer, also in the 50 µg PfAMA1 adjuvanted<br />

with Montanide group, developed moderate induration nine days after the<br />

second immunisation which lasted approximately one week. Six days after the<br />

third immunisation he developed induration at his left arm (the site of the first<br />

<strong>and</strong> last immunisation) which eventually started fluctuating. A total of 130 ml<br />

brown, opaque fluid was collected by means of two punctures. Thereafter,<br />

spontaneous percutaneous drainage occurred <strong>and</strong> the lesion resolved 57 days<br />

after the third immunisation. Both volunteers did not have any systemic<br />

symptoms such as fever during this time period. Abscesses were only mildly<br />

painful, but limited volunteers daily activities because of their size.<br />

The aspirated fluids from both volunteers were abundant in red blood cells <strong>and</strong><br />

lymphocytes with low Creatinine Kinase (CK) levels. Repeated cultures did not<br />

reveal any bacterial contamination. Serum CK levels were normal. Circulating<br />

43


44 Chapter 2<br />

Figure 2. Mean log anti-AMA-1 titres with st<strong>and</strong>ard deviation for low <strong>and</strong><br />

high dose per adjuvant group. Anti-AMA-1 titres were determined by ELISA<br />

for the six different groups, immunized with Alhydrogel, Montanide <strong>and</strong><br />

AS02 adjuvanted PfAMA1 vaccine. Dashed lines represent high dose of<br />

PfAMA1 (50 µg), continuous lines represent low dose groups (10 µg).<br />

Measurements were performed at baseline, 28 days after the first, second<br />

<strong>and</strong> third immunisation (day 28, 56 <strong>and</strong> 84 respectively) <strong>and</strong> day 140 <strong>and</strong><br />

365.<br />

levels of C-reactive protein remained below detection levels (indicating that the<br />

reaction was a local response). Ultrasound examination suggested an<br />

intramuscular <strong>and</strong> subcutaneous localisation of the fluid-filled cavity.<br />

Systemic reactions were infrequent in the Alhydrogel <strong>and</strong> Montanide groups<br />

<strong>and</strong> occurred mainly in the AS02 groups. The systemic adverse events occurred<br />

within 24 hours of immunisation <strong>and</strong> usually resolved within two days. The most<br />

prevalent systemic adverse events were headache (77.8-87.5%) <strong>and</strong> malaise<br />

(66.7-87.5%) in the AS02 groups. Four of those volunteers reported grade 3<br />

headache or malaise. Most of the systemic adverse events occurred after dose<br />

2. There was no effect of antigen dose on reactogenicity. No changes in blood<br />

pressure were noted in any of these volunteers.


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

Figure 3. Representative immunofluorescent microscopy picture, showing<br />

recognition of native antigen on merozoites by induced anti-AMA1 antibodies.<br />

Immunofluorescence picture of merozoites incubated with 40x diluted anti-AMA1<br />

plasma from an immunized volunteer one month after final immunisation stained<br />

with Rabbit anti-human immunoglobulin FITC (A) <strong>and</strong> DAPI (B). Photo was taken at<br />

magnification 400x. Incubation with monoclonal antibody 4G2, a pan-specific anti-<br />

AMA1 antibody, confirmed the surface staining pattern (not shown).<br />

Humoral immune response<br />

Peak antibody titres were observed one month after the final immunisation.<br />

100% of volunteers in the 10 <strong>and</strong> 50 µg AS02 <strong>and</strong> 50 µg Montanide groups<br />

showed a greater than four-fold increase in antibody titre over preimmunisation<br />

compared to 60% in the 10 µg Alhydrogel, 80% in the 50 µg<br />

Alhydrogel <strong>and</strong> 90% in the 10 µg Montanide groups (Figure 2). All vaccinees<br />

had reached IgG titres comparable to or higher than semi immune sera. Two <strong>and</strong><br />

four months post final immunisation both AS02 groups <strong>and</strong> the Montanide 50 µg<br />

group showed the highest IgG titres but given the small sample sizes there was<br />

no power to detect statistical differences between groups. Antibody titres<br />

decreased further one year post immunisation, with the steepest decline being<br />

in the Montanide groups, to a level comparable with the reference Alhydrogel<br />

groups.<br />

One year post vaccination, titres in the 10 µg AS02 group were significantly<br />

higher as compared to the reference group (post hoc Bonferroni when<br />

compared with low dose Alhydrogel reference group p


46 Chapter 2<br />

Figure 4. ELISPOT assay for IFNγ (A) <strong>and</strong> IL-5 (B) after stimulation with 6 µg PfAMA1.<br />

Peripheral Blood Mononuclear Cells from immunised volunteers 28 days after the<br />

second immunisation <strong>and</strong> 28 days after the third immunisation (day 56 <strong>and</strong> 84<br />

respectively) were stimulated with 6 µg of PfAMA1 vaccine. Production of IFNγ <strong>and</strong> IL-<br />

5 was measured by counting spots in ELISPOT plates. Box plots <strong>and</strong> whiskers show the<br />

range <strong>and</strong> the 25th, 50th <strong>and</strong> 75th percentile of spots per 2x10 5 cells. Circles<br />

represent outliers.


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

Figure 5. Stimulation indices in response to 6 µg/ml PfAMA1 presented as box<br />

plots <strong>and</strong> whiskers. Peripheral Blood Mononuclear Cells from immunised<br />

volunteers 28 days after the second immunisation <strong>and</strong> 28 days after the third<br />

immunisation (Day 56 <strong>and</strong> 84 respectively) were stimulated with 6 µg of<br />

PfAMA1 vaccine. Cell proliferation was measured by adding 3H thymidine <strong>and</strong><br />

calculated relative to control wells. Box plots <strong>and</strong> whiskers show the range <strong>and</strong><br />

the 25th, 50th <strong>and</strong> 75th percentile. Circles represent outliers. Measurements<br />

were performed at baseline, 28 days after the second immunisation <strong>and</strong> 28<br />

days after the third immunisation (day 56 <strong>and</strong> 84 respectively).<br />

Sera from vaccinees could be shown to recognise the native PfAMA1 by<br />

immunofluorescence in a dose dependent manner. Eight of fifteen samples were<br />

positive in IFA, amongst which were four samples with the highest antibody<br />

titres. The staining pattern found in positive samples localised to the same<br />

structures as 4G2 rat monoclonal antibody (Figure 3).<br />

Cellular immune response<br />

In all groups, induction of IFNγ <strong>and</strong> IL-5 cytokines could be demonstrated (Figure<br />

4). The magnitude of cytokine production was not dose dependent or depen-<br />

47


48 Chapter 2<br />

Figure 6. Percentage of growth inhibition of FVO-strain P. falciparum<br />

parasites after addition of 5 or 10 mg/ml IgG. Serum samples from volunteers<br />

immunised with PfAMA1 were obtained four weeks after the final<br />

immunisation by per protocol analysis <strong>and</strong> included in a merozoite growth<br />

inhibition assay. Growth inhibition is expressed as a percentage to control.<br />

Boxes show 25th, 50th <strong>and</strong> 75th percentile growth inhibition, whiskers show<br />

the range, circles are outliers.<br />

dent on the number of immunisations. Rather, IFNγ production in many samples<br />

decreased after the third immunisation. For both cytokines, PfAMA1 induction<br />

was comparable or higher than that following stimulation with 5 µg Tetanus<br />

Toxoid (data not shown). Cytokine production in the different groups did not<br />

differ significantly from each other (for IFNγ p = 0.18, for IL-5 p = 0.14). Ratio’s of<br />

IFNγ / IL-5 production were also not significantly different between adjuvant<br />

groups (data not shown), but showed a trend towards higher ratio in the<br />

Montanide <strong>and</strong> AS02 groups (Day 84 mean ratio Alhydrogel: 1.16 (95% CI: 0.08<br />

to 2.23), Montanide: 2.82 (95% CI: 1.44 to 4.21), AS02: 2.66 (95% CI: 0.57 to<br />

4.76)).<br />

All groups showed Peripheral Blood Mononuclear Cell proliferation upon<br />

stimulation with PfAMA1 (Figure 5). Stimulation indices between PBMC’s<br />

stimulated with 30, 6 or 1.2 µg/ml PfAMA1 were similar. All groups of volunteers


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

showed significant increase in proliferation upon stimulation with PfAMA1 after<br />

the second immunisation. After the third immunisation none of the groups<br />

showed a further increase in stimulation index, rather the 10 µg Montanide<br />

group showed a significant decrease after the third immunisation (p=0.03).<br />

There were no significant differences in stimulation index between the different<br />

adjuvant or dose groups.<br />

In vitro parasite growth inhibition<br />

To estimate functionality of the induced antibodies, an in vitro GIA was<br />

performed. Results are shown as percentage inhibition compared to prevaccination<br />

sera from the same individual (Figure 6). At a concentration of 10<br />

mg/ml, the median growth inhibition in the Montanide 50 µg <strong>and</strong> AS02 groups<br />

was about 30% <strong>and</strong> 50% respectively. In the Alhydrogel <strong>and</strong> Montanide 10 µg<br />

groups median inhibition was lower, ranging from 4 to 17%. Only differences<br />

between Alhydrogel <strong>and</strong> AS02 groups were significant (p=0.002).<br />

Discussion<br />

This trial demonstrates that reactogenicity of PfAMA1-FVO[25-545] varies,<br />

depending on the adjuvant. Immunogenicity at both high <strong>and</strong> low doses <strong>and</strong> in<br />

all adjuvant formulations is good, although the type <strong>and</strong> magnitude of immune<br />

response varied among different adjuvant groups.<br />

PfAMA1-FVO[25-545] mixed with the adjuvants Alhydrogel, Montanide <strong>and</strong><br />

AS02 tended to be locally reactogenic, mainly causing short lasting injection site<br />

pain when administered to healthy adult volunteers. Most post immunisations<br />

adverse events were mild-to-moderate in intensity <strong>and</strong> have been seen<br />

previously with other vaccines [32–37]. Because this was the first time Pichia<br />

pastoris produced FVO PfAMA1 antigen was being given to humans, the<br />

occurrence of a grade 3 adverse event was a stopping criterion, which led to<br />

withdrawal of seven subjects post dose 2 for grade 3 (>50mm) erythema.<br />

However, the erythema observed resolved spontaneously within three days of<br />

onset without any sequelae. The erythema is not considered a hindrance for<br />

further vaccination with the AS02 adjuvant. In terms of systemic adverse events,<br />

most were related to the AS02 adjuvant <strong>and</strong> transient resolving within two days<br />

with no sequelae. The pattern of transient, primarily mild to moderate systemic<br />

adverse events has been reported with another AMA-1 antigen adjuvanted with<br />

AS02 [36].<br />

49


50 Chapter 2<br />

Three immunisations with 50 µg PfAMA1 adjuvanted by Montanide induced a<br />

sterile abscess in two of ten volunteers. Progression of induration to a sterile<br />

abscess has been previously reported before after immunisation with<br />

Montanide [38–40]. In all reports the development of an abscess followed<br />

intramuscular immunisation <strong>and</strong> was accompanied by enhanced<br />

immunogenicity. The increased reactogenicity of Montanide-adjuvanted<br />

vaccines has been attributed to a combination of antigen dose <strong>and</strong> the<br />

formation of a vaccine depot that may persist locally <strong>and</strong> that is inherent to<br />

water-in-oil emulsions [41]. Similarly, induration at the previous immunisation<br />

site has been attributed to persistent antigen in previous trials [31]. A less<br />

condensed vaccination regimen <strong>and</strong> avoidance of the same injection sites may<br />

be measures to avoid induration.<br />

To date, there are four other reports on clinical phase Ia trials of a PfAMA1<br />

vaccine. These trials employed different PfAMA1 constructs <strong>and</strong> utilized<br />

different adjuvants. The constructs were of P. pastoris or E. coli origin or used a<br />

virally vectored delivery system [9,10,36,42].<br />

The P. pastoris-produced PfAMA1 comprised recombinant proteins based on<br />

sequences from the ectodomains of FVO <strong>and</strong> 3D7 strain AMA1 adjuvanted with<br />

Alhydrogel have been tested both in a phase Ia <strong>and</strong> Ib trial. As with previous<br />

studies in which malarial antigens have been adjuvanted with Alhydrogel, this<br />

c<strong>and</strong>idate vaccine showed an acceptable reactogenicity profile but a limited<br />

immune response. The Malkin et al. phase Ia trial shows a GIA response in only 4<br />

of 22 subjects despite high seroconversion rates [10], similar to the data<br />

obtained here with Alhydrogel. Interestingly, in our study, the P. pastoris<br />

PfAMA1 combined with Alhydrogel was much less reactogenic <strong>and</strong> did not<br />

produce any erythema or induration, even though the doses were comparable.<br />

The lower Alhydrogel dose (500 µg per immunisation) used in this trial, as<br />

compared to Malkin et al. (800 µg) may also play a role in its decreased<br />

reactogenicity.<br />

There are two trials utilizing the E. coli-produced 3D7 strain AMA1, one<br />

reconstituted in Montanide <strong>and</strong> a second formulated in AS02. The AMA1-<br />

Montanide combination was considered safe but the trial was compromised by<br />

apparent loss of potency [42]. The AMA1-AS02 combination [36] showed<br />

comparable local <strong>and</strong> systemic reactogenicity. Although Polhemus et al. were<br />

able to show recognition of the native antigen by IFA, growth inhibition results<br />

were approximately two fold lower than those found in this study.


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

Lastly, PfAMA1 has also been evaluated in a multi-antigen malaria vaccine<br />

delivered in an attenuated vaccinia virus. Although weak protective effects were<br />

found, immunogenicity in that trial was poor [9].<br />

After one year follow-up, we found antibody levels still to be significantly higher<br />

than baseline for all groups. This is in sharp contrast to the results of Malkin et<br />

al. [10] who reported detectable antibodies in only 50-90% of volunteers by day<br />

364, even though they had been boosted much later (on Day 180). This suggests<br />

that a more condensed immunisation regime may affect the persistence of<br />

antibodies.<br />

In this trial we have shown that the combination of clinical grade PfAMA1 FVO<br />

[25-545] P. pastoris expressed material with either Montanide or AS02 is<br />

significantly more immunogenic than previous PfAMA1 formulations, being<br />

capable of inducing high levels of antibodies for both dosages in both adjuvant<br />

groups. A positive trend between antigen dose, antibody response <strong>and</strong> in vitro<br />

parasite growth inhibition could be detected, although the effect of antigen<br />

dose on immunogenicity was negligible compared to the effect of varying the<br />

adjuvant. The wide variety of immune responses found in different adjuvant<br />

formulations stresses the importance of adjuvants as a critical component in<br />

malaria vaccine development.<br />

The functionality of vaccine induced antibodies was assessed by growth<br />

inhibition assay. Although this assay has not been validated as a correlate of<br />

<strong>protection</strong>, this trial demonstrates that the st<strong>and</strong>ardised assay is able to<br />

demonstrate recognition of the native protein <strong>and</strong> thus functionality in vitro.<br />

Different adjuvants are known to prompt immune responses towards Th1 or<br />

Th2. It has been previously reported that AS02 induces an immune response<br />

skewed towards Th1 [37], with production of primarily IFNγ. In contrast<br />

Alhydrogel is known to be Th2 inducer [43]. In this study, ratio’s of cytokine<br />

production at day 84 showed relatively more IFNγ over IL-5 production in the<br />

Montanide <strong>and</strong> AS02 groups suggesting a pro-Th1 response, although<br />

statistically non-significant. Interestingly, the additional third immunisation<br />

generally did not lead to a further increase in IFNγ or IL-5 production or in<br />

lymphocyte proliferation. Rather, many volunteers showed a reduction in the<br />

response after the third immunisation. This difference could not be explained by<br />

inter-test variability. It remains to be investigated if it indicates a shift in the<br />

relative balance between immediate effector cells <strong>and</strong> long-lived memory cells.<br />

51


52 Chapter 2<br />

Although this phase I trial is limited with respect to the size <strong>and</strong> generalizibility<br />

to the target population, it met its objectives to outline a generalizable safety<br />

profile. Specifically the direct comparison of the safety profile of different<br />

adjuvants is valuable for future development of AMA1 <strong>and</strong> other malaria<br />

vaccines. Furthermore, the malaria vaccine c<strong>and</strong>idate AMA1 provides the<br />

possibility of assessing functionality of the immune response by a parasite<br />

growth inhibition assay. However, it must be noted that the growth inhibition<br />

assay is not validated as a correlate of <strong>protection</strong>, <strong>and</strong> is as such a limited<br />

predictor for efficacy.<br />

With this study we have shown that the PfAMA1 vaccine combined with<br />

different adjuvants, Alhydrogel, Montanide <strong>and</strong> AS02 provided distinct<br />

reactogenicity profiles. All vaccine formulation were immunogenic at both<br />

dosages. Growth inhibition results indicate that induction of functional immune<br />

responses is probably dependent on adjuvant, underscoring the need for strong<br />

immunopotentiators for malaria vaccines. Altogether, these results are<br />

promising for a future development of a PfAMA1 malaria vaccine.<br />

Acknowledgements<br />

We thank Dominique Lemoine <strong>and</strong> Natalie Imbault for their comments <strong>and</strong><br />

contribution to the manuscript<br />

Funding<br />

This work was supported by a grant from the European Malaria Vaccine<br />

Initiative.


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

References<br />

1. Girard MP, Reed ZH, Friede M, Kieny MP. A review of human vaccine research <strong>and</strong><br />

development: malaria. Vaccine 2007; 25:1567-1580.<br />

2. Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E et al. Duration of <strong>protection</strong><br />

with RTS,S/AS02A malaria vaccine in prevention of Plasmodium falciparum<br />

disease in Mozambican children: single-blind extended follow-up of a r<strong>and</strong>omised<br />

controlled trial. Lancet 2005; 366:2012-2018.<br />

3. Aponte JJ, Aide P, Renom M, M<strong>and</strong>om<strong>and</strong>o I, Bassat Q et al. Safety of the<br />

RTS,S/AS02D c<strong>and</strong>idate malaria vaccine in infants living in a highly endemic area of<br />

Mozambique: a double blind r<strong>and</strong>omised controlled phase I/IIb trial. Lancet 2007;<br />

370:1543-1551.<br />

4. Mitchell GH, Thomas AW, Margos G, Dluzewski AR, Bannister LH. Apical<br />

membrane antigen 1, a major malaria vaccine c<strong>and</strong>idate, mediates the close<br />

attachment of invasive merozoites to host red blood cells. Infect Immun 2004;<br />

72:154-158.<br />

5. Hodder AN, Crewther PE, Anders RF. Specificity of the protective antibody<br />

response to apical membrane antigen 1. Infect Immun 2001; 69:3286-3294.<br />

6. Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A et al. A role for apical<br />

membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum<br />

sporozoites. J Biol Chem 2004; 279:9490-9496.<br />

7. Howell SA, Withers-Martinez C, Kocken CH, Thomas AW, Blackman MJ. Proteolytic<br />

processing <strong>and</strong> primary structure of Plasmodium falciparum apical membrane<br />

antigen-1. J Biol Chem 2001; 276: 31311-31320.<br />

8. Remarque EJ, Faber BW, Kocken CH, Thomas AW. Apical membrane antigen 1: a<br />

malaria vaccine c<strong>and</strong>idate in review. Trends Parasitol 2008; 24:74-84.<br />

9. Ockenhouse CF, Sun PF, Lanar DE, Wellde BT, Hall BT et al. Phase I/IIa safety,<br />

immunogenicity, <strong>and</strong> efficacy trial of NYVAC-Pf7, a pox-vectored, multiantigen,<br />

multistage vaccine c<strong>and</strong>idate for Plasmodium falciparum malaria. J Infect Dis<br />

1998; 177:1664-1673.<br />

10. Malkin EM, Diemert DJ, McArthur JH, Perreault JR, Miles AP et al. Phase 1 clinical<br />

trial of apical membrane antigen 1: an asexual blood-stage vaccine for<br />

Plasmodium falciparum malaria. Infect Immun 2005; 73:3677-3685.<br />

11. Schijns VE, Degen WG. Vaccine immunopotentiators of the future. Clin <strong>Pharma</strong>col<br />

Ther 2007; 82:750-755.<br />

12. Keitel WA, Kester KE, Atmar RL, White AC, Bond NH et al. Phase I trial of two<br />

recombinant vaccines containing the 19kd carboxy terminal fragment of<br />

Plasmodium falciparum merozoite surface protein 1 (msp-1(19)) <strong>and</strong> T helper<br />

epitopes of tetanus toxoid. Vaccine 1999; 18:531-539.<br />

53


54 Chapter 2<br />

13. Ballou WR, Hoffman SL, Sherwood JA, Hollingdale MR, Neva FA et al. Safety <strong>and</strong><br />

efficacy of a recombinant DNA Plasmodium falciparum sporozoite vaccine. Lancet<br />

1987; 1:1277-1281.<br />

14. Amador R, Moreno A, Valero V, Murillo L, Mora AL et al. The first field trials of the<br />

chemically synthesized malaria vaccine SPf66: safety, immunogenicity <strong>and</strong><br />

protectivity. Vaccine 1992; 10:179-184.<br />

15. Lawrence G, Cheng QQ, Reed C, Taylor D, Stowers A et al. Effect of vaccination<br />

with 3 recombinant asexual-stage malaria antigens on initial growth rates of<br />

Plasmodium falciparum in non-immune volunteers. Vaccine 2000; 18:1925-1931.<br />

16. Lawrence GW, Saul A, Giddy AJ, Kemp R, Pye D. Phase I trial in humans of an oilbased<br />

adjuvant SEPPIC Montanide ISA 720. Vaccine 1997; 15:176-178.<br />

17. Saul A, Lawrence G, Smillie A, Rzepczyk CM, Reed C et al. Human phase I vaccine<br />

trials of 3 recombinant asexual stage malaria antigens with Montanide ISA720<br />

adjuvant. Vaccine 1999; 17:3145-3159.<br />

18. Genton B, Betuela I, Felger I, Al-Yaman F, Anders RF et al. A recombinant bloodstage<br />

malaria vaccine reduces Plasmodium falciparum density <strong>and</strong> exerts selective<br />

pressure on parasite populations in a phase 1-2b trial in Papua New Guinea. J<br />

Infect Dis 2002; 185:820-827.<br />

19. Aucouturier J, Dupuis L, Deville S, Ascarateil S, Ganne V. Montanide ISA 720 <strong>and</strong><br />

51: a new generation of water in oil emulsions as adjuvants for human vaccines.<br />

Expert Rev Vaccines 2002; 1:111-118.<br />

20. Heppner DG, Jr., Kester KE, Ockenhouse CF, Tornieporth N, Ofori O et al. Towards<br />

an RTS,S-based, multi-stage, multi-antigen vaccine <strong>against</strong> falciparum malaria:<br />

progress at the Walter Reed Army Institute of Research. Vaccine 2005; 23:2243-<br />

2250.<br />

21. Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E et al. Duration of <strong>protection</strong><br />

with RTS,S/AS02A malaria vaccine in prevention of Plasmodium falciparum<br />

disease in Mozambican children: single-blind extended follow-up of a r<strong>and</strong>omised<br />

controlled trial. Lancet 2005; 366:2012-2018.<br />

22. Aponte JJ, Aide P, Renom M, M<strong>and</strong>om<strong>and</strong>o I, Bassat Q et al. Safety of the<br />

RTS,S/AS02D c<strong>and</strong>idate malaria vaccine in infants living in a highly endemic area of<br />

Mozambique: a double blind r<strong>and</strong>omised controlled phase I/IIb trial. Lancet 2007;<br />

370:1543-1551.<br />

23. Bojang KA, Milligan PJ, Pinder M, Vigneron L, Alloueche A et al. Efficacy of<br />

RTS,S/AS02 malaria vaccine <strong>against</strong> Plasmodium falciparum <strong>infection</strong> in semiimmune<br />

adult men in The Gambia: a r<strong>and</strong>omised trial. Lancet 2001; 358:1927-<br />

1934.<br />

24. Gordon DM, McGovern TW, Krzych U, Cohen JC, Schneider I et al. Safety,<br />

immunogenicity, <strong>and</strong> efficacy of a recombinantly produced Plasmodium


Safety <strong>and</strong> Immunogenicity of a Recombinant Plasmodium falciparum AMA1<br />

Malaria vaccine Adjuvanted with Alhydrogel TM, Montanide ISA 720 or AS02<br />

falciparum circumsporozoite protein-hepatitis B surface antigen subunit vaccine. J<br />

Infect Dis 1995; 171:1576-1585.<br />

25. Sun P, Schwenk R, White K, Stoute JA, Cohen J et al. Protective immunity induced<br />

with malaria vaccine, RTS,S, is linked to Plasmodium falciparum circumsporozoite<br />

protein-specific CD4+ <strong>and</strong> CD8+ T cells producing IFN-gamma. J Immunol 2003;<br />

171:6961-6967.<br />

26. Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE et al. A preliminary<br />

evaluation of a recombinant circumsporozoite protein vaccine <strong>against</strong><br />

Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J<br />

Med 1997; 336:86-91.<br />

27. Kocken CH, Withers-Martinez C, Dubbeld MA, Van Der WA, Hackett F et al. Highlevel<br />

expression of the malaria blood-stage vaccine c<strong>and</strong>idate Plasmodium<br />

falciparum apical membrane antigen 1 <strong>and</strong> induction of antibodies that inhibit<br />

erythrocyte invasion. Infect Immun 2002; 70:4471-4476.<br />

28. Faber BW, Remarque EJ, Kocken CH, Cheront P, Cingolani D et al. Production,<br />

quality control, stability <strong>and</strong> pharmacotoxicity of cGMP-produced Plasmodium<br />

falciparum AMA1 FVO strain ectodomain expressed in Pichia pastoris. Vaccine<br />

2008; 26:6143-6150.<br />

29. Kester KE, McKinney DA, Tornieporth N, Ockenhouse CF, Heppner DG, Jr. et al. A<br />

phase I/IIa safety, immunogenicity, <strong>and</strong> efficacy bridging r<strong>and</strong>omized study of a<br />

two-dose regimen of liquid <strong>and</strong> lyophilized formulations of the c<strong>and</strong>idate malaria<br />

vaccine RTS,S/AS02A in malaria-naive adults. Vaccine 2007; 25:5359-5366.<br />

30. Human D, Crawley F, IJesselmuiden C. Revised declaration of Helsinki. WMA will<br />

continue to revise policy as medicine <strong>and</strong> research changes. BMJ 2001; 323:283-<br />

284.<br />

31. Hermsen CC, Verhage DF, Telgt DS, Teelen K, Bousema JT et al. Glutamate-rich<br />

protein (GLURP) induces antibodies that inhibit in vitro growth of Plasmodium<br />

falciparum in a phase 1 malaria vaccine trial. Vaccine 2007; 25:2930-2940.<br />

32. Zepp F, Knuf M, Habermehl P, Mannhardt-Laakmann W, Howe B et al. Safety of<br />

reduced-antigen-content tetanus-diphtheria-acellular pertussis vaccine in<br />

adolescents as a sixth consecutive dose of acellular pertussis-containing vaccine. J<br />

Pediatr 2006; 149:603-610.<br />

33. Kshirsagar N, Mur N, Thatte U, Gogtay N, Viviani S et al. Safety, immunogenicity,<br />

<strong>and</strong> antibody persistence of a new meningococcal group A conjugate vaccine in<br />

healthy Indian adults. Vaccine 2007; 25 suppl1:A101-A107<br />

34. Malkin E, Long CA, Stowers AW, Zou L, Singh S et al. Phase 1 Study of Two<br />

Merozoite Surface Protein 1 (MSP1(42)) Vaccines for Plasmodium falciparum<br />

Malaria. PLoS Clin Trials 2007; 2:e12.<br />

55


56 Chapter 2<br />

35. Kester KE, McKinney DA, Tornieporth N, Ockenhouse CF, Heppner DG, Jr. et al. A<br />

phase I/IIa safety, immunogenicity, <strong>and</strong> efficacy bridging r<strong>and</strong>omized study of a<br />

two-dose regimen of liquid <strong>and</strong> lyophilized formulations of the c<strong>and</strong>idate malaria<br />

vaccine RTS,S/AS02A in malaria-naive adults. Vaccine 2007; 25:5359-5366.<br />

36. Polhemus ME, Magill AJ, Cummings JF, Kester KE, Ockenhouse CF et al. Phase I<br />

dose escalation safety <strong>and</strong> immunogenicity trial of Plasmodium falciparum apical<br />

membrane protein (AMA-1) FMP2.1, adjuvanted with AS02A, in malaria-naive<br />

adults at the Walter Reed Army Institute of Research. Vaccine 2007; 25:4203-<br />

4212.<br />

37. V<strong>and</strong>epapeliere P, Rehermann B, Koutsoukos M, Moris P, Garcon N et al. Potent<br />

enhancement of cellular <strong>and</strong> humoral immune responses <strong>against</strong> recombinant<br />

hepatitis B antigens using AS02A adjuvant in healthy adults. Vaccine 2005;<br />

23:2591-2601.<br />

38. Langermans JA, Schmidt A, Vervenne RA, Birkett AJ, Calvo-Calle JM et al. Effect of<br />

adjuvant on reactogenicity <strong>and</strong> long-term immunogenicity of the malaria Vaccine<br />

ICC-1132 in macaques. Vaccine 2005; 23:4935-4943.<br />

39. Langermans JA, Hensmann M, van GM, Zhang D, Pan W et al. Preclinical<br />

evaluation of a chimeric malaria vaccine c<strong>and</strong>idate in Montanide ISA 720:<br />

immunogenicity <strong>and</strong> safety in rhesus macaques. Hum Vaccin 2006; 2:222-226.<br />

40. Toledo H, Baly A, Castro O, Resik S, Laferte J et al. A phase I clinical trial of a multiepitope<br />

polypeptide TAB9 combined with Montanide ISA 720 adjuvant in non-HIV-<br />

1 infected human volunteers. Vaccine 2001; 19:4328-4336.<br />

41. Miles AP, McClellan HA, Rausch KM, Zhu D, Whitmore MD et al. Montanide ISA<br />

720 vaccines: quality control of emulsions, stability of formulated antigens, <strong>and</strong><br />

comparative immunogenicity of vaccine formulations. Vaccine 2005; 23:2530-<br />

2539.<br />

42. Saul A, Lawrence G, Allworth A, Elliott S, Anderson K et al. A human phase 1<br />

vaccine clinical trial of the Plasmodium falciparum malaria vaccine c<strong>and</strong>idate<br />

apical membrane antigen 1 in Montanide ISA720 adjuvant. Vaccine 2005;<br />

23:3076-3083.<br />

43. Ulanova M, Tarkowski A, Hahn-Zoric M, Hanson LA The Common vaccine adjuvant<br />

aluminum hydroxide up-regulates accessory properties of human monocytes via<br />

an interleukin-4-dependent mechanism. Infect Immun 2001; 69:1151-1159.


Chapter 3<br />

Humoral immune responses to a single<br />

allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

Edmond J. Remarque* 1 , Meta Roestenberg 2 , Sumera Younis 1 , Vanessa<br />

Walraven 1 , Nicole van der Werff 1 , Bart W. Faber 1 , Odile Leroy 3 , Robert<br />

Sauerwein 2 , Clemens H.M. Kocken 1 Alan W. Thomas 1<br />

Biomedical Primate Research Centre, Rijswijk, The Netherl<strong>and</strong>s<br />

Radboud University Nijmegen Medical Centre, Nijmegen, The Netherl<strong>and</strong>s<br />

European Vaccine Initiative, Heidelberg, Germany<br />

PLoS ONE, 2012;7(6):e38898.


Chapter 3 58<br />

Abstract<br />

Plasmodium falciparum apical membrane antigen 1 (AMA1) is a c<strong>and</strong>idate<br />

malaria vaccine antigen expressed on merozoites <strong>and</strong> sporozoites. The<br />

polymorphic nature of AMA1 may compromise vaccine induced <strong>protection</strong>.<br />

The humoral response induced by two dosages (10 <strong>and</strong> 50 µg) of a single allele<br />

AMA1 antigen formulated with Alhydrogel, Montanide ISA 720 or AS02 was<br />

investigated in 47 malaria-naïve adult volunteers. Volunteers were vaccinated 3<br />

times at 4 weekly intervals <strong>and</strong> serum samples obtained four weeks after the<br />

third immunization were analysed for (i) antibody responses to various allelic<br />

variants, (ii) domain specificity, (iii) avidity, (iv) IgG subclass levels, by ELISA <strong>and</strong><br />

(v) functionality of antibody responses by Growth Inhibition Assay.<br />

About half of the antibodies induced by vaccination cross reacted with<br />

heterologous AMA1 alleles. The choice of adjuvant determined the magnitude of<br />

the antibody response, but had only a marginal influence on specificity, avidity,<br />

domain recognition or subclass responses. The highest antibody responses were<br />

observed for AMA1 formulated with AS02. The Growth Inhibition Assay (GIA)<br />

activity of the antibodies was proportional to the amount of antigen specific IgG<br />

<strong>and</strong> the functional capacity of the antibodies was similar for heterologous<br />

AMA1-expressing laboratory strains.


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

Introduction<br />

Malaria is a serious public health problem causing high levels of morbidity <strong>and</strong><br />

mortality in malaria-endemic regions [1]. An effective malaria vaccine will<br />

contribute to reducing the burden of malaria in addition to existing measures<br />

like insecticide-treated bed nets, antimalarials <strong>and</strong> vector control.<br />

Apical Membrane Antigen 1 (AMA1) is a promising malaria vaccine c<strong>and</strong>idate<br />

(reviewed in [2]), expressed on merozoites <strong>and</strong> sporozoites as a type I integral<br />

membrane protein [3,4] <strong>and</strong> is initially located in the micronemes [3,5]. In<br />

Plasmodium falciparum AMA1 is expressed as an 83 kDa protein <strong>and</strong> relocates<br />

as a 66 kDa protein to the parasite surface following cleavage of the prosequence<br />

at the time of invasion [6], where it is subsequently shed as 44 <strong>and</strong> 48<br />

kDa forms [6]. The AMA1 ectodomain consists of an N-terminal pro-sequence<br />

<strong>and</strong> three tightly interacting domains [7,8]. AMA1 is believed to play an essential<br />

role in red blood cell invasion [9] <strong>and</strong> may also be implicated in liver cell invasion<br />

by sporozoites [4]. The protective efficacy of AMA1-based vaccines has been<br />

demonstrated in numerous mouse <strong>and</strong> simian models (reviewed in [2]).<br />

AMA1 is a polymorphic antigen [10], <strong>and</strong> this polymorphism is entirely due to<br />

single amino acid substitutions [7]. These polymorphisms have been found to be<br />

restricted to the surface of AMA1, predominantly mapping to one molecular<br />

face [7,8,11]. Studies with the rodent malaria P. chabaudi have shown that<br />

polymorphism in AMA1 can ablate vaccine efficacy [12]. Rabbit immunisation<br />

studies have demonstrated that, although antibodies raised <strong>against</strong> one PfAMA1<br />

allele show excellent inhibition of strains expressing a homologous allele, strains<br />

expressing a heterologous PfAMA1 allele are inhibited to a variable lesser<br />

degree depending on the antigenic differences <strong>and</strong> their locations [13–17],<br />

suggesting that PfAMA1 polymorphism reduces the efficacy of PfAMA1 based<br />

vaccines. Results from a Phase IIb vaccine study in Malian children suggests that<br />

the specificity of the AMA1 immune response is crucial in <strong>protection</strong> [18].<br />

Most of the data addressing the impact of polymorphism have been generated<br />

in laboratory animals <strong>and</strong> relatively little is known from human vaccination<br />

studies. A phase Ia study with 10 or 50 µg of a single allele vaccine (PfAMA1 FVO<br />

25-545) formulated with three adjuvants [19] in malaria-naïve subjects offers a<br />

unique opportunity to perform an in depth analysis of antibody responses in<br />

humans. Total IgG <strong>and</strong> GIA responses to the homologous antigen have been<br />

59


60 Chapter 3<br />

reported previously [19]. The analysis reported here comprises ELISA <strong>and</strong><br />

Growth Inhibition Assay (GIA) titres to the homologous <strong>and</strong> heterologous AMA1<br />

antigens, domain specificity, subclass distribution, avidity <strong>and</strong> the relation<br />

between GIA <strong>and</strong> Elisa titres after 3 immunisations with PfAMA1 FVO 25-545. In<br />

addition, it also offers the opportunity to investigate the effect of various<br />

adjuvants on the quality <strong>and</strong> quantity of the humoral immune response.<br />

Adjuvant Dose N Age Minimum Maximum<br />

Alhydrogel 10 9 23.3 ± 2.9 21.1 29.5<br />

Alhydrogel 50 10 23.2 ± 2.1 19.4 26.9<br />

ISA720 10 8 23.1 ± 5.2 18.5 33.7<br />

ISA720 50 9 23.2 ± 3.6 19.7 31.1<br />

AS02 10 7 24.5 ± 8.3 20.0 42.8<br />

AS02 50 4 24.3 ± 4.8 20.5 31.3<br />

Table 1: Characteristics of the subjects at the time of first vaccination (per<br />

protocol)<br />

Materials <strong>and</strong> Methods<br />

Participants<br />

Fifty-six malaria-naïve healthy male study participants were recruited at the<br />

Radboud University Nijmegen Medical Centre as previously reported [19]. A<br />

total of 47 subjects completing all 3 immunisations were included for the per<br />

protocol analysis in the current paper [19]. The characteristics of the subjects at<br />

the time of first vaccination are presented in Table 1. All volunteers provided<br />

written informed consent. The study was approved by the Institutional Review<br />

Board (CMO Regio Arnhem-Nijmegen, 2005/015). The study was conducted in<br />

accordance with the Declaration of Helsinki principles for the conduct of clinical<br />

trials <strong>and</strong> the International Committee of Harmonization Good Clinical Practice<br />

Guidelines <strong>and</strong> registered at www.clinicaltrials.gov (NCT00730782).<br />

Vaccines, vaccination <strong>and</strong> blood samples<br />

Clinical grade PfAMA1 FVO[25-545] consisting of P. falciparum FVO-strain AMA1<br />

ectodomain (amino acids 25 to 545) was produced as previously described [20].<br />

The cGMP produced PfAMA1 FVO[25-545] was available in multidose vials<br />

containing 120 µg lyophilised AMA1 (44 µg EDTA, 180 µg saccharose <strong>and</strong> 120 µg<br />

NaHCO3, Lot B) or 62.5 µg lyophilised AMA1 (23 µg EDTA, 25 mg saccharose, 226<br />

µg K2HPO4 <strong>and</strong> 187 µg NaH2PO4, Lot C). PfAMA1 vaccines of 0.5 mL were<br />

prepared at two dosages (10 <strong>and</strong> 50 µg) with three different adjuvants


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

*----30---*----40---* ---50 --*----60---*----70---*----80---*----90---*----100--*----110--*----<br />

120--<br />

FVO ∆ Glyc<br />

QNYWEHPYQKSDVYHPINEHREHPKEYEYPLHQEHTYQQEDSGEDENTLQHAYPIDHEGAEPAPQEQNLFSSIEIVERSNYMGNPWTEYMAKYDIEEVHG<br />

3D7 ∆.Glyc ---------N----R----------------------------------------------------------------------------------<br />

---<br />

HB3 ∆ Glyc ---------N----R---------------------------------------------------------------------------------<br />

K---<br />

CAMP ∆ Glyc ---------N—N---------------Q---------------------------------------------------------------------<br />

---<br />

*----130--*----140--*----150--*----160--*----170--*----180--*----190--*----200--*----210--*----<br />

220--<br />

FVO ∆ Glyc<br />

SGIRVDLGEDAEVAGTQYRLPSGKCPVFGKGIIIENSKTTFLKPVATGNQDLKDGGFAFPPTNPLISPMTLNGMRDFYKNNEYVKNLDELTLCSRHAGNM<br />

3D7 ∆ Glyc ------------------------------------------T-------Y-----------E—-M-----DE—-H---D-K---------------<br />

---<br />

HB3 ∆ Glyc ------------------------------------------T----E--------------E--------DQ—-HL—-D-----------------<br />

---<br />

CAMP ∆ Glyc --------------------------------------------------------------E----------------------------------<br />

---<br />

*----230--*----240--*----250--*----260--*----270--*----280--*----290--*----300--*----310--*----<br />

320--<br />

FVO ∆ Glyc<br />

NPDNDKNSNYKYPAVYDYNDKKCHIL<strong>TI</strong>AAQENNGPRYCNKDQSKRNSMFCFRPAKDKLFENYVYLSKNVVDNWEEVCPRKNLENAKFGLWVDGNCEDIP<br />

3D7 ∆ Glyc I----------------DK-----------------------E--------------IS-Q--------------K-------Q-------------<br />

---<br />

HB3 ∆ Glyc ------------------E-----------------------E------------------------------------------------------<br />

---<br />

CAMP ∆ Glyc ---K-E-----------DK-----------------------E---------------S-Q--------------K---------------------<br />

---<br />

*----330--*----340--*----350--*----360--*----370--*----380--*----390--*----400--*----410--*----<br />

420--<br />

FVO ∆ Glyc<br />

HVNEFSANDLFECNKLVFELSASDQPKQYEQHLTDYEKIKEGFKNKNADMIKSAFLPTGAFKADRYKSHGKGYNWGNYNRETQKCEIFNVKPTCLINDKS<br />

3D7 ∆ Glyc -----P-I-----------------------------------------------------------------------T-----------------<br />

---<br />

HB3 ∆ Glyc --------------------------------------------------------------------R----------T-----------------<br />

--<br />

CAMP ∆ Glyc --------------------------------------------------------------------------------K-H--------------<br />

---<br />

*----430--*----440--*----450--*----460--*----470--*----480--*----490--*----500--*----510--*----<br />

520--<br />

FVO ∆ Glyc<br />

YIATTALSHPIEVEHNFPCSLYKDEIKKEIERESKRIKLNDNDDEGNKKIIAPRIFISDDKDSLKCPCDPEMVSQSTCRFFVCKCVERRAEVTSNNEVVV<br />

3D7 ∆ Glyc --------------N-----------M----------------------------------------------------------------------<br />

---<br />

HB3 ∆ Glyc ----------N---N--------------------------------------------------------I------N--------K---------<br />

---<br />

CAMP ∆ GlyC --------------N--------N—-M----------------------------------------------------------------------<br />

----<br />

*----530--*----540--*<br />

FVO ∆ Glyc KEEYKEDYADIPEHKPTYDNM<br />

3D7 ∆ Glyc -------------------K-<br />

HB3 ∆ Glyc ---------------------<br />

CAMP ∆ Glyc ---------------------<br />

Figure 1: Alignment of AMA1 protein sequences amino acids 25 through 545 used for Elisa<br />

measurements. Potential N-glycosylation sited were changed (six for FVO, 3D7 <strong>and</strong> CAMP<br />

<strong>and</strong> five for HB3) to avoid unwanted N-glycosylation: N 162 K (For FVO, 3D7 <strong>and</strong> CAMP.<br />

HB3 AMA1 has K at position 162). T 288 V, S 273 D, N 422 D, S 423 K <strong>and</strong> N 499 Q.<br />

(Alhydrogel, Montanide ISA 720 or AS02) as previously described [19].<br />

Alhydrogel <strong>and</strong> Montanide ISA 720 vaccines were prepared with lot B PfAMA1<br />

<strong>and</strong> AS02 vaccines were prepared with lot C PfAMA1. Formulated vaccines were<br />

kept at 4°C for a maximum of six hours until administration. Vaccine<br />

formulations were tested for stability as previously described [20] <strong>and</strong> all<br />

fulfilled the pre-set specifications. Immunisations were given intramuscularly in<br />

the deltoid region of alternate arms at days 0, 28 <strong>and</strong> 56. Blood was collected in<br />

VacutainerTM CPT tubes (Becton <strong>and</strong> Dickinson), plasma collected after<br />

centrifugation <strong>and</strong> stored at -20°C.<br />

61


62 Chapter 3<br />

Quantification ELISA for total IgG <strong>and</strong> IgG subclasses<br />

Enzyme-linked immunosorbent assay (ELISA) was performed in duplicate on<br />

plasma samples in 96 well flat-bottomed microtitre plates (Greiner, Alphen a/d<br />

Rijn, The Netherl<strong>and</strong>s), coated with 1 µg/mL purified AMA1 antigens according<br />

to published methods [10]. The sequences of the four Pichia pastoris-expressed<br />

AMA1 proteins used for ELISA are shown in Figure 1, potential N-glycosylation<br />

sites were removed as previously described [10,15] <strong>and</strong> proteins were produced<br />

as previously described [20]. The P. pastoris-expressed AMA1 from 3D7, HB3 <strong>and</strong><br />

CAMP used in the ELISA’s differ by 26 (2, 17, 5 <strong>and</strong> 2 for prodomain <strong>and</strong> domains<br />

I, II <strong>and</strong> III, respectively), 20 (2, 11, 4, 3 for prodomain <strong>and</strong> domains I, II <strong>and</strong> III,<br />

respectively) <strong>and</strong> 17 (3, 9, 3, 2 for prodomain <strong>and</strong> domains I, II <strong>and</strong> III,<br />

respectively) amino acid positions in the ectodomains (aa 25-545) (Figure 1). The<br />

secondary antibody was goat anti-human IgG conjugated to alkaline<br />

phosphatase (Pierce, Rockford, IL), or mouse anti-human IgG subclass<br />

conjugated to horseradish peroxidase (Sanquin, Amsterdam, The Netherl<strong>and</strong>s).<br />

A st<strong>and</strong>ard curve was included on each plate <strong>and</strong> antibody levels in the<br />

unknowns were calculated using a four-parameter fit. Titres are expressed as<br />

arbitrary units, where 1 AU yields an OD of 1 over background. Thus the amount<br />

of AU of a sample is the reciprocal dilution at which an OD of 1 over background<br />

will be achieved.<br />

Competition Elisa<br />

Competition ELISA was performed as previously described [16]. Dilutions that<br />

resulted in 2 AU were calculated for each plasma sample <strong>and</strong> used in the<br />

subsequent antigen competition assay. The assay involved co-incubation of<br />

different allelic forms of PfAMA1, or PfAMA1-FVO domains with test plasma in<br />

plates coated with PfAMA1 FVO allele, such that competition occurs between<br />

the added (competitor) antigens <strong>and</strong> the coated antigen for binding to test IgG’s.<br />

For the AMA1 allelic forms the competitor/soluble antigens were titrated 3-fold<br />

over 7 wells from 100 to 0.137 µg /ml with PfAMA1 from the FVO, 3D7, HB3 or<br />

CAMP alleles (Figure 1). For the PfAMA1-FVO domains competition ELISA, plates<br />

were coated with FVO AMA1 amino acids 97 through 545 (Domains I, II <strong>and</strong> III) a<br />

fixed concentration of 30 µg/ml of various FVO AMA1 domain constructs were<br />

used as competitors (pro-DI-II-III aa 25-545, DI-II aa 97 - 442, DII-III aa 303 - 545<br />

<strong>and</strong> DII aa 303 - 442). The appropriately diluted plasma was then added <strong>and</strong><br />

after incubation for 2 h, plates were developed as described above. Values are<br />

presented as the fraction remaining relative to the initial amount.


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

Avidity ELISA<br />

The avidities of the antibodies were determined by sodium isothiocyanate<br />

(NaSCN) elution ELISA as previously described [16]. Briefly, microtitre plates<br />

were coated with AMA1 variant proteins (Figure 1) as described above, <strong>and</strong> after<br />

blocking, incubated with a pre-determined titre (1 AU) of sera for 1 h. Plates<br />

were then washed <strong>and</strong> incubated with an increasing concentration of NaSCN<br />

(ranging from 0 to 3.0 M in 0.25 M steps) in duplicate wells for 15 min. Plates<br />

were washed <strong>and</strong> developed with goat anti-human IgG alkaline phosphatase<br />

conjugate <strong>and</strong> substrate as previously described [16]. The avidity index is<br />

expressed as the concentration of NaSCN required for 50% dissociation of bound<br />

antibodies (relative to duplicate wells without NaSCN).<br />

Growth Inhibition Assay (GIA)<br />

Antibodies used for growth inhibition assays (GIA) were purified from CPT<br />

plasma on protein A columns (Immunopure Plus Pierce, St Louis, MO, USA),<br />

exchanged into RPMI 1640 using Amicon Ultra-15 concentrators (30 kDa cut-off,<br />

Millipore, Irel<strong>and</strong>), filter-sterilised <strong>and</strong> stored at -20°C until use. IgG<br />

concentrations were determined using a Nanodrop ND-1000 spectrophotometer<br />

(Nanodrop Technologies, Wilmington, DE, USA). P. falciparum strains FCR3, 3D7<br />

<strong>and</strong> HB3 were cultured in vitro using st<strong>and</strong>ard culture techniques in an<br />

atmosphere of 5% CO2, 5% O2 <strong>and</strong> 90% N2. FCR3 AMA1 (accession no. M34553)<br />

differs by one amino acid in the pro-sequence (D36G) from FVO AMA1<br />

(accession no. AJ277646). The ectodomain (amino acids 25-545) of 3D7<br />

(accession no. U65407) differs by 26 amino acids (2, 17, 5 <strong>and</strong> 2 for prodomain<br />

<strong>and</strong> domains I, II <strong>and</strong> III, respectively) from FVO <strong>and</strong> the ectodomain of HB3<br />

(accession no. U33277) differs by 21 amino acids (2, 12, 4 <strong>and</strong> 3 for prodomain<br />

<strong>and</strong> domains I, II <strong>and</strong> III, respectively) from FVO.<br />

The GIA was performed as previously described [14]. Briefly, the effect of<br />

purified IgG antibodies on in vitro parasite growth was evaluated at two IgG<br />

concentrations (5 <strong>and</strong> 10 mg/mL, respectively) <strong>and</strong> each participants preimmune<br />

IgG was used as negative control. A IgG concentration of 10 mg /ml<br />

approximates the amount of IgG (9.5 to 11.5 mg /ml) found in undiluted human<br />

plasma [21]. Samples were run in triplicate using 96 well flat-bottomed plates<br />

with alanine-synchronized cultures of P. falciparum schizonts at an initial<br />

parasitemia of 0.2–0.4%, a haematocrit of 2.0% <strong>and</strong> a final volume of 100 µL.<br />

After 40 to 42 hours, cultures were resuspended, <strong>and</strong> 50 µL was transferred into<br />

200 µL ice-cold PBS. The cultures were then centrifuged, the supernatant<br />

63


64 Chapter 3<br />

AMA1 IgG (AU/mL)<br />

65536<br />

32768<br />

16384<br />

8192<br />

4096<br />

2048<br />

1024<br />

512<br />

FVO<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

3D7<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Treatment<br />

removed <strong>and</strong> the plates were frozen. Parasite growth was assessed by<br />

measuring parasite lactate dehydrogenase levels with the lactate diaphorase<br />

APAD substrate system, <strong>and</strong> plates were read at 655 nm after 30 min incubation<br />

in the dark. Parasite growth inhibition was expressed as: 100 x (1 – (OD655<br />

Day84 – OD655 RBC)/(OD655 Day0 – OD655 RBC)), Where: OD655Day84 is the<br />

OD655 for IgG purified from day 84 plasma, OD655Day0 is the OD655 for IgG<br />

purified from day 0 plasma <strong>and</strong> OD655RBC is the average OD655 of RBC control<br />

wells. The data are presented as the arithmetic mean % inhibition from each<br />

sample triplicate.<br />

Statistics<br />

Antibody titres (IgG <strong>and</strong> IgG subclasses) were log-transformed to obtain<br />

normality <strong>and</strong> are presented as geometric means with 95% confidence intervals.<br />

GIA titres, IgG avidity <strong>and</strong> levels of depletion were approximately normally<br />

distributed <strong>and</strong> are therefore presented as arithmetic means with 95% confi-<br />

HB3<br />

CAMP<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Figure 2: IgG levels to the vaccine allele <strong>and</strong> three variant AMA1 alleles in serum<br />

obtained on day 84. IgG antibody levels to the vaccine antigen (FVO) <strong>and</strong> 3<br />

heterologous AMA1 alleles (3D7, HB3 <strong>and</strong> CAMP). Values for each subject within a<br />

treatment group are indicated by a specific symbol (same symbol within each<br />

treatment group is same subject throughout graphs 2, 4, 5, 6 <strong>and</strong> 7), boxes indicate<br />

median <strong>and</strong> quartile ranges. Treatment groups are indicated by Adjuvant name <strong>and</strong><br />

AMA1 dose, respectively.


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

Fraction of IgG remaining (%)<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

0<br />

0.14<br />

0.41<br />

Alhydrogel 10 Alhydrogel 50<br />

ISA 720 10 ISA 720 50<br />

AS02 10 AS02 50<br />

1.23<br />

3.7<br />

11.11<br />

33.33<br />

100<br />

0<br />

Competitor concentration (µg/mL)<br />

Figure 3. Competition ELISA using four different competitor antigens on FVO coated<br />

plates in serum obtained on day 84.<br />

Concentration of competitor antigen is depicted on the x-axis (µg /ml) <strong>and</strong> the fraction<br />

remaining bound to the ELISA plate is depicted on the y-axis (%). Light Blue = FVO AMA1<br />

(homologous), Orange = HB3 AMA1, Green = CAMP AMA1 <strong>and</strong> Dark Blue = 3D7 AMA1.<br />

Treatment groups are indicated by Adjuvant name <strong>and</strong> AMA1 dose, respectively.<br />

0.14<br />

0.41<br />

1.23<br />

3.7<br />

11.11<br />

33.33<br />

100<br />

65


66 Chapter 3<br />

dence intervals. The statistical significance of between group differences was<br />

initially evaluated using one way (comparing the 6 adjuvant-dose groups<br />

directly) Analysis of Variance (ANOVA).<br />

Significant between group differences were further evaluated by Tukey’s Honest<br />

Significant Difference post-hoc test which applies a correction on the p-value for<br />

multiple comparisons <strong>and</strong> provides estimates with 95% confidence intervals<br />

(95% CI) for the between group comparisons.<br />

The relation between GIA <strong>and</strong> IgG titres was investigated by non linear least<br />

squares regression using the following formula: GIA = 100 * 1 / [1 + Exp(( Ln IC50<br />

– Ln IgG ) * Slope) ], where IgG is the antibody titre, IC50 represents the IgG<br />

concentration yielding 50% inhibition <strong>and</strong> slope is a parameter indicating the<br />

steepness of the curve, with steeper curves at higher (absolute) values. As Slope<br />

is rather difficult to interpret, it is expressed as the fold-increase in IgG required<br />

to raise GIA levels from 10 to 50% <strong>and</strong>, as the sigmoid is symmetrical around<br />

50%, also from 50 to 90%. It can be shown algebraically that this is<br />

approximately e(2.197 / Slope).<br />

Results<br />

IgG responses to AMA1 alleles<br />

All plasma samples obtained 4 weeks after the third vaccination (Day 84) were<br />

titrated in a single laboratory run on 4 different AMA1 variants; IgG antibody<br />

titres are depicted in Figure 2. Results obtained for the FVO AMA1 are in<br />

agreement with those previously reported (r = 0.916, p < 0.0001)[19]. IgG<br />

antibody levels were lowest in the Alhydrogel groups (10 <strong>and</strong> 50 µg) <strong>and</strong> in the<br />

Montanide ISA 720 10 µg group as compared to the Montanide ISA 720 50 µg<br />

<strong>and</strong> both AS02 groups, this trend was consistently observed for all AMA1<br />

variants under investigation (Figure 2). The IgG antibody levels to the<br />

heterologous AMA1 (3D7, HB3 <strong>and</strong> CAMP) variants, were approximately 50%<br />

lower as compared to the homologous FVO AMA1 (Figure 2). IgG antibody levels<br />

to all AMA1 variants under investigation were 4 to 5 fold higher in the<br />

Montanide ISA 720 50 µg <strong>and</strong> AS02 10 µg groups as compared to the Alhydrogel<br />

10 µg group (Figure 2, all p < 0.02).<br />

The specificity of the IgG responses in day 84 samples was further characterised<br />

using competition ELISA, where increasing amounts of competitor antigen were<br />

co-incubated with a fixed amount of antibodies. Figure 3 shows that the addition


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

Figure 4. Competition ELISA using AMA1 domain constructs in serum obtained on<br />

day 84.The fraction of antibodies remaining bound after competition with 30 µg<br />

of the indicated competitor are depicted on the y-axis, boxes indicate median <strong>and</strong><br />

quartile ranges. P-DI-II-III = FVO AMA1 ectodomain amino acids 25 – 545, DI-II =<br />

FVO-AMA1 amino acids 97 – 442, DII-III = FVO AMA1 amino acids 303 – 545 <strong>and</strong><br />

DII = FVO AMA1 amino acids 303 – 442. Treatment groups are indicated by<br />

Adjuvant name <strong>and</strong> AMA1 dose, respectively.<br />

of increasing amounts of antigen reduces the amount of bound antibody for all<br />

competitor antigens, with the most pronounced reduction observed for the<br />

homologous antigen; the heterologous antigens also reduced the amount of IgG<br />

remaining bound, but less efficiently as compared to the homologous antigen.<br />

The depletion curves per competitor antigen were similar, as judged by IC50 <strong>and</strong><br />

slope parameters, for the adjuvant <strong>and</strong> dose groups, suggesting neither adjuvant<br />

nor dose markedly influenced antibody specificities (Figure 3). The amount of<br />

competitor antigen required to remove 50% of antibodies (IC50) was lowest for<br />

FVO at 0.23 µg /mL (95% CI 0.21 to 0.26), 1.47 µg/ml (95% CI 1.08 to 2.02) for<br />

HB3, 4.39 µg /mL-(95% CI 2.93 to 6.57) for CAMP <strong>and</strong> 20.15 µg/mL (95% CI 14.17<br />

to 28.65) for 3D7 (Figure 3). The increase in competitor antigen concentration<br />

required to reduce the remaining amount of bound antibodies from 50 to 10%<br />

was 38-fold (95% CI 34 to 43) for FVO AMA1. The slopes for the heterologous<br />

AMA1 proteins indicated that a much larger increase in competitor concentra-<br />

67


68 Chapter 3<br />

AMA1 IgG avidity (M NaSCN)<br />

2.5<br />

2.0<br />

1.5<br />

1.0<br />

0.5<br />

FVO<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

3D7<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Alhydrogel 10<br />

Treatment<br />

HB3<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

CAMP<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Figure 5. Antibody avidity to the vaccine allele <strong>and</strong> three variant alleles in serum<br />

obtained on day 84, boxes indicate median <strong>and</strong> quartile ranges. Antibody avidity is<br />

expressed as the amount of NaSCN required to dissociate 50% of bound antibodies.<br />

Treatment groups are indicated by Adjuvant name <strong>and</strong> AMA1 dose, respectively.<br />

tion was required to reduce the amount of bound antibodies from 50 to<br />

10%: 1015-fold (95% CI 702 to 1535), 1328-fold (95% CI 920 to 2001) <strong>and</strong> 910fold<br />

(95% CI 471 to 2058) for HB3, CAMP <strong>and</strong> 3D7 AMA1, respectively (Figure 3).<br />

IgG responses to AMA1 domains<br />

The domain specificity of the antibody response at day 84 was investigated using<br />

competition ELISA’s, where fixed amounts (30 µg) of PfAMA1-FVO domains were<br />

added. Figure 4 shows that 7.2% (95% CI 3.3 to 11.2) of the IgG remained bound<br />

after the addition of 30 µg of the full length antigen, indicating over 90%<br />

depletion. The fraction of IgG remaining after the addition of the full length<br />

antigen (p-DI-II-III) did not differ for the groups (p=0.36). After addition of<br />

domains I-II approximately 19.0% (95% CI 17.6 to 20.5) of the IgG remained<br />

bound, indicating more than 80% depletion. The addition of domains I-II resulted<br />

in significant between group differences in the fraction remaining bound (p =<br />

0.0009, ANOVA). More IgG remained bound In the Montanide ISA 720 10 µg<br />

group when compared to the Alhydrogel 50 µg <strong>and</strong> AS02 50 µg groups, (7.8 %<br />

(95% CI 2.0 to 13.6, p = 0.0030, Tukey HSD) <strong>and</strong> 10.9 (95% CI 3.4 to 18.4. p =


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

AMA1 IgGx (AU/mL)<br />

2048<br />

512<br />

128<br />

32<br />

8<br />

2<br />

0.5<br />

0.125<br />

0.03125<br />

IgG1<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

IgG2<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Treatment<br />

IgG3<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Figure 6. IgG subclasses to the vaccine antigen in serum obtained on day 84, boxes<br />

indicate median <strong>and</strong> quartile ranges. Treatment groups are indicated by Adjuvant<br />

name <strong>and</strong> AMA1 dose, respectively.<br />

0.0011, Tukey HSD). A similar trend, albeit not significant, was observed when<br />

comparing the Montanide ISA 720 50 µg group with the AS02 50 µg group<br />

(7.2 % difference, 95% CI -0.17 to 14.5, p = 0.059, Tukey HSD). This suggests<br />

that Montanide ISA 720 induces more antibodies directed <strong>against</strong> domain III. No<br />

significant differences were observed between the treatment groups following<br />

depletion with domains II-III or domain II only (p values 0.16 <strong>and</strong> 0.051,<br />

respectively, ANOVA). The average amount of IgG remaining bound was 21.1%<br />

(95% CI 19.2 to 23.0) <strong>and</strong> 61.5% (95% CI 57.1 to 66.0) for domains II-III <strong>and</strong><br />

domain II, respectively.<br />

Antibody Avidity<br />

Antibody avidity in plasma samples collected on day 84 was quantified using a<br />

sodium thiocyanate (NaSCN) elution ELISA <strong>and</strong> expressed as the concentration<br />

of NaSCN required to reduce the amount of bound antibody by 50%, shown in<br />

Figure 5. Adjuvant or antigen dose did not significantly influence the average<br />

avidity to the AMA1 variants under investigation (p=0.169, p=0.725, p=0.864 <strong>and</strong><br />

p=0.609 for FVO, 3D7, HB3 <strong>and</strong> CAMP, respectively). The average avidity was<br />

0.694 M NaSCN (95% CI 0.660 to 0.725) for the homologous FVO AMA1. Average<br />

avidities for the heterologous antigens were: 0.609 M (95% CI 0.542 to 0.676),<br />

IgG4<br />

69


70 Chapter 3<br />

0.711 M (95% CI 0.602 to 0.820) <strong>and</strong> 0.929 M (95% CI 0.775 to 1.083) for 3D7,<br />

HB3 <strong>and</strong> CAMP AMA1, respectively. Antibody avidity to the 3D7 AMA1 was<br />

significantly lower than to the homologous FVO AMA1 (0.085 M NaSCN, 95% CI<br />

0.025 to 0.1435, p = 0.006, paired t-test). By contrast, antibody avidity to the<br />

CAMP AMA1 was significantly higher than to the homologous FVO AMA1 (0.235<br />

M NaSCN, 95% CI 0.0885 to 0.3821, p = 0.002, paired t-test). Antibody avidities<br />

to the HB3 allele were similar to those observed for the FVO allele (p = 0.742,<br />

paired t-test).<br />

Subclass distribution<br />

IgG subclass levels to the homologous FVO AMA1 allele determined in plasma<br />

samples taken on day 84 are shown in Figure 6. The ranking of IgG subclasses in<br />

descending titre order was IgG1 > IgG3 ≈ IgG4 > IgG2. IgG1 <strong>and</strong> IgG3 antibody<br />

levels showed a similar pattern as to what was observed for total IgG, with<br />

lowest titres in the Alhydrogel groups (10 <strong>and</strong> 50 µg) <strong>and</strong> in the Montanide ISA<br />

720 10 µg group as compared to the Montanide ISA 720 50 µg <strong>and</strong> both AS02<br />

groups (Figure 6). Interestingly, titre differences between the treatment arms<br />

were slightly higher for IgG3 as compared to IgG1 (Figure 6).<br />

GIA<br />

The results of the Growth Inhibition Assay (GIA) for three laboratory strains<br />

obtained with IgG fractions purified from plasma samples collected at day 84 are<br />

shown in Figure 7. GIA titres determined on the FCR3 laboratory strain<br />

expressing an AMA1 similar to the vaccine antigen were below 20% in most<br />

(22/27) subjects in the Alhydrogel (10 <strong>and</strong> 50 µg) <strong>and</strong> Montanide ISA 720 10 µg<br />

groups; only 5 out of 27 subjects had GIA titres higher than 20% at 10 mg/ ml<br />

total IgG. By contrast, 14 out of 20 subjects in the Montanide ISA 720 (50 µg)<br />

<strong>and</strong> AS02 (10 <strong>and</strong> 50 µg) groups had GIA titres exceeding 20% at 10 mg /ml total<br />

IgG (Figure 7). Average GIA responses were significantly higher (38.7 %-points,<br />

95% CI 4.7 to 72.6%, p = 0.018, Tukey HSD) in the AS02 10 µg group as compared<br />

to the Alhydrogel 10 µg group. The GIA values at 10 mg /ml total IgG in<br />

Montanide ISA 720 <strong>and</strong> AS02 50 µg groups were also higher than the Alhydrogel<br />

10 µg group, but this failed to achieve statistical significance (p=0.17 <strong>and</strong> 0.20,<br />

respectively, Tukey HSD, Figure 7). The GIA titres determined on the FCR3 strain<br />

at 5 mg/ ml total IgG were lower <strong>and</strong> showed a ranking similar to what was<br />

observed at 10 mg/ ml, but none of the between group comparisons reached<br />

statistical significance (p=0.070, ANOVA, Figure 7).


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

Growth Inhibition (%)<br />

80<br />

60<br />

40<br />

20<br />

0<br />

−20<br />

FCR3<br />

400<br />

800<br />

1600<br />

3200<br />

6400<br />

12800<br />

25600<br />

51200<br />

102400<br />

3D7<br />

400<br />

800<br />

1600<br />

3200<br />

6400<br />

12800<br />

25600<br />

51200<br />

AMA1 IgG (AU/mL)<br />

102400<br />

400<br />

800<br />

1600<br />

3200<br />

6400<br />

12800<br />

25600<br />

51200<br />

102400<br />

Figure 8. Relation between AMA1-specific IgG titre <strong>and</strong> GIA titre for the<br />

homologous (FCR3) <strong>and</strong> two heterologous strains (3D7 <strong>and</strong> HB3). The GIA value<br />

for the designated strain measured at 10 mg /mL total IgG is depicted on the yaxis<br />

<strong>and</strong> the corresponding Elisa titre to the corresponding AMA1 allele is<br />

depicted on the x-axis. Estimated IC50 values are indicated by dashed lines.<br />

Symbols represent treatment groups: white circles = Alhydrogel 10 µg, black<br />

circles = Alhydrogel 50 µg, white squares = Montanide ISA 720 10 µg, black<br />

squares = Montanide ISA 720 10 µg, white diamonds = AS02 10 µg <strong>and</strong> black<br />

diamonds = AS02 50 µg, where the 10 or 50 µg refers to the AMA1 dose.<br />

GIA titres determined on the 3D7 laboratory strain at 10 mg/ml IgG were lower<br />

than those to the FCR3 strain at the same total IgG concentration <strong>and</strong> no<br />

significant differences were observed between the treatment groups (p=0.095,<br />

ANOVA). Interestingly, GIA titres on the 3D7 strain at 10 mg/ml were similar to<br />

GIA titres to the homologous FCR3 strain at 5 mg/ml total IgG (Figure 7). GIA<br />

titres on the 3D7 laboratory strain at 5 mg/ml IgG were low; only 2 out of 47<br />

subjects had GIA titres higher than 20% (Figure 7).<br />

GIA titres determined on the HB3 laboratory strain at 10 mg /ml IgG were lower<br />

than to the FCR3 strain <strong>and</strong> similar to the titres observed for the 3D7 strain.<br />

Significant treatment group differences were observed (p=0.004, ANOVA). GIA<br />

titres in the AS02 10 µg group were significantly higher than in the Alhydrogel 10<br />

µg group (24.6 %-points, 95% CI 3.4 to 45.8%, p = 0.014, Tukey HSD) <strong>and</strong> GIA<br />

titres in the AS02 10 µg group tended to be higher as compared to the<br />

Alhydrogel 50 µg group (20.5%-points, 95% CI -0.2 to 41.3%, p = 0.053, Tukey<br />

HB3<br />

71


72 Chapter 3<br />

GIA (%)<br />

80<br />

60<br />

40<br />

20<br />

0<br />

−20<br />

80<br />

60<br />

40<br />

20<br />

0<br />

−20<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

10<br />

FCR3<br />

5<br />

FCR3<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

10<br />

3D7<br />

5<br />

3D7<br />

ISA 720 10<br />

ISA 720 50<br />

Treatment<br />

AS02 10<br />

AS02 50<br />

Alhydrogel 10<br />

Alhydrogel 50<br />

10<br />

HB3<br />

5<br />

HB3<br />

. Figure 7. GIA to 3 laboratory strains (FCR3, homologous <strong>and</strong> 3D7 <strong>and</strong> HB3,<br />

heterologous) at 10 <strong>and</strong> 5 mg /ml total IgG obtained at day 84. Top row: GIA at<br />

10 mg /ml <strong>and</strong> bottom row GIA at 5 mg /ml IgG. Boxes indicate median <strong>and</strong><br />

quartile ranges. Treatment groups are indicated by Adjuvant name <strong>and</strong> AMA1<br />

dose, respectively<br />

ISA 720 10<br />

ISA 720 50<br />

AS02 10<br />

AS02 50


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

HSD) <strong>and</strong> the Montanide ISA 720 10 µg group (21.2%-points, 95% CI -0.6 to<br />

42.9%, p = 0.061, Tukey HSD). GIA titres on the HB3 laboratory strain at 5 mg /ml<br />

total IgG were low, with only 4 out of 47 subjects having GIA titres of over 20%.<br />

GIA titres on the HB3 laboratory strain at 5 mg /ml IgG in the AS02 10 µg group<br />

tended to be higher, similar to what was observed at 10 mg ml total IgG, but this<br />

failed to reach statistical significance (p=0.091, ANOVA).<br />

Relation between IgG <strong>and</strong> GIA titres<br />

The relation between AMA1 variant-specific IgG <strong>and</strong> GIA was investigated by<br />

modelling sigmoid curves using non-linear least squares regression. The data are<br />

graphically presented in Figure 8. The correlation coefficients were 0.911 (95%<br />

CI 0.845 to 0.950, p < 0.0001) for FCR3, 0.795 (95% CI 0.658 to 0.8810, p <<br />

0.0001) for 3D7 <strong>and</strong> 0.794 (95% CI 0.657 to 0.881, p < 0.0001) for HB3. The IC50<br />

values, i.e. the amount of AMA1 specific IgG required for 50% inhibition were<br />

estimated at 31.3 kAU/ ml (95% CI 27.7 to 35.4) for FCR3, 39.8 kAU/ ml (95% CI<br />

30.8 to 51.3) for 3D7 <strong>and</strong> 38.9 kAU/ ml (95% CI 28.9 to 52.5) for HB3 (Figure 8),<br />

which was not significantly different from the value for the FCR3 strain (p = 0.08<br />

<strong>and</strong> 0.20, respectively). The fold-increase in AMA1 variant specific IgG required<br />

to raise the GIA titre from 10 to 50% or from 50 to 90% was 3.5-fold (95% CI 2.7<br />

to 5.0) for FCR3, 2.9-fold (95% CI 2.2 to 5.7) for 3D7 <strong>and</strong> 4.5-fold (95% CI 3.1 to<br />

9.5) for HB3, the values for the 3D7 <strong>and</strong> HB3 strains were not significantly<br />

different from the value for the FCR3 strain (p = 0.50 <strong>and</strong> p = 0.33, respectively).<br />

Discussion<br />

The main finding of this study is that AMA1 antibody responses induced by<br />

vaccination of malaria-naïve human volunteers with a single AMA1 variant are<br />

biased towards the vaccine allele. This is in agreement with previously published<br />

results from a similar human Phase I vaccine trial [22] <strong>and</strong> observations in<br />

rabbits [14,16]. Approximately half of the IgG induced by vaccination of human<br />

volunteers reacts in ELISA with heterologous AMA1 variants, similar to what has<br />

been observed in rabbits [14,16]. The functionality of the antibody response<br />

(GIA titre) to heterologous AMA1 expressing strains reflects differences in the<br />

quantity of IgG; suggesting that, although absolute antibody levels to<br />

heterologous AMA1 variants are lower, these antibodies are, on per mass basis,<br />

equally capable of inhibiting parasite growth.<br />

73


74 Chapter 3<br />

The data presented here also demonstrate that both adjuvant <strong>and</strong> dose can<br />

have a profound impact on antibody levels, while other aspects of the antibody<br />

response like avidity, domain recognition, breadth <strong>and</strong> subclass distribution, are<br />

much less influenced by adjuvant or dose. This is, with exception of the subclass<br />

distribution, in agreement with what has been observed in rabbit studies [23].<br />

The data presented here show that the widely used adjuvant Alhydrogel yields<br />

relatively low antibody levels, <strong>and</strong> increasing the antigen dose from 10 to 50 µg<br />

only marginally improved antibody responses. The data confirm that Alhydrogel<br />

is not potent enough to induce high levels of functional antibodies in malaria<br />

naïve subjects, as was previously reported [24,25]. The low potency of<br />

Alhydrogel at inducing a functional response is even more pronounced when the<br />

functionality of the antibody response is evaluated on strains expressing<br />

heterologous AMA1 alleles. Therefore adjuvants more potent than Alhydrogel<br />

are definitively required for the induction of functional antibody responses to<br />

AMA1 variants not included in a vaccine. Compared to Alhydrogel, the water in<br />

oil emulsion Montanide ISA 720, appeared to yield improved antibody responses<br />

when combined with a 50 µg AMA1 dose, but also gave increased numbers of<br />

adverse events [19]. Moreover, Montanide ISA 720 can chemically modify the<br />

vaccine antigen [26], potentially resulting in loss of potency <strong>and</strong> thus vaccine<br />

failure [27]. The proprietary adjuvant AS02 yielded relatively high (functional)<br />

antibody responses at both antigen doses tested, in agreement with previously<br />

published results [22, 28].<br />

Recently, vaccination induced efficacy <strong>against</strong> the AMA1 vaccine allele<br />

formulated with AS02 in a Phase IIb study was estimated at 64%, whereas<br />

overall vaccine efficacy was estimated at 17%, indicating the importance of<br />

covering AMA1 variants [18]. Thus, the challenge for an AMA1-based vaccine<br />

appears to be with covering AMA1 polymorphism. Theoretically, two options<br />

would be available: i) The induction of a broad antibody response <strong>and</strong> ii) The<br />

maximisation of heterologous antibody responses by using potent adjuvants or<br />

prime boost strategies combined with a potent adjuvant, such that the response<br />

induced with a single variant would be high enough to also be sufficiently<br />

functional <strong>against</strong> heterologous strains. As the latter may not be possible with<br />

adjuvants currently available, a more practical approach would be the<br />

combination of the induction of the broadest response possible with the highest<br />

response possible.


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

The breadth of the antibody response can be improved by vaccination with a<br />

mixture of several AMA1 variants, either naturally occurring [14, 16, 23, 29], or<br />

artificial ones [10, 29]. Three artificial diversity covering (DiCo) AMA1 sequences<br />

[10] have recently been produced under cGMP <strong>and</strong> will enter clinical testing in<br />

the near future.<br />

The magnitude of the antibody response can be improved by the use of potent<br />

adjuvants, as shown here. A novel proprietary adjuvant, CoVaccine HT, has<br />

yielded promising results in rhesus macaques <strong>and</strong> rabbits [23, 30]. Moreover, P.<br />

knowlesi AMA1 formulated with CoVaccine HT has induced <strong>protection</strong> <strong>against</strong><br />

blood-stage challenge with P. knowlesi in the rhesus macaque model <strong>and</strong> the<br />

degree of <strong>protection</strong> was correlated with GIA titre [31]. It would therefore be<br />

interesting to combine DiCo AMA1 with CoVaccine HT in a Phase I trial.<br />

Antibody avidity was determined with a sodium thiocyanate elution ELISA <strong>and</strong><br />

average antibody avidities ranged between 0.6 <strong>and</strong> 0.9 M for the various<br />

antigens. These values are lower than what was previously observed in rabbits<br />

(0.9 to 1.4 M) [16]. Of note is that the average avidity to the heterologous CAMP<br />

AMA1 was higher than the homologous antigen. This could possibly be explained<br />

by the fact that half of FVO AMA1-specific antibodies bound to CAMP AMA1 <strong>and</strong><br />

that this fraction may bind with higher avidity. Conversely, avidities to the<br />

heterologous 3D7 AMA1 were lower than the homologous avidities. This may<br />

represent antigenic relatedness of the respective AMA1 molecules, with CAMP<br />

being more close to FVO AMA1 <strong>and</strong> 3D7 more distant.<br />

The antibody response to AMA1 appears to be mainly directed <strong>against</strong> domains I<br />

<strong>and</strong> II, as competition with a construct comprising these domains removes about<br />

80% of antibodies bound. Competition with a domain II-III construct removes a<br />

similar amount of antibodies as the I-II construct, suggesting that the majority of<br />

the response would be directed <strong>against</strong> domain II. This is, however, not<br />

supported by the depletion obtained by a domain II construct. Of note here is<br />

that only constructs including domains I-II induce functionally active antibodies<br />

in rabbits [32, 33], suggesting conformational authenticity. The Domain II-III <strong>and</strong><br />

the domain II constructs both failed to elicit functionally active antibodies in<br />

rabbits [32, 33]. The results obtained here warrant the statement that the<br />

majority of the antibody response to AMA1 is directed <strong>against</strong> domains I <strong>and</strong> II<br />

<strong>and</strong> a further subdivision for these two domains is not possible with the data<br />

hitherto obtained. The importance of domains I <strong>and</strong> II in the antibody response<br />

is in agreement with what has been found in rabbits [32,33].<br />

75


76 Chapter 3<br />

The subclass distribution found in vaccinated malaria naive volunteers was<br />

similar to what was observed in exposed children [34, 35] <strong>and</strong> reflects the<br />

expected subclass distribution for a protein antigen [36]. Antigen dose or<br />

adjuvant only marginally influenced the subclass distribution.<br />

In conclusion, vaccination with a single allele AMA1 vaccine induces a humoral<br />

immune response that is biased towards the vaccine allele. The magnitude of<br />

the response can be enhanced by a potent adjuvant, in contrast other<br />

parameters of the humoral response like breadth, avidity <strong>and</strong> subclass<br />

distribution appear much less influenced by the adjuvant. Future vaccine<br />

development should focus on improving both breadth <strong>and</strong> magnitude of<br />

antibody responses to AMA1.<br />

Acknowledgements<br />

The authors wish to thank Glaxo Smith Kline, Rixensart, Belgium for supplying<br />

AS02 <strong>and</strong> SEPPIC, Paris, France for supplying Montanide ISA 720.<br />

Funding<br />

This work was supported by a grant from the European Malaria Vaccine<br />

Initiative.


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

References<br />

1. Snow RW, Guerra CA, Noor AM, Myint HY, Hay SI. The global distribution of<br />

clinical episodes of Plasmodium falciparum malaria. Nature 2005; 434:214-217.<br />

2. Remarque EJ, Faber BW, Kocken CH, Thomas AW. Apical membrane antigen 1: a<br />

malaria vaccine c<strong>and</strong>idate in review. Trends Parasitol 2008; 24:74-84.<br />

3. Bannister LH, Hopkins JM, Dluzewski AR, Margos G, Williams IT et al.<br />

Plasmodium falciparum apical membrane antigen 1 (PfAMA-1) is translocated<br />

within micronemes along subpellicular microtubules during merozoite<br />

development. J Cell Sci 2003; 116:3825-3834.<br />

4. Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A et al. A role for apical<br />

membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum<br />

sporozoites. J Biol Chem 2004; 279:9490-9496.<br />

5. Healer J, Crawford S, Ralph S, McFadden G, Cowman AF. Independent<br />

translocation of two micronemal proteins in developing Plasmodium falciparum<br />

merozoites. Infect Immun 2002; 70:5751-5758.<br />

6. Howell SA, Withers-Martinez C, Kocken CH, Thomas AW, Blackman MJ.<br />

Proteolytic processing <strong>and</strong> primary structure of Plasmodium falciparum apical<br />

membrane antigen-1. J Biol Chem 2001; 276:31311-31320.<br />

7. Chesne-Seck ML, Pizarro JC, Vulliez-Le Norm<strong>and</strong> B, Collins CR, Blackman MJ et<br />

al. Structural comparison of apical membrane antigen 1 orthologues <strong>and</strong><br />

paralogues in apicomplexan parasites. Mol Biochem Parasitol 2005; 144:55-67.<br />

8. Pizarro JC, Vulliez-Le Norm<strong>and</strong> B, Chesne-Seck ML, Collins CR, Withers-Martinez<br />

C et al. Crystal structure of the malaria vaccine c<strong>and</strong>idate apical membrane<br />

antigen 1. Science 2005; 308:408-411.<br />

9. Mitchell GH, Thomas AW, Margos G, Dluzewski AR, Bannister LH. Apical<br />

membrane antigen 1, a major malaria vaccine c<strong>and</strong>idate, mediates the close<br />

attachment of invasive merozoites to host red blood cells. Infect Immun 2004;<br />

72:154-158.<br />

10. Remarque EJ, Faber BW, Kocken CH, Thomas AW. A diversity-covering approach<br />

to immunization with Plasmodium falciparum apical membrane antigen 1<br />

induces broader allelic recognition <strong>and</strong> growth inhibition responses in rabbits.<br />

Infect Immun 2008; 76:2660-2670.<br />

11. Bai T, Becker M, Gupta A, Strike P, Murphy VJ et al. Structure of AMA1 from<br />

Plasmodium falciparum reveals a clustering of polymorphisms that surround a<br />

conserved hydrophobic pocket. Proc Natl Acad Sci U S A 2005; 102:12736-<br />

12741.<br />

12. Crewther PE, Matthew ML, Flegg RH, Anders RF. Protective immune responses<br />

to apical membrane antigen 1 of Plasmodium chabaudi involve recognition of<br />

strain-specific epitopes. Infect Immun 1996; 64:3310-3317.<br />

13. Healer J, Murphy V, Hodder AN, Masciantonio R, Gemmill AW et al. Allelic<br />

polymorphisms in apical membrane antigen-1 are responsible for evasion of<br />

antibody-mediated inhibition in Plasmodium falciparum. Mol Microbiol 2004;<br />

52:159-168.<br />

77


78 Chapter 3<br />

14. Kennedy MC, Wang J, Zhang Y, Miles AP, Chitsaz F et al. In vitro studies with<br />

recombinant Plasmodium falciparum apical membrane antigen 1 (AMA1):<br />

production <strong>and</strong> activity of an AMA1 vaccine <strong>and</strong> generation of a multiallelic<br />

response. Infect Immun 2002; 70:6948-6960.<br />

15. Kocken CHM, Withers-Martinez C, Dubbeld MA, van der Wel A, Hackett F et al.<br />

High-level expression of the malaria blood-stage vaccine c<strong>and</strong>idate Plasmodium<br />

falciparum apical membrane antigen 1 <strong>and</strong> induction of antibodies that inhibit<br />

erythrocyte invasion. Infect Immun 2002; 70:4471-4476.<br />

16. Kusi KA, Faber BW, Thomas AW, Remarque EJ. Humoral immune response to<br />

mixed PfAMA1 alleles; multivalent PfAMA1 vaccines induce broad specificity.<br />

PLoS One 2009; 4:e8110.<br />

17. Dutta S, Lee SY, Batchelor AH, Lanar DE. Structural basis of antigenic escape of a<br />

malaria vaccine c<strong>and</strong>idate. Proc Natl Acad Sci U S A 2007; 104:12488-12493.<br />

18. Thera MA, Doumbo OK, Coulibaly D, Laurens MB, Ouattara A et al. A Field Trial<br />

to Assess a Blood-Stage Malaria Vaccine. New Engl<strong>and</strong> Journal of Medicine<br />

2011; 365:1004-1013.<br />

19. Roestenberg M, Remarque E, de Jonge E, Hermsen R, Blythman H et al. Safety<br />

<strong>and</strong> immunogenicity of a recombinant Plasmodium falciparum AMA1 malaria<br />

vaccine adjuvanted with Alhydrogel, Montanide ISA 720 or AS02. PLoS One<br />

2008; 3:e3960.<br />

20. Faber BW, Remarque EJ, Kocken CH, Cheront P, Cingolani D et al. Production,<br />

quality control, stability <strong>and</strong> pharmacotoxicity of cGMP-produced Plasmodium<br />

falciparum AMA1 FVO strain ectodomain expressed in Pichia pastoris. Vaccine<br />

2008; 26:6143-6150.<br />

21. Introduction to the immune system. In: Kasper DL, Braunwald E, Fauci AS,<br />

Hauser SL, Longo DL et al., editors. Harrison's principles of internal medicine.<br />

New York: McGraw-Hill. pp. 1907-1930.<br />

22. Spring MD, Cummings JF, Ockenhouse CF, Dutta S, Reidler R et al. Phase 1/2a<br />

study of the malaria vaccine c<strong>and</strong>idate apical membrane antigen-1 (AMA-1)<br />

administered in adjuvant system AS01B or AS02A. PLoS One 2009; 4:e5254.<br />

23. Kusi KA, Faber BW, Riasat V, Thomas AW, Kocken CH et al. Generation of<br />

humoral immune responses to multi-allele PfAMA1 vaccines; effect of adjuvant<br />

<strong>and</strong> number of component alleles on the breadth of response. PLoS One 2010;<br />

5:e15391.<br />

24. Malkin EM, Diemert DJ, McArthur JH, Perreault JR, Miles AP et al. Phase 1<br />

clinical trial of apical membrane antigen 1: an asexual blood-stage vaccine for<br />

Plasmodium falciparum malaria. Infect Immun 2005; 73:3677-3685.<br />

25. Mullen GE, Ellis RD, Miura K, Malkin E, Nolan C et al. Phase 1 trial of AMA1-<br />

C1/Alhydrogel plus CPG 7909: an asexual blood-stage vaccine for Plasmodium<br />

falciparum malaria. PLoS One 2008; 3:e2940.<br />

26. Miles AP, McClellan HA, Rausch KM, Zhu D, Whitmore MD et al. Montanide ISA<br />

720 vaccines: quality control of emulsions, stability of formulated antigens, <strong>and</strong><br />

comparative immunogenicity of vaccine formulations. Vaccine 2005; 23:2530-<br />

2539.


Humoral immune responses to a single allele PfAMA1 vaccine in healthy malarianaïve<br />

adults.<br />

27. Saul A, Lawrence G, Allworth A, Elliott S, Anderson K et al. A human phase 1<br />

vaccine clinical trial of the Plasmodium falciparum malaria vaccine c<strong>and</strong>idate<br />

apical membrane antigen 1 in Montanide ISA720 adjuvant. Vaccine 2005;<br />

23:3076-3083.<br />

28. Polhemus ME, Magill AJ, Cummings JF, Kester KE, Ockenhouse CF et al. Phase I<br />

dose escalation safety <strong>and</strong> immunogenicity trial of Plasmodium falciparum<br />

apical membrane protein (AMA-1) FMP2.1, adjuvanted with AS02A, in malarianaive<br />

adults at the Walter Reed Army Institute of Research. Vaccine 2007;<br />

25:4203-4212.<br />

29. Kusi KA, Faber BW, van der Eijk M, Thomas AW, Kocken CH et al. Immunization<br />

with different PfAMA1 alleles in sequence induces clonal imprint humoral<br />

responses that are similar to responses induced by the same alleles as a vaccine<br />

cocktail in rabbits. Malar J 2011; 10:40.<br />

30. Draper SJ, Biswas S, Spencer AJ, Remarque EJ, Capone S et al. Enhancing bloodstage<br />

malaria subunit vaccine immunogenicity in rhesus macaques by<br />

combining adenovirus, poxvirus, <strong>and</strong> protein-in-adjuvant vaccines. J Immunol<br />

2010; 185:7583-7595.<br />

31. Mahdi Abdel Hamid M, Remarque EJ, van Duivenvoorde LM, van der Werff N,<br />

Walraven V et al. Vaccination with Plasmodium knowlesi AMA1 Formulated in<br />

the Novel Adjuvant Co-Vaccine HT Protects <strong>against</strong> Blood-Stage Challenge in<br />

Rhesus Macaques. PLoS One 2011; 6:e20547.<br />

32. Lalitha PV, Ware LA, Barbosa A, Dutta S, Moch JK et al. Production of the<br />

subdomains of the Plasmodium falciparum apical membrane antigen 1<br />

ectodomain <strong>and</strong> analysis of the immune response. Infect Immun 2004; 72:4464-<br />

4470.<br />

33. Faber BW, Remarque EJ, Morgan WD, Kocken CH, Holder AA et al. Malaria<br />

vaccine-related benefits of a single protein comprising Plasmodium falciparum<br />

apical membrane antigen 1 domains I <strong>and</strong> II fused to a modified form of the 19kilodalton<br />

C-terminal fragment of merozoite surface protein 1. Infect Immun<br />

2007; 75:5947-5955.<br />

34. Dodoo D, Aikins A, Kusi KA, Lamptey H, Remarque E et al. Cohort study of the<br />

association of antibody levels to AMA1, MSP1-19, MSP3 <strong>and</strong> GLURP with<br />

<strong>protection</strong> from clinical malaria in Ghanaian children. Malar J 2008; 7:142.<br />

35. Nebie I, Diarra A, Ouedraogo A, Soulama I, Bougouma EC et al. Humoral<br />

responses to Plasmodium falciparum blood-stage antigens <strong>and</strong> association with<br />

incidence of clinical malaria in children living in an area of seasonal malaria<br />

transmission in Burkina Faso, West Africa. Infect Immun 2008; 76:759-766.<br />

36. Hammarstrom L, Smith CIE. IgG subclass changes in response to vaccination. In:<br />

Skakib F, editors. Monographs in allergy (vol 19): Basic <strong>and</strong> clinical aspects of<br />

IgG subclasses. Basel: Karcher. 1986; pp. 241-251.<br />

79


Chapter 4<br />

A saturation of antibody avidity <strong>and</strong><br />

concentration induced by malaria vaccine<br />

c<strong>and</strong>idate Apical Membrane Antigen 1<br />

Meta Roestenberg 1 , Ed Remarque 2 , Bart Faber 2 , Clemens Kocken 2 , Alan<br />

Thomas 2 , Cornelus C. Hermsen 1 , Will Roeffen 1 , Robert W. Sauerwein 1<br />

1 Radboud University Nijmegen Medical Centre, department of Medical<br />

Microbiology 268, P.O Box 9101, 6500 HB, Nijmegen, The Netherl<strong>and</strong>s<br />

2 Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The<br />

Netherl<strong>and</strong>s<br />

Manuscript in preparation


Chapter 4 82<br />

Abstract<br />

The functional activity of antibodies is determined by their quantity <strong>and</strong> quality.<br />

Antibody avidity describes the quality of antigen-antibody binding, but this<br />

parameter has received only limited attention in vaccine design. The asexual<br />

erythrocytic stage antigen Apical Membrane Antigen 1 (AMA1) is a potential<br />

Plasmodium falciparum vaccine c<strong>and</strong>idate because of its critical role in red blood<br />

cell invasion. AMA1 induces antibody-mediated <strong>protection</strong> <strong>against</strong> <strong>infection</strong> in<br />

animals by blocking invasion of the parasite. Assuming a simple 1:1 interaction<br />

model of immunoglobulin with antigen, we determined anti-AMA1 antibody<br />

concentration <strong>and</strong> avidity in rabbit <strong>and</strong> human sera post-vaccination by Surface<br />

Plasmon Resonance (SPR) technology. We show that anti-AMA1 antibody<br />

concentration <strong>and</strong> avidity are inversely correlated <strong>and</strong> become saturated at a<br />

maximum fraction of bound antigen. The matured AMA1 antibody response,<br />

whether induced through vaccination or natural exposure, thus always has a<br />

maximum saturated, constant capacity to bind AMA1 antigen, although there is<br />

some interindividual variation in the balance between concentration <strong>and</strong> avidity.<br />

The functionality of these antibodies in vitro was tested by the parasite growth<br />

inhibition assay. Growth inhibition in vitro correlated with antibody<br />

concentration, but not with avidity. The relevance of these findings in vivo<br />

warrants further research, as the appreciation of a saturated antibody response<br />

may guide AMA1 vaccine development.


A saturation of antibody avidity <strong>and</strong> concentration induced by malaria vaccine<br />

c<strong>and</strong>idate Apical Membrane Antigen 1<br />

Introduction<br />

The functional activity of antibodies depends on epitope specificity, titre <strong>and</strong><br />

affinity. The avidity of antibodies defines the sum of the individual affinities<br />

between the antibody binding sites <strong>and</strong> single epitopes [1, 2] <strong>and</strong> is an<br />

important parameter defining antibody quality. Both concentration <strong>and</strong> avidity<br />

of antibodies should be addressed in vaccines that depend on humoral<br />

responses to convey <strong>protection</strong>. One c<strong>and</strong>idate immunogen is Plasmodium<br />

falciparum Apical Membrane Antigen 1 (PfAMA1). Located at the surface of<br />

blood-stage malaria parasites, AMA1 plays an essential role in the process of red<br />

blood cell invasion [3-5]. Functional antibody binding has been shown to prevent<br />

parasite invasion [6], protecting mice <strong>and</strong> monkeys <strong>against</strong> parasite<br />

multiplication <strong>and</strong> disease [7]. The full-length ectodomain recombinant PfAMA1<br />

(amino acids 25-545) of the Plasmodium falciparum FVO strain has been<br />

expressed under cGCP in Pichia pastoris [8]. The product was formulated with<br />

Alhydrogel, Montanide ISA 720 <strong>and</strong> AS02A <strong>and</strong> tested for safety <strong>and</strong><br />

immunogenicity in humans [9]. We investigated the concentration <strong>and</strong> avidity of<br />

antibodies induced by recombinant AMA1 in rabbits <strong>and</strong> human immunizations<br />

studies by Surface Plasmon Resonance. We correlated these data with the<br />

capacity of sera to functionally inhibit parasite growth in vitro.<br />

Materials <strong>and</strong> Methods<br />

Antigens<br />

Six different recombinant PfAMA1 constructs were prepared as described<br />

previously [10]. Pf3mH comprises the prosequence, subdomain I <strong>and</strong> subdomain<br />

II, Pf8mH comprises subdomain II only, Pf9mH comprises subdomain II <strong>and</strong><br />

subdomain III <strong>and</strong> Pf10mH comprises subdomain III only, covering amino-acid<br />

residues 25-442, 303-442, 303-544 <strong>and</strong> 419-544 of the FVO AMA1 antigen<br />

respectively (Figure 1). The Pf4mH construct comprises the prosequence,<br />

subdomain I, subdomain II <strong>and</strong> subdomain III, representing residues 25 to 544<br />

from the Pf FVO strain as described by Kocken et al. [11]. Construct Pf11.0<br />

comprises amino-acid residues 25-545, but lacks the additional Myc-His tag<br />

(Figure 1). Based on rabbit immunization studies, the latter construct was used<br />

for a human phase I trial [9].<br />

83


84 Chapter 4<br />

Figure 1. Schematic representation of the different AMA1 antigen<br />

constructs used for the Rabbit immunizations. Construct Pf11.0 was<br />

used for the Phase I clinical trial.<br />

Sera<br />

Immunizations of rabbits were performed as described previously [11]. Briefly,<br />

injection of 100 μg of purified FVO strain AMA1 antigen with Freund's complete<br />

adjuvant was followed by three booster injections of 100 μg AMA1 at days 14,<br />

28, <strong>and</strong> 56 with Freund's incomplete adjuvant. Rabbits were immunized with the<br />

different AMA1 constructs shown in Figure 1. A total number of 18 rabbits were<br />

immunized, with minimum sets of 2 rabbits receiving the same recombinant<br />

antigen. Antisera obtained 4 weeks after the last injection were used for<br />

antibody evaluation. Sera from rabbits immunized with the same antigen were<br />

pooled to obtain enough material for kinetic analysis. Pre-immunization sera<br />

were used as a negative control.<br />

Human sera were obtained from a previously published phase I trial [9]. In short,<br />

healthy malaria-naïve Dutch volunteers were immunized with either 10 or 50µg<br />

of PfAMA1-FVO [25-545] adjuvanted with either Alhydrogel TM , Montanide ISA<br />

720 (SEPPIC, Paris, France) or AS02A (a proprietary Adjuvant System from<br />

GlaxoSmithKline Biologicals) at three occasions with monthly intervals. Sera<br />

were obtained one month after the third immunisation. For SPR analysis<br />

immunoglobulins (Ig) from a r<strong>and</strong>om selection of six human sera from every trial<br />

arm (18 total) were purified according to manufacturer’s instructions using<br />

HiTrap TM MabSelect SuRe TM (GE Healthcare, Diegem, Belgium) <strong>and</strong> concentrated<br />

to their original volume by Vivaspin® 20 (GE Healthcare, Diegem, Belgium). Sera<br />

from ten volunteers living in malaria endemic areas in Burkina Faso, Came-


A saturation of antibody avidity <strong>and</strong> concentration induced by malaria vaccine<br />

c<strong>and</strong>idate Apical Membrane Antigen 1<br />

Figure 2. Example of a BIAcore sensorgram. The sensorgram shows<br />

immobilization of Pf4mH starting at 1, injection of a serum sample at 2<br />

<strong>and</strong> initiation of regeneration with glycine pH2.0 at 3.<br />

roon, Gambia, Madagascar, Senegal <strong>and</strong> Tanzania, previously obtained for other<br />

purposes, were pooled for analysis.<br />

Surface Plasmon Resonance (SPR) analysis<br />

A Biacore TM 2000 (Biacore International SA, Uppsala, Sweden) was used for SPR<br />

measurements at 25°C using CM5 chips (Carboxylmethylated dextrane, Biacore<br />

International SA, Uppsala, Sweden). Rabbit anti-myc antibodies (PA1-981 Affinity<br />

BioReagents, Golden USA) were immobilized until approximately 13,000<br />

resonance units (RU) using st<strong>and</strong>ard amine coupling in 10 mM acetate pH 5.0.<br />

according to manufacturer’s instructions, after which the immobilized surface<br />

was washed with injections of 50 mM HCl. All subsequent measurements on the<br />

biosensor were performed in duplicate. Pre-immunization sera were used as a<br />

negative control. Correction for non-specific binding was performed by<br />

subtraction of the control channel.<br />

Chips were coated with approximately 350 RU Pf4mH construct. Samples were<br />

serially diluted ranging from 1:25 to 1:800, <strong>and</strong> 25 µl of diluted sample were<br />

injected at 5 µl/min. Measurement of antibody concentrations was performed<br />

by evaluating the association rate 25 seconds after initiation injection,<br />

measuring the concentration of binding antibodies only. Regeneration of the<br />

anti-myc antibody chip surface was performed with 50 µl glycine pH 2.0 at a flow<br />

rate of 30µl/min. Calibration curves were prepared with the rat monoclonal<br />

antibody 4G2 [12].<br />

For analysis of antibody avidity, Pf4mH was injected at 20 µl/min for 1.5 minute<br />

in the experimental flow cells to obtain a total immobilization level of approx<br />

85


86 Chapter 4<br />

Concentration<br />

(mg/ml, range)<br />

Rabbit 1.31 (0.58 – 2.98)<br />

Human<br />

0.066 (0.007 - 0.50)<br />

Mean ka<br />

(1/Ms, range)<br />

1.26x10 5<br />

(9.1x10 3 – 3.7x10 5 )<br />

1.50 x 10 5<br />

(1.19x10 4 – 6.22 x 10 5 )<br />

Mean kd<br />

(1/s, range)<br />

2.80x10 -3<br />

(1.10 x10 -3 – 5.53 x10 -3 )<br />

2.65 x 10 -3<br />

(2.41 – 9.45 x 10 -3 )<br />

Table 1. Mean antibody concentration, association constant (ka) <strong>and</strong><br />

dissociation constant (kd) for rabbit (n=38) <strong>and</strong> human (n=72) sera.<br />

1/Ms = per Molar per second, 1/s = per second.<br />

150 RU. The abovementioned dilution series of samples was injected at 30<br />

µl/min. An example of a Biacore sensorgram is provided in figure 2. The<br />

association rate constant (ka) <strong>and</strong> the dissociation rate constant (kd) were<br />

obtained by fitting the sensorgrams with the use of BIAevaluation software 3.2<br />

(Biacore International SA, Uppsala, Sweden). Curves were fitted according to a<br />

simple 1:1 Langmuir binding model, with correction for a linear drifting baseline<br />

to adjust for slow dissociation of the Pf4mH coat from anti-myc antibodies.<br />

Rabbit sera <strong>and</strong> human sera were each measured using one chip.<br />

Growth inhibition assay<br />

Antibodies used for parasite inhibition assays were purified on protein A<br />

columns (Immunopure Plus Pierce, St Louis, MO, USA) using st<strong>and</strong>ard protocols,<br />

exchanged into RPMI 1640 using Amicon Ultra-15 concentrators (30 kDa cut-off,<br />

Millipore, Irel<strong>and</strong>), filter-sterilised <strong>and</strong> stored at -20 o C until use. IgG<br />

concentrations were determined using a Nanodrop ND-1000 spectrophotometer<br />

(Nanodrop Technologies, Wilmington, DE, USA).<br />

Pf strain FCR3 was cultured in vitro using st<strong>and</strong>ard Pf culture techniques in an<br />

atmosphere of 5% CO2, 5% O2 <strong>and</strong> 90% N2. FCR3 AMA1 (accession no. M34553)<br />

differs by one amino acid in the pro-sequence from FVO AMA1 (accession no.<br />

AJ277646).<br />

The effect of purified IgG antibodies on parasite invasion was evaluated in<br />

triplicate using 96-well flat-bottom plates (Greiner Bio-One, Alphen a/d Rijn, The<br />

Netherl<strong>and</strong>s) with synchronized cultures of Pf schizonts at a starting parasitemia<br />

of 0.2-0.4% <strong>and</strong> a haematocrit of 2.0% in a final volume of 100 µL containing<br />

10% control non-immune human serum, 20 µg /ml gentamicin in RPMI 1640 <strong>and</strong><br />

5 mg/mL purified IgG. After 40 to 42 hours, cultures were resuspended, <strong>and</strong> 50<br />

µL was transferred into 200 µL ice-cold PBS. The cultures were then centrifuged,<br />

the supernatant removed <strong>and</strong> the plates were frozen. Inhibition of parasite<br />

growth was estimated using the pLDH assay as previously described [13].


A saturation of antibody avidity <strong>and</strong> concentration induced by malaria vaccine<br />

c<strong>and</strong>idate Apical Membrane Antigen 1<br />

Figure 3: AMA1 antibody concentration <strong>and</strong> association constant (ka) of pooled rabbit<br />

sera (A) or human sera (B). Dashed lines show inverse curve fit of data in panel A.<br />

A: Inverse correlation between concentration <strong>and</strong> ka of AMA1 antibodies (clear circles)<br />

<strong>and</strong> concentration <strong>and</strong> ka of antibodies measured <strong>against</strong> heterologous AMA1 sequences<br />

from strain 3D7 (grey) or HB3 (black)<br />

B: Inverse correlation between concentration <strong>and</strong> ka of AMA1 antibodies in naturally<br />

exposed semi-immunes (stars) <strong>and</strong> concentration <strong>and</strong> ka of AMA1 antibodies in human<br />

volunteers post-vaccination with FVO PfAMA1 [25-545] vaccine (construct Pf11.0)<br />

adjuvanted by Alhydrogel® (white), Montanide ISA 720 (grey) or AS02A (black).<br />

Parasite growth inhibition was calculated based on optical density (OD)<br />

measurements as follows: 100 - ((ODexperimental - ODbackground)/ (ODcontrol -<br />

ODbackground) x 100). IgG purified from pre- immunization plasma was used as a<br />

control <strong>and</strong> culture medium was used to measure the background OD.<br />

Statistical Analysis<br />

Statistical fitting of interaction kinetics was performed using BIAevaluation<br />

software 3.2, whereby the closeness of fit is represented by the statistical value<br />

chi2. Chi2 reduces to the average squared residual per data point. All other<br />

statistical analyses were performed in SPSS 16.0 (SPSS Inc.). For comparison<br />

between means of independent data sets the Kruskall-Wallis was used for three<br />

or more groups, the Mann-Whitney U for two independent groups <strong>and</strong> Wilcoxon<br />

singed ranks for paired samples. Non-parametric Spearman analyses were used<br />

to describe correlations. Curve fits were assessed by ANOVA. All statistical<br />

analysis were two-tailed <strong>and</strong> p values less than 0.05 were considered significant.<br />

Results<br />

Sera obtained from rabbit immunization studies <strong>and</strong> purified immunoglobulins<br />

(Ig) obtained from volunteers in a phase I clinical trial were tested for their<br />

87


88 Chapter 4<br />

Figure 4: Correlation between residue number of the vaccine construct <strong>and</strong> AMA1<br />

antibody concentration (A) or association constant ka (B) post-vaccination in rabbit sera.<br />

binding kinetics to the full length homologous ectodomain of recombinant<br />

PfAMA1 by Surface Plasmon Resonance (construct Pf4mH, Figure 1). Association<br />

constants (ka) <strong>and</strong> dissociation constants (kd) of immunoglobulin-antigen (IgAg)<br />

interactions were determined separately by fitting kinetic curves to a simple 1:1<br />

Langmuir binding model. Fits to the model showed an average chi2 of 2.98 for<br />

rabbit samples (n=38 measurements) <strong>and</strong> 0.85 for human samples (n=72<br />

measurements). Two rabbit samples could not be fitted with the model <strong>and</strong><br />

were excluded from analyses. Mean ka <strong>and</strong> kd values of all other measurements<br />

are shown in Table 1. Kinetic constants of rabbit <strong>and</strong> human samples covered a<br />

similar range. Values for kd were considerably smaller than for ka (~10 8 fold<br />

difference), reflecting a stable antigen-antibody complex.<br />

Antibody concentration (of binding antibodies) <strong>and</strong> ka values of pooled rabbit<br />

sera were inversely correlation (Figure 3A), approximated by inverse curve fit<br />

(p=0.002, R 2 = 0.871). The implication of these results are best illustrated by<br />

analysis of a simple non-covalent interaction system, whereby the increase in<br />

IgAg over time (δ[IgAg]/ δt) is described by concentration of immunoglobulin<br />

[Ig], antigen [Ag] <strong>and</strong> ka <strong>and</strong> kd as follows: δ[IgAg]/ δt = ka*[Ig]*[Ag] –<br />

kd*[IgAg].<br />

In a state of equilibrium, when the increase in immunoglobulin-antigen complex<br />

over time is zero, concentration <strong>and</strong> binding constants are balanced<br />

(ka*[Ig]*[Ag] = kd*[IgAg]). Algebraic rearrangement will result in ka/kd*[Ig] =<br />

[IgAg]/[Ag], where [IgAg]/[Ag] represents the fraction of antigen bound by<br />

antibody. Assuming kd is negligible, the observed inverse relationship in rabbit<br />

sera (Figure 3A) thus suggests that the fraction of bound antigen is constant <strong>and</strong><br />

identical for all investigated sera.


A saturation of antibody avidity <strong>and</strong> concentration induced by malaria vaccine<br />

c<strong>and</strong>idate Apical Membrane Antigen 1<br />

Figure 5. Correlation between in vitro growth inhibition <strong>and</strong> AMA1 antibody<br />

concentration (A) or association constant ka (B) in human sera post-vaccination with<br />

FVO PfAMA1 [25-545] vaccine (construct Pf11.0) adjuvanted by Alhydrogel® (white),<br />

Montanide ISA 720 (grey) or AS02A (black).<br />

The balance between concentration <strong>and</strong> ka was influenced by the vaccine<br />

construct, whereby the number of amino-acid residues of the antigen used for<br />

immunization was positively correlated with the induced concentration of<br />

antibodies <strong>and</strong> inversely related to ka (p=0.007 R 2 =0.89 <strong>and</strong> p=0.003, R 2 =-0.92<br />

respectively using Pf4mH for all measurements, Figure 4). PfAMA1-FVO<br />

subdomain I, being the largest subdomain, was the major contributor to this<br />

effect <strong>and</strong> immunizations using antigen constructs spanning the entire domain<br />

typically induced high concentrations <strong>and</strong> lower avidity antibodies (p=0.03 for<br />

both parameters). When the same sera were tested <strong>against</strong> PfAMA1 antigen<br />

from heterologous strains 3D7 <strong>and</strong> HB3, the total fraction of bound antigen was<br />

generally lower (Figure 3A). Antibodies binding the 3D7 strain PfAMA1 showed a<br />

(non-significant) trend towards lower ka, corresponding with an increased<br />

number of discordant residues (FVO vs 3D7 25 residues, FVO vs HB3 18 residues<br />

[14]).<br />

PfAMA1 antibodies from subjects participating in a phase I vaccine trial also<br />

adhered to the inverse relation similar to rabbit sera. Of note, a number of<br />

samples, particularly from the Alhydrogel TM adjuvanted group, showed a lower<br />

total fraction of bound antigen <strong>and</strong> did not follow this inverse correlation (Figure<br />

3B). We additionally studied human AMA1 antibodies from highly-exposed<br />

individuals living in malaria endemic areas <strong>and</strong> again found a similar inverse<br />

relationship between concentration <strong>and</strong> antibody avidity (Figure 3B).<br />

Finally, human post-immunization sera were tested for their capacity to inhibit<br />

asexual parasite growth in vitro. Growth inhibiting capacity of antibodies<br />

89


90 Chapter 4<br />

positively correlated with anti-AMA1 antibody concentration but not with the<br />

mean ka (p


A saturation of antibody avidity <strong>and</strong> concentration induced by malaria vaccine<br />

c<strong>and</strong>idate Apical Membrane Antigen 1<br />

thus establish an equilibrium depending on their competition for common<br />

resources, of which antigen is only one [17]. The availability of other resources,<br />

such as growth factors, interleukins <strong>and</strong> the presence of adhesion molecules<br />

may provide critical advantage preserving diversity <strong>and</strong> preventing dominance of<br />

one B-cell pool [18]. This is an interesting hypothesis, because it suggests that<br />

possible addition of resources to the antigen (such as cytokine adjuvants) would<br />

offer an opportunity to increase the specific B-cell population beyond the<br />

current maximum.<br />

Few sera did not reach the saturated level, particularly in the Alhydrogel®<br />

adjuvanted group. We hypothesize that the antibody response in these sera may<br />

not have undergone full maturation <strong>and</strong> have not (yet) reached a maximum<br />

fraction of bound antigen. Similarly, allergen-specific IgE from atopic children<br />

also lacked a negative relationship between concentration <strong>and</strong> avidity [19],<br />

whereas this was apparent for the more matured antibody responses in adults<br />

[16].<br />

These data are particularly relevant for vaccine development, since the avidity of<br />

antibodies may determine susceptibility to diseases such as the Respiratory<br />

Syncytial virus or meningococcal disease [20-22]. For AMA1 antibodies, we<br />

found a relation between in vitro parasite growth inhibition <strong>and</strong> antibody<br />

concentration, but not avidity. Caution should be taken, however, when<br />

concluding that the functionality of AMA1 antibodies in vitro seems to be<br />

governed primarily by concentration of antibodies, as epitope functionality has<br />

not been taken into account in this study. Particularly PfAMA1 domain I <strong>and</strong><br />

domain II antibodies are thought to harbour the major targets for inhibitory<br />

responses [7]. Moreover, the results of in vitro growth inhibition so far do not<br />

correlate with in vivo susceptibility [23].<br />

In conclusion, our data illustrate an inverse relation between concentration <strong>and</strong><br />

avidity, that becomes saturated at a maximum, constant capacity to bind<br />

antigen. Any increase in antibody concentration at this saturated level will be<br />

balanced by a decrease in antibody avidity. The appreciation of a saturated<br />

antibody response may be crucial for the future development of PfAMA1<br />

vaccine, since it provides guidance to the search for more effective antigens,<br />

adjuvants or delivery platforms.<br />

91


92 Chapter 4<br />

Acknowledgements<br />

The authors wish to thank GlaxoSmithKline Biologicals, Rixensart, Belgium for<br />

supplying AS02A <strong>and</strong> SEPPIC, Paris, France for supplying Montanide ISA 720.<br />

Funding<br />

This work was supported by a grant from the European Malaria Vaccine<br />

Initiative.


A saturation of antibody avidity <strong>and</strong> concentration induced by malaria vaccine<br />

c<strong>and</strong>idate Apical Membrane Antigen 1<br />

References<br />

1. Ferreira MU, Katzin AM. The assessment of antibody affinity distribution by<br />

thiocyanate elution: a simple dose-response approach. J Immunol Methods<br />

1995; 187:297-305.<br />

2. Kaattari SL, Zhang HL, Khor IW, Kaattari IM, Shapiro DA. Affinity maturation in<br />

trout: clonal dominance of high affinity antibodies late in the immune response.<br />

Dev Comp Immunol 2002; 26:191-200.<br />

3. Mitchell GH, Thomas AW, Margos G, Dluzewski AR, Bannister LH. Apical<br />

membrane antigen 1, a major malaria vaccine c<strong>and</strong>idate, mediates the close<br />

attachment of invasive merozoites to host red blood cells. Infect Immun 2004;<br />

72:154-8.<br />

4. Dutta S, Haynes JD, Barbosa A, et al. Mode of Action of Invasion-Inhibitory<br />

Antibodies Directed <strong>against</strong> Apical Membrane Antigen 1 of Plasmodium<br />

falciparum. Infect Immun 2005; 73:2116-22.<br />

5. Thomas AW, Deans JA, Mitchell GH, Alderson T, Cohen S. The Fab fragments of<br />

monoclonal IgG to a merozoite surface antigen inhibit Plasmodium knowlesi<br />

invasion of erythrocytes. Mol Biochem Parasitol 1984; 13:187-99.<br />

6. Treeck M, Tamborrini M, Daubenberger CA, Gilberger TW, Voss TS. Caught in<br />

action: mechanistic insights into antibody-mediated inhibition of Plasmodium<br />

merozoite invasion. Trends Parasitol 2009; 25:494-7.<br />

7. Remarque EJ, Faber BW, Kocken CH, Thomas AW. Apical membrane antigen 1: a<br />

malaria vaccine c<strong>and</strong>idate in review. Trends Parasitol 2008; 24:74-84.<br />

8. Faber BW, Remarque EJ, Kocken CH, et al. Production, quality control, stability<br />

<strong>and</strong> pharmacotoxicity of cGMP-produced Plasmodium falciparum AMA1 FVO<br />

strain ectodomain expressed in Pichia pastoris. Vaccine 2008; 26:6143-50.<br />

9. Roestenberg M, Remarque E, de JE, et al. Safety <strong>and</strong> immunogenicity of a<br />

recombinant Plasmodium falciparum AMA1 malaria vaccine adjuvanted with<br />

Alhydrogel, Montanide ISA 720 or AS02. PLoS One 2008; 3:e3960.<br />

10. Polley SD, Mwangi T, Kocken CH, et al. Human antibodies to recombinant<br />

protein constructs of Plasmodium falciparum Apical Membrane Antigen 1<br />

(AMA1) <strong>and</strong> their associations with <strong>protection</strong> from malaria. Vaccine 2004;<br />

23:718-28.<br />

11. Kocken CH, Withers-Martinez C, Dubbeld MA, et al. High-level expression of the<br />

malaria blood-stage vaccine c<strong>and</strong>idate Plasmodium falciparum apical<br />

membrane antigen 1 <strong>and</strong> induction of antibodies that inhibit erythrocyte<br />

invasion. Infect Immun 2002; 70:4471-6.<br />

12. Kocken CH, van der Wel AM, Dubbeld MA, et al. Precise timing of expression of<br />

a Plasmodium falciparum-derived transgene in Plasmodium berghei is a critical<br />

93


94 Chapter 4<br />

determinant of subsequent subcellular localization. J Biol Chem 1998;<br />

273:15119-24.<br />

13. Amador R, Moreno A, Valero V, et al. The first field trials of the chemically<br />

synthesized malaria vaccine SPf66: safety, immunogenicity <strong>and</strong> protectivity.<br />

Vaccine 1992; 10:179-84.<br />

14. Remarque EJ, Faber BW, Kocken CH, Thomas AW. A diversity-covering approach<br />

to immunization with Plasmodium falciparum apical membrane antigen 1<br />

induces broader allelic recognition <strong>and</strong> growth inhibition responses in rabbits.<br />

Infect Immun 2008; 76:2660-70.<br />

15. de Voer RM, van der Klis FR, Schepp RM, Rijkers GT, S<strong>and</strong>ers EA, Berbers GA.<br />

Age-Related Immunity to Meningococcal Serogroup C Vaccination: An Increase<br />

in the Persistence of IgG2 Correlates with a Decrease in the Avidity of IgG. PLoS<br />

One 2011; 6:e23497.<br />

16. Pierson-Mullany LK, Jackola DR, Blumenthal MN, Rosenberg A. Evidence of an<br />

affinity threshold for IgE-allergen binding in the percutaneous skin test reaction.<br />

Clin Exp Allergy 2002; 32:107-16.<br />

17. McLean AR, Rosado MM, Agenes F, Vasconcellos R, Freitas AA. Resource<br />

competition as a mechanism for B cell homeostasis. Proc Natl Acad Sci U S A<br />

1997; 94:5792-7.<br />

18. Freitas AA, Rosado MM, Viale AC, Gr<strong>and</strong>ien A. The role of cellular competition<br />

in B cell survival <strong>and</strong> selection of B cell repertoires. Eur J Immunol 1995;<br />

25:1729-38.<br />

19. Jackola DR, Liebeler CL, Lin CY, Chiu YK, Blumenthal MN, Rosenberg A. Evidence<br />

that negative feedback between antibody concentration <strong>and</strong> affinity regulates<br />

humoral response consolidation to a non-infectious antigen in infants. Mol<br />

Immunol 2005; 42:19-30.<br />

20. Freitas GR, Silva DA, Yokosawa J, et al. Antibody response <strong>and</strong> avidity of<br />

respiratory syncytial virus-specific total IgG, IgG1, <strong>and</strong> IgG3 in young children. J<br />

Med Virol 2011; 83:1826-33.<br />

21. Granoff DM, Maslanka SE, Carlone GM, et al. A modified enzyme-linked<br />

immunosorbent assay for measurement of antibody responses to<br />

meningococcal C polysaccharide that correlate with bactericidal responses. Clin<br />

Diagn Lab Immunol 1998; 5:479-85.<br />

22. Delgado MF, Coviello S, Monsalvo AC, et al. Lack of antibody affinity maturation<br />

due to poor Toll-like receptor stimulation leads to enhanced respiratory<br />

syncytial virus disease. Nat Med 2009; 15:34-41.<br />

23. Spring MD, Cummings JF, Ockenhouse CF, et al. Phase 1/2a study of the malaria<br />

vaccine c<strong>and</strong>idate apical membrane antigen-1 (AMA-1) administered in<br />

adjuvant system AS01B or AS02A. PLoS One 2009; 4:e5254.


Section 2<br />

Controlled human malaria <strong>infection</strong> model


Chapter 5<br />

Comparison of clinical <strong>and</strong> parasitological<br />

data from experimental human malaria<br />

challenge trials<br />

Meta Roestenberg 1 , Geraldine A. O’Hara 2 , Christopher J.A. Duncan 2 , Judith E.<br />

Epstein 4 , Nick J. Edwards 2 , Anja Scholzen 1 , André J.A.M. van der Ven 3 , Cornelus<br />

C. Hermsen 1 , Adrian V.S. Hill 2 , Robert W. Sauerwein 1<br />

1 Radboud University Nijmegen Medical Centre, Department of Medical<br />

Microbiology, Nijmegen, The Netherl<strong>and</strong>s<br />

2 Centre for Clinical Vaccinology <strong>and</strong> Tropical Medicine <strong>and</strong> Jenner Institute,<br />

Churchill Hospital, University of Oxford.<br />

3Radboud University Nijmegen Medical Centre, Department of General<br />

Internal Medicine, Nijmegen, The Netherl<strong>and</strong>s<br />

4Naval Medical Research Centre, US Military Malaria Vaccine Program,<br />

Maryl<strong>and</strong>, United States<br />

PLoS ONE, 2012;7(6):e38434


Chapter 5 98<br />

Abstract<br />

Exposing healthy human volunteers to Plasmodium falciparum-infected<br />

mosquitoes is an accepted tool to evaluate preliminary efficacy of malaria<br />

vaccines. To accommodate the dem<strong>and</strong> of the malaria vaccine pipeline,<br />

controlled <strong>infection</strong>s are carried out in an increasing number of centres<br />

worldwide. We assessed their safety <strong>and</strong> reproducibility.<br />

We reviewed safety <strong>and</strong> parasitological data from 128 malaria-naïve subjects<br />

participating in controlled malaria <strong>infection</strong> trials conducted at the University of<br />

Oxford, UK, <strong>and</strong> the Radboud University Nijmegen Medical Centre, The<br />

Netherl<strong>and</strong>s. Results were compared to a report from the US Military Malaria<br />

Vaccine Program.<br />

We show that controlled human malaria <strong>infection</strong> trials are safe <strong>and</strong><br />

demonstrate a consistent safety profile with minor differences in the<br />

frequencies of arthralgia, fatigue, chills <strong>and</strong> fever between institutions. But<br />

prepatent periods show significant variation. Detailed analysis of Q-PCR data<br />

reveals highly synchronous blood stage parasite growth <strong>and</strong> multiplication rates.<br />

Procedural differences can lead to some variation in safety profile <strong>and</strong> parasite<br />

kinetics between institutions. Further harmonization <strong>and</strong> st<strong>and</strong>ardization of<br />

protocols will be useful for wider adoption of these cost-effective small-scale<br />

efficacy trials. Nevertheless, parasite growth rates are highly reproducible,<br />

illustrating the robustness of controlled <strong>infection</strong>s as a valid tool for malaria<br />

vaccine development.


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

Introduction<br />

Deliberate exposure of healthy human volunteers to the bites of laboratoryreared<br />

Plasmodium falciparum (Pf)-infected mosquitoes in a controlled<br />

experimental setting is an accepted tool in malaria vaccine development. Such<br />

controlled human malaria <strong>infection</strong> (CHMI) trials can be used to investigate Pf<br />

immunology [1] or to provide data on the efficacy of malaria vaccine c<strong>and</strong>idates<br />

[2] as a precursor to more costly <strong>and</strong> logistically challenging Phase IIb field<br />

efficacy trials. In CHMI, development of blood stage parasites in test subjects is<br />

assessed by blood smears at regular time points <strong>and</strong> anti-malarial treatment is<br />

given as soon as blood stage parasites are detected microscopically, keeping<br />

blood stage parasitemia low (treatment threshold: four parasites/µl) <strong>and</strong><br />

confined to a short (two to eight day) period [3]. A comparison of the interval<br />

between exposure <strong>and</strong> parasite detection (prepatent period) among vaccinated<br />

<strong>and</strong> control subjects, together with sterile efficacy rates in vaccinees, provides<br />

an important efficacy estimate for the c<strong>and</strong>idate vaccine. Because prepatent<br />

periods without information on parasite growth rates provide only an estimate<br />

of vaccine efficacy, molecular techniques have been developed to more<br />

accurately quantify blood parasites <strong>and</strong> provide parasite kinetic data [3-5].<br />

Decades of extensive efforts to find an efficacious malaria vaccine have lead to<br />

the development of about 38 Pf c<strong>and</strong>idate (sub-unit) malaria vaccines or vaccine<br />

components (www.who.int/vaccine_research/links/Rainbow/en/index.html). To<br />

meet the dem<strong>and</strong>s of the growing malaria vaccine development pipeline, CHMI<br />

will likely be conducted in an increasing number of sites worldwide. We have<br />

performed a comparative analysis of safety <strong>and</strong> parasitological data from trials<br />

performed at the Radboud University Nijmegen Medical Centre (RUNMC), The<br />

Netherl<strong>and</strong>s, <strong>and</strong> the University of Oxford in the United Kingdom, two of a total<br />

of five different institutions <strong>and</strong> the only non-US institutions currently routinely<br />

performing CHMI. Where possible, data were compared with a previously<br />

published report from the US Military Malaria Vaccine Program (USMMVP),<br />

Naval Medical Research Centre Component, Silver Spring, Maryl<strong>and</strong> [6]. Based<br />

on this data, we provide a perspective on future strengthening of <strong>and</strong><br />

improvements to the CHMI model.<br />

99


100 Chapter 5<br />

Cohort RUNMC I RUNMC II Oxford USMMVP<br />

Number of volunteers 20 43 65 47<br />

Demographics<br />

Mean age (stdev) 29 (8.3) 22 (2.5) 27 (6.2) 27 (UNK)<br />

Sex (males) 10 14 32 27<br />

Immunized, non-protected volunteers 0 12 0 31<br />

Methodology<br />

Mosquito strain NF54 NF54 3D7 NF54<br />

Number of infected mosquitoes 4-7 5 5 5<br />

Exposure time to mosq. (min) 10 10 5 5<br />

Threshold microscopy (parasites/ul) 2 4 2 3<br />

Clinical follow-up frequency (times daily) 3 3 2 1<br />

Anti-malarial treatment<br />

Chloroquine 20 30 47<br />

Artemether/lumefantrine 33 35<br />

Atovaquone/proguanil 10<br />

Parasitological data<br />

Median prepatent period (days) 9,0 10,0 11,2 11,0<br />

Range prepatent period (days) 7.3-10.3 7.0-12.3 8.0-14.5 9.0-14.0<br />

Geometric mean peak parasitemia (Pf/ml) 7076 15901 9055<br />

Geometric mean parasitemia first cycle (Pf/ml) 567 456 48<br />

Geometric multiplication factor 11,8 11,1 11,6<br />

Laboratory safety parameters<br />

Mean platelet count day 7-10 (x10e9/l) 242 261 239<br />

References [7] [8-10] [11-17] [6]<br />

Table 1: Cohort characteristics.<br />

Methods<br />

Volunteers participating in CHMI studies performed at the RUNMC in Nijmegen,<br />

The Netherl<strong>and</strong>s <strong>and</strong> the Centre for Clinical Vaccinology <strong>and</strong> Tropical Medicine<br />

at the University of Oxford, United Kingdom were included from 1999 until 2010<br />

<strong>and</strong> from 2000 to 2010 respectively. Data were compared with a previously<br />

published report of trials performed between 1998 <strong>and</strong> 2002 at the USMMVP,<br />

Silver Spring, Maryl<strong>and</strong> [6].<br />

Patient population<br />

Data from three different cohorts were assessed. A summary of the cohort<br />

characteristics is provided in Table 1. Data from eight studies at RUNMC were<br />

analyzed in two cohorts [7-10]. The first cohort (RUNMC I) has been previously<br />

described by Verhage et al [7]. Five volunteers from this cohort received anti-


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

malarial treatment with 48 hours delay after parasites were detected by<br />

microscopy. The RUNMC II cohort includes volunteers from 2004 onwards, when<br />

more stringent cardiovascular inclusion criteria (based on SCORE cardiovascular<br />

risk [11]) were adapted following a case of myocardial infarction in a malarianegative<br />

volunteer [7] <strong>and</strong> an increased threshold for microscopic parasite<br />

detection was implemented. RUNMC II includes 12 volunteers who participated<br />

in a c<strong>and</strong>idate malaria vaccine trial, but were not protected (Nieman et al.<br />

manuscript in preparation). All other volunteers were unimmunized In Oxford,<br />

65 infectivity control volunteers participated in 14 studies [12-18].<br />

All included subjects were healthy, male <strong>and</strong> female, malaria-naïve volunteers<br />

between the ages of 18 <strong>and</strong> 50 years (Table 1). Malaria naiveté was confirmed<br />

by medical <strong>and</strong> travel history. Volunteers from the RUNMC cohorts were also<br />

confirmed negative for antibodies <strong>against</strong> blood-stage Pf by ELISA [19].<br />

Volunteers were excluded in case of known allergies to anti-malarials,<br />

pregnancy, systemic disease or chronic use of medication. Volunteers were<br />

screened by a physician based on medical history, physical examination,<br />

complete blood count, liver <strong>and</strong> renal function tests, pregnancy test <strong>and</strong><br />

serological testing for HIV, hepatitis B <strong>and</strong> C. Volunteers provided written<br />

informed consent <strong>and</strong> all studies were approved by either the RUNMC<br />

Committee on Research involving Human Subjects or Central Committee on<br />

Research involving Human Subjects (CMO 0004-0090, 0011-0262, 2001/203,<br />

2002/170, 2004/129, 2006/207, NL14715.000.06, NL24193.091.09) or the<br />

Oxfordshire Research Ethics Committee or the UK Gene Therapy Advisory<br />

Committee (C01.111, C02.069, C02.152, C02.153, C02.266, C02.268, C02.293,<br />

C02.305, CL03.100, C03.088, 04/Q1604/93, 06/Q1604/55, 05/Q1604/69, GTAC<br />

160-02).<br />

Infection procedures.<br />

Anopheles stephensi mosquitoes were infected with the NF54 strain of Pf<br />

(RUNMC) or 3D7, a clone originally derived from NF54, (Oxford) following<br />

previously described procedures [20]. Both strains are chloroquine sensitive<br />

(data not shown).<br />

Fixed numbers of mosquitoes were allowed to bite volunteers during five<br />

(Oxford) or ten (RUNMC) minutes. Fully blood-engorged mosquitoes were<br />

confirmed positive for salivary gl<strong>and</strong> sporozoites by dissection (a threshold of<br />

>10 sporozoites/gl<strong>and</strong> was used in all centres). If necessary, feeding sessions<br />

were repeated until exactly the predefined number of infected mosquitoes were<br />

101


102 Chapter 5<br />

fully engorged, i.e. five mosquitoes, except for the RUNMC I cohort, where<br />

volunteers were exposed to the bites of four to seven mosquitoes (Table 1).<br />

Monitoring took place twice (Oxford) or thrice daily (RUNMC) using microscopy<br />

of Giemsa-stained blood smears starting on day five at RUNMC or on the<br />

afternoon of day six at Oxford. Volunteers were treated with a st<strong>and</strong>ard<br />

therapeutic regimen of chloroquine, arthemether/lumefantrine or<br />

atovaquone/proguanil as soon as microscopy confirmed the presence of<br />

parasites or by the discretion of the physician.<br />

Trial volunteers were followed on an outpatient basis <strong>and</strong> lived in the vicinity of<br />

the hospital. An active tracking policy using mobile phones <strong>and</strong>/or home visits<br />

was operational at both institutions during the monitoring period. Adverse<br />

events were recorded at every visit. Investigators evaluated the potential<br />

relation of adverse events with trial procedures. All probable or possible related<br />

events were included in the analysis, with exception of the five volunteers for<br />

whom anti-malarial treatment was delayed by 48 hours. The USMMVP report<br />

included adverse events from day seven after challenge [6]. Severity of<br />

symptoms in RUNMC II <strong>and</strong> the USMMVP report were assessed according to<br />

st<strong>and</strong>ard guidelines (http://www.fda.gov/BiologicsBloodVaccines/GuidanceCom<br />

plianceRegulatoryInformation/Guidances/Vaccines/ucm074775.htm).<br />

Mild symptoms (grade 1) did not interfere with daily activities, moderate<br />

symptoms (grade 2) interfered with daily activities, severe symptoms (grade 3)<br />

prevented daily activity. Symptom severity was not consistently assessed in the<br />

other cohorts, with exception of fever, which was graded mild when 37.5 to<br />

37.9°C, moderate when 38 to 38.9°C <strong>and</strong> severe when ≥39°C in all cohorts. All<br />

centres recorded oral temperatures, which were measured at least once daily,<br />

either by volunteers themselves or by the attending physician at the clinical site.<br />

RUNMC also recorded auricular temperatures at the clinical site up to three<br />

times daily. Serious adverse events (grade 4) were defined according to<br />

International Conference of Harmonization Good Clinical Practice Guidelines.<br />

Clinical haematological laboratory data were available on a daily basis from day<br />

five post-challenge until three days after anti-malaria treatment for RUNMC<br />

cohorts. Biochemical parameters in RUNMC cohorts were assessed once, at<br />

three days after anti-malarial treatment. Clinical laboratory parameters in<br />

Oxford were not routinely recorded during challenge in any trials. For the<br />

USMMVP cohort, clinical laboratory parameters were reported at days 10-12<br />

after challenge.


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

Parasitological data<br />

Prepatent period was defined as time from exposure to positive thick smear. The<br />

threshold for microscopic detection of parasites varied between cohorts<br />

depending on the local st<strong>and</strong>ard operating procedure (Table 1). Centres use<br />

different microscopes, blood volume <strong>and</strong> slide surface area. RUNMC <strong>and</strong> Oxford<br />

readers complete 200 fields, yielding a threshold of approximately two parasites<br />

per μl blood in RUNMC I <strong>and</strong> Oxford (slide was deemed positive if one parasite<br />

was found), which was confirmed by Q-PCR in Oxford. A threshold of four<br />

parasites per μl blood was achieved in RUNMC II (slide was deemed positive if<br />

two parasites were found). USMMVP readers completed 5 passes (72<br />

fields/pass), yielding a threshold of approximately 3 parasites per μl blood (slide<br />

positive if two parasites were found). In all centres, slides were read by two<br />

independent readers at 1000x magnification.<br />

Simultaneously, a quantitative PCR (Q-PCR) for Pf was used in Oxford to support<br />

microscopy. Parasite densities were measured by Q-PCR for RUNMC <strong>and</strong> Oxford<br />

cohorts as previously described [3, 5]. Although methodology of the Q-PCR<br />

differed, there was no inter-institutional difference in measured densities,<br />

confirmed by an exchange of samples between both institutions (data not<br />

shown). Peak parasitemia was defined as the highest parasite density during<br />

<strong>infection</strong> measured by Q-PCR. Any cycle threshold above 45 was plotted as zero<br />

parasitemia. For calculations, these samples were given a value of half the<br />

detection threshold (ten parasites/ml).<br />

Data analysis<br />

Data were assessed in SPSS 16.0 with correction for multiple analyses.<br />

Differences between frequencies <strong>and</strong> prepatent period were compared by<br />

Kruskal-Wallis tests when comparing multiple groups or Mann-Whitney U tests<br />

when comparing two groups. Dunn’s multiple comparisons test was performed<br />

as post-hoc analysis when appropriate. Analysis of parasitological PCR data was<br />

performed on log-transformed data using independent-samples t-test when<br />

comparing two groups <strong>and</strong> one-way ANOVA when comparing three groups. The<br />

multiplication rate of blood stage parasites was calculated by the ratio of the<br />

geometric mean parasitemia in the second cycle with the first cycle (day 8.6-10.5<br />

vs 6.6-8.5) or the third cycle with the second cycle (day 10.6-12.5 vs 8.6-10.5), or<br />

if possible, the mean of both ratios.<br />

103


104 Chapter 5<br />

p-value*<br />

0.44<br />


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

Correlations were assessed by Pearson’s correlation when parametric <strong>and</strong><br />

Spearman’s when non-parametric. Two-sided p-values below 0.05 were<br />

considered significant unless stated otherwise.<br />

Results<br />

Demographics<br />

A total of 128 volunteers were grouped into three different cohorts: RUNMC I<br />

<strong>and</strong> II <strong>and</strong> Oxford <strong>and</strong> compared to published data from 47 infected individuals<br />

at USMMVP [6], for a total of 175 volunteers. Individuals were generally 20-40<br />

years old, with equal distribution between male <strong>and</strong> female participants (Table<br />

1). There were no significant differences in sex distribution between the<br />

different cohorts (Kruskal-Wallis, p=0.22), but RUNMC II volunteers were<br />

significantly younger (Kruskal-Wallis, p37.5°C) occurred with significant higher frequency in<br />

RUNMC <strong>and</strong> USMMVP.<br />

105


106 Chapter 5<br />

Figure 1. Time to microscopically detected parasitemia by cohort<br />

Survival curve for four cohorts: RUNMC I (orange), RUNMC II (blue), USMMVP<br />

(grey) <strong>and</strong> Oxford (green).<br />

Unsolicited adverse events were infrequent <strong>and</strong> never severe, <strong>and</strong> all resolved<br />

spontaneously. Events included vasovagal collapse, epistaxis, flatulence,<br />

insomnia, tinnitus, hyperesthesia, psychiatric complaints associated with<br />

chloroquine [7], pleuritic chest pain, sore throat, migraine, gingivitis,<br />

palpitations, numbness in fingers, dizziness, drowsiness, photosensitivity <strong>and</strong><br />

stiff neck.<br />

Laboratory safety parameters<br />

Clinical laboratory parameters in RUNMC <strong>and</strong> USMMVP did not show significant<br />

abnormalities in haemoglobin content; parameters were not routinely available<br />

in Oxford. The majority of volunteers experienced a mild to moderate decrease<br />

in leucocyte count, none of which was severe (


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

Figure 2: Geometric mean parasite density by Q-PCR per cohort<br />

Three cohorts are depicted RUNMC I (orange), RUNMC II (blue) <strong>and</strong> Oxford<br />

(green).<br />

No abnormalities of urea <strong>and</strong> creatinine were found, but USMMVP reported six<br />

cases of haemoglobinuria <strong>and</strong> one case of proteinuria [6]. Urinary parameters<br />

were not checked in Oxford or RUNMC. Incidental increases particularly in<br />

alanine aminotransferase <strong>and</strong> aspartate aminotransferase were reported in both<br />

RUNMC <strong>and</strong> USMMVP. Severe increases (>2.5x ULN) were found in five cases,<br />

three from USMMVP (twice at day 10, once at day 14-16) <strong>and</strong> two from RUNMC<br />

II (day 16 <strong>and</strong> 17). All tests normalized at the end of the trial.<br />

Parasitemia by microscopy<br />

Prepatent period between the four cohorts was significantly different (Kruskall-<br />

Wallis p


108 Chapter 5<br />

Figure 3. Statistics of Q-PCR parasitemia per cohort.<br />

Peak parasitemia (A, one-way ANOVA p=0.13), geometric mean parasitemia during<br />

the first blood stage parasite multiplication cycle, day 6.5 to 8.5 (B, one-way<br />

ANOVA p45 was assigned a parasitemia of 10 parasites/ml.<br />

3A), although there seemed to be a trend towards higher parasitemia at RUNMC<br />

after the threshold for microscopic parasite detection changed. The first blood<br />

stage parasite growth cycle was significantly lower in Oxford as compared to<br />

RUNMC (p


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

Figure 4. Correlation of Q-PCR parasitemia with prepatent period<br />

Correlation between peak parasitemia (A) or geometric mean<br />

parasitemia during the first multiplication cycle from day 6.5 to day 8.5<br />

(B) with prepatent period (R 2 = 0.27 <strong>and</strong> -0.73, p=0.006 <strong>and</strong> p=


110 Chapter 5<br />

in time to microscopically detected parasitemia whereas clinical symptoms are<br />

broadly similar. Although there are several methodological differences between<br />

the CHMI protocols that limit direct comparability, this difference in prepatency<br />

likely results from variation in the mean parasite burden in the first blood stage,<br />

possibly as a result of different inoculum size, whilst blood stage multiplication<br />

factors are equal.<br />

The analysis of adverse events from 175 non-immune participants of sporozoite<br />

challenge trials shows that serious adverse events (grade 4) are rare. One<br />

serious cardiac adverse event was reported; the true nature <strong>and</strong> pathophysiological<br />

explanation of that event remains unclear [21]. Severe adverse<br />

events (grade 3) related to clinical malaria occur in up to half of the volunteers<br />

<strong>and</strong> persist for several days. We conclude that CHMI are generally safe, but may<br />

lead to severe (grade 3) symptoms, though not serious adverse events, in a<br />

significant proportion of subjects. Several precautions are taken in both<br />

institutions to ensure safety of volunteers, such as 24-hour phone access, medicalert<br />

cards <strong>and</strong> emergency contact procedures. Nevertheless, the exposure of<br />

volunteers to the likelihood of some severe adverse events should be carefully<br />

weighed <strong>against</strong> the benefits of the information to be gained [22].<br />

We find significant differences in frequencies of fever, fatigue, arthralgia <strong>and</strong><br />

chills between institutions. Cohorts with a longer prepatent period or a higher<br />

peak parasitemia do not consistently show a higher frequency of adverse events.<br />

These data, combined with methodological differences in the recording of<br />

adverse events (e.g. home-monitoring of oral temperature in RUNMC), lead us<br />

to conclude that biologically relevant variation in patho-physiology between<br />

institutions seems unlikely. Nevertheless, st<strong>and</strong>ardized assessment of adverse<br />

events <strong>and</strong> harmonization of solicited events would advance the interpretability<br />

<strong>and</strong> comparability of CHMI in different settings worldwide.<br />

Laboratory safety parameters show a severe decrease in platelet count<br />

(


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

An increase in threshold for microscopic detection of parasites leads to a<br />

prolonged prepatent period but not an increase in the number of adverse<br />

events, as illustrated by comparing the RUNMC I <strong>and</strong> II cohorts. St<strong>and</strong>ardized<br />

reading of blood smears is thus essential to harmonize trial endpoints<br />

worldwide; recent efforts by the WHO have resulted in a proposed<br />

harmonization document (Laurens MB, Roestenberg M, <strong>and</strong> Moorthy VS<br />

manuscript in preparation). Variability in prepatent period among institutions,<br />

however, cannot be explained solely by microscopy methodology. A detailed<br />

analysis of parasitemia by Q-PCR revealed a ~10 fold difference in the parasite<br />

burden during the first blood stage growth cycle. Taking into account a<br />

replication factor of approximately 10 every 48 hours, this difference accounts<br />

for a 48 hour shorter prepatent period at RUNMC. The variation in parasite load<br />

may reflect variation in liver stage development of the parasites or,<br />

alternatively, the number of inoculated parasites. The number of inoculated<br />

parasites is estimated to vary widely (ranging from 5-10 [26] to 100-300<br />

sporozoites per bite [27, 28]). Whether the intensity of mosquito <strong>infection</strong> (e.g.<br />

sporozoite salivary gl<strong>and</strong> load) or exposure time influences the number of<br />

sporozoites inoculated is controversial [29, 30]. However, also a formal relation<br />

between the number of parasites inoculated <strong>and</strong> prepatent period has never<br />

been established [7, 28, 30-33]. Similarly, the role of mosquito infectivity (i.e.<br />

number of sporozoites per mosquito), parasite strain (3D7 vs NF54), exposure<br />

time (5 vs 10 minutes) or viability of inoculated sporozoites in determining the<br />

inoculated dose is unclear [30, 31]. Efforts to st<strong>and</strong>ardize the sporozoite dose<br />

should ideally be tested for their impact on comparability <strong>and</strong> reproducibility of<br />

CHMI. St<strong>and</strong>ardization may be achieved by harmonization of mosquito breeding<br />

<strong>and</strong> feeding protocols or by needle injection of cryopreserved sporozoites. CHMI<br />

trials are underway to test the infectiousness of cryopreserved sporozoites by<br />

needle injection (NCT01086917) [34].<br />

We show that blood stage parasite growth is cyclical <strong>and</strong> highly synchronous<br />

within <strong>and</strong> between institutions. Importantly, the duration of parasite liver stage<br />

development as well as the blood stage multiplication rate are highly<br />

reproducible. Thus vaccine efficacy can be robustly evaluated in any of the CHMI<br />

centres if a non-protected, malaria-naïve control group is included.<br />

CHMI trials do not fully mimic conditions in endemic regions where pre-existing<br />

immunity may augment or impair vaccine efficacy. A limited number of<br />

comparisons between Phase IIa preliminary efficacy trials <strong>and</strong> Phase IIb field<br />

111


112 Chapter 5<br />

efficacy trials shows that results are generally in line, but more comparisons are<br />

required before definite conclusions can be drawn [2]. Another potential<br />

difference is reflected by the almost instant delivery of parasites by five infected<br />

mosquitoes, which has been considered unnatural <strong>and</strong> a stringent test for<br />

vaccine-induced immune responses [35]. However, although the frequency of<br />

infectious mosquito bites is generally lower in malaria-endemic areas, intense<br />

transmission can occur. A person may be subjected to 35–96 mosquito bites per<br />

night, <strong>and</strong> in certain areas approximately 10% of mosquitoes are infected with Pf<br />

[36].<br />

The present data show that CHMI can be safely conducted, but will lead to grade<br />

3 adverse events in a proportion of volunteers. The primary parasitological<br />

outcome of such experiments is highly reproducible within institutions but may<br />

vary between trial centres. With an increasing number of CHMI centres being<br />

installed, priority should be given to initiatives to st<strong>and</strong>ardize challenge<br />

procedures [37]. The implementation of guidelines will enhance the<br />

comparability of CHMI; a critical <strong>and</strong> indispensable component of malaria<br />

vaccine development worldwide.<br />

Acknowledgements<br />

We would like to acknowledge Geert-Jan van Gemert, Marga van de Vegte-<br />

Bolmer, Matthew McCall, An-Emmie Nieman, Theo Arens, Pieter Beckers, Karina<br />

Teelen, Jorien Wiersma <strong>and</strong> the staff of the Clinical Research Center Nijmegen<br />

for their continuing support of controlled malaria <strong>infection</strong> trials at the RUNMC.<br />

We thank the clinical staff of the CCVTM <strong>and</strong> scientists of Jenner Institute Labs<br />

who contributed to these trials in Oxford. We thank all participating volunteers.<br />

Judith Epstein is a military service member. This work was prepared as part of<br />

her official duties. The views expressed in this article are those of the authors<br />

<strong>and</strong> do not necessarily reflect the official policy or position of the Department of<br />

the Navy, Department of Defense, nor the U.S. Government. Title 17 U.S.C. §105<br />

provides that ‘Copyright <strong>protection</strong> under this title is not available for any work<br />

of the United States Government.’ Title 17 U.S.C. §101 defines a U.S.<br />

Government work as a work prepared by a military service member or employee<br />

of the U.S. Government as part of that person’s official duties.


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

Funding<br />

This work was supported by Top Institute <strong>Pharma</strong> [grant number T4-102] <strong>and</strong><br />

Dioraphte Foundation. The UK trials were supported by the UK Medical Research<br />

Council, the Wellcome Trust, the European Commission framework<br />

programmes, the European Vaccine Initiative, the PATH Malaria Vaccine<br />

Initiative <strong>and</strong> the UK National Institute for Health Research.<br />

113


114 Chapter 5<br />

References<br />

1. McCall MB, Netea MG, Hermsen CC et al. Plasmodium falciparum <strong>infection</strong><br />

causes proinflammatory priming of human TLR responses. J Immunol 2007;<br />

179:162-171.<br />

2. Sauerwein RW, Roestenberg M, Moorthy VS. <strong>Experimental</strong> human challenge<br />

<strong>infection</strong>s can accelerate clinical malaria vaccine development. Nat Rev<br />

Immunol 2011; 11:57-64.<br />

3. Andrews L, Andersen RF, Webster D et al. Quantitative real-time polymerase<br />

chain reaction for malaria diagnosis <strong>and</strong> its use in malaria vaccine clinical trials.<br />

Am J Trop Med Hyg 2005; 73:191-198.<br />

4. Felger I, Genton B, Smith T, Tanner M, Beck HP. Molecular monitoring in<br />

malaria vaccine trials. Trends Parasitol 2003; 19:60-63.<br />

5. Hermsen CC, Telgt DS, Linders EH et al. Detection of Plasmodium falciparum<br />

malaria parasites in vivo by real-time quantitative PCR. Mol Biochem Parasitol<br />

2001; 118:247-251.<br />

6. Epstein JE, Rao S, Williams F et al. Safety <strong>and</strong> clinical outcome of experimental<br />

challenge of human volunteers with Plasmodium falciparum-infected<br />

mosquitoes: an update. J Infect Dis 2007; 196:145-154.<br />

7. Verhage DF, Telgt DS, Bousema JT et al. Clinical outcome of experimental<br />

human malaria induced by Plasmodium falciparum-infected mosquitoes. Neth J<br />

Med 2005; 63:52-58.<br />

8. McCall MB, Beynon AJ, Mylanus EA, van d, V, Sauerwein RW. No hearing loss<br />

associated with the use of artemether-lumefantrine to treat experimental<br />

human malaria. Trans R Soc Trop Med Hyg 2006; 100:1098-1104.<br />

9. Roestenberg M, McCall M, Hopman J et al. Protection <strong>against</strong> a malaria<br />

challenge by sporozoite inoculation. N Engl J Med 2009; 361:468-477.<br />

10. Roestenberg M, Teirlinck AC, McCall MB et al. Long-term <strong>protection</strong> <strong>against</strong><br />

malaria after experimental sporozoite inoculation: an open-label follow-up<br />

study. Lancet 2011.<br />

11. Conroy RM, Pyorala K, Fitzgerald AP et al. Estimation of ten-year risk of fatal<br />

cardiovascular disease in Europe: the SCORE project. Eur Heart J 2003; 24:987-<br />

1003.<br />

12. Thompson FM, Porter DW, Okitsu SL et al. Evidence of blood stage efficacy with<br />

a virosomal malaria vaccine in a phase IIa clinical trial. PLoS One 2008; 3:e1493.<br />

13. Dunachie SJ, Walther M, Epstein JE et al. A DNA prime-modified vaccinia virus<br />

ankara boost vaccine encoding thrombospondin-related adhesion protein but<br />

not circumsporozoite protein partially protects healthy malaria-naive adults<br />

<strong>against</strong> Plasmodium falciparum sporozoite challenge. Infect Immun 2006;<br />

74:5933-5942.<br />

14. Walther M, Thompson FM, Dunachie S et al. Safety, immunogenicity, <strong>and</strong><br />

efficacy of prime-boost immunization with recombinant poxvirus FP9 <strong>and</strong><br />

modified vaccinia virus Ankara encoding the full-length Plasmodium falciparum<br />

circumsporozoite protein. Infect Immun 2006; 74:2706-2716.


Comparison of clinical <strong>and</strong> parasitological data from experimental human malaria<br />

challenge trials<br />

15. Webster DP, Dunachie S, Vuola JM et al. Enhanced T cell-mediated <strong>protection</strong><br />

<strong>against</strong> malaria in human challenges by using the recombinant poxviruses FP9<br />

<strong>and</strong> modified vaccinia virus Ankara. Proc Natl Acad Sci U S A 2005; 102:4836-<br />

4841.<br />

16. Walther M, Dunachie S, Keating S et al. Safety, immunogenicity <strong>and</strong> efficacy of a<br />

pre-erythrocytic malaria c<strong>and</strong>idate vaccine, ICC-1132 formulated in Seppic ISA<br />

720. Vaccine 2005; 23:857-864.<br />

17. McConkey SJ, Reece WH, Moorthy VS et al. Enhanced T-cell immunogenicity of<br />

plasmid DNA vaccines boosted by recombinant modified vaccinia virus Ankara<br />

in humans. Nat Med 2003; 9:729-735.<br />

18. Porter DW, Thompson FM, Berthoud TK et al. A human Phase I/IIa malaria<br />

challenge trial of a polyprotein malaria vaccine. Vaccine 2011; 29:7514-7522.<br />

19. Bousema JT, Roeffen W, van der KM et al. Rapid onset of transmission-reducing<br />

antibodies in javanese migrants exposed to malaria in Papua,Iindonesia. Am J<br />

Trop Med Hyg 2006; 74:425-431.<br />

20. Ponnudurai T, Lensen AH, van Gemert GJ, Bensink MP, Bolmer M, Meuwissen<br />

JH. Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes.<br />

Parasitology 1989; 98 Pt 2:165-173.<br />

21. Nieman AE, de MQ, Roestenberg M et al. Cardiac complication after<br />

experimental human malaria <strong>infection</strong>: a case report. Malar J 2009; 8:277.<br />

22. Visser HK. <strong>Experimental</strong> malaria in human volunteers: ethical aspects. Neth J<br />

Med 2005; 63:41-42.<br />

23. de Mast Q, Groot E, Lenting PJ et al. Thrombocytopenia <strong>and</strong> release of activated<br />

von Willebr<strong>and</strong> Factor during early Plasmodium falciparum malaria. J Infect Dis<br />

2007; 196:622-628.<br />

24. de Mast Q, de Groot PG, van Heerde WL et al. Thrombocytopenia in early<br />

malaria is associated with GP1b shedding in absence of systemic platelet<br />

activation <strong>and</strong> consumptive coagulopathy. Br J Haematol 2010; 151:495-503.<br />

25. Ali H, Ahsan T, Mahmood T, Bakht SF, Farooq MU, Ahmed N. Parasite density<br />

<strong>and</strong> the spectrum of clinical illness in falciparum malaria. J Coll Physicians Surg<br />

Pak 2008; 18:362-368.<br />

26. Beier JC, Davis JR, Vaughan JA, Noden BH, Beier MS. Quantitation of<br />

Plasmodium falciparum sporozoites transmitted in vitro by experimentally<br />

infected Anopheles gambiae <strong>and</strong> Anopheles stephensi. Am J Trop Med Hyg<br />

1991; 44:564-570.<br />

27. Jin Y, Kebaier C, V<strong>and</strong>erberg J. Direct microscopic quantification of dynamics of<br />

Plasmodium berghei sporozoite transmission from mosquitoes to mice. Infect<br />

Immun 2007; 75:5532-5539.<br />

28. Frischknecht F, Baldacci P, Martin B et al. Imaging movement of malaria<br />

parasites during transmission by Anopheles mosquitoes. Cell Microbiol 2004;<br />

6:687-694.<br />

29. Pumpuni CB, Mendis C, Beier JC. Plasmodium yoelii sporozoite infectivity varies<br />

as a function of sporozoite loads in Anopheles stephensi mosquitoes. J Parasitol<br />

1997; 83:652-655.<br />

115


116 Chapter 5<br />

30. Ponnudurai T, Lensen AH, van Gemert GJ, Bolmer MG, Meuwissen JH. Feeding<br />

behaviour <strong>and</strong> sporozoite ejection by infected Anopheles stephensi. Trans R Soc<br />

Trop Med Hyg 1991; 85:175-180.<br />

31. Rickman LS, Jones TR, Long GW et al. Plasmodium falciparum-infected<br />

Anopheles stephensi inconsistently transmit malaria to humans. Am J Trop Med<br />

Hyg 1990; 43:441-445.<br />

32. Jeffery GM, Young MD, Burgess RW, Eyles DE. Early activity in sporozoiteinduced<br />

Plasmodium falciparum <strong>infection</strong>s. Ann Trop Med Parasitol 1959;<br />

53:51-58.<br />

33. Rosenberg R, Wirtz RA, Schneider I, Burge R. An estimation of the number of<br />

malaria sporozoites ejected by a feeding mosquito. Trans R Soc Trop Med Hyg<br />

1990; 84:209-212.<br />

34. Hoffman SL, Billingsley PF, James E et al. Development of a metabolically active,<br />

non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria.<br />

Hum Vaccin 2010; 6:97-106.<br />

35. Genton B, D'Acremont V, Lurati-Ruiz F et al. R<strong>and</strong>omized double-blind<br />

controlled Phase I/IIa trial to assess the efficacy of malaria vaccine PfCS102 to<br />

protect <strong>against</strong> challenge with P. falciparum. Vaccine 2010; 28:6573-6580.<br />

36. Trape JF, Zoulani A. Malaria <strong>and</strong> urbanization in central Africa: the example of<br />

Brazzaville. Part II: Results of entomological surveys <strong>and</strong> epidemiological<br />

analysis. Trans R Soc Trop Med Hyg 1987; 81 Suppl 2:10-18.<br />

37. Moorthy VS, Diggs C, Ferro S et al. Report of a consultation on the optimization<br />

of clinical challenge trials for evaluation of c<strong>and</strong>idate blood stage malaria<br />

vaccines, 18-19 March 2009, Bethesda, MD, USA. Vaccine 2009; 27:5719-5725.


Chapter 6<br />

Efficacy of pre-erythrocytic <strong>and</strong> blood-stage<br />

malaria vaccines can be assessed in small<br />

sporozoite challenge trials in human<br />

volunteers<br />

Meta Roestenberg 1 , Sake J. de Vlas 3 , An-Emmie Nieman 2 , Robert W. Sauerwein 1 ,<br />

Cornelus C. Hermsen 1<br />

1<br />

Radboud University Nijmegen Medical Centre, dept. of Medical Microbiology,<br />

Nijmegen, The Netherl<strong>and</strong>s<br />

2<br />

VU Medical Centre, dept. of Medical Microbiology, Amsterdam, The<br />

Netherl<strong>and</strong>s<br />

3<br />

Erasmus Medical Centre, dept. of Public Health, Rotterdam, The Netherl<strong>and</strong>s<br />

J. Infect. Dis. 2012 Aug; 206(3):319-23


Chapter 6 118<br />

Abstract<br />

The development of a vaccine <strong>against</strong> malaria has public health priority. In a<br />

controlled setting, preliminary data on the efficacy of Plasmodium falciparum<br />

(Pf) vaccine c<strong>and</strong>idates can be obtained by exposing immunized human<br />

volunteers to the bites of laboratory-reared Pf-infected mosquitoes. Using<br />

empirical data we show that these trials, with small numbers of volunteers, are<br />

sufficiently powered to detect protective biological effects induced by preerythrocytic<br />

<strong>and</strong>/or blood-stage c<strong>and</strong>idate vaccines if parasitemia is measured<br />

daily by quantitative PCR. Sporozoite challenge trials are thus a powerful tool for<br />

early selection of c<strong>and</strong>idates that warrant efficacy of trials in the field.


Efficacy of pre-erythrocytic <strong>and</strong> blood-stage malaria vaccines can be assessed in small<br />

sporozoite challenge trials in human volunteers<br />

Introduction<br />

The development of an effective vaccine <strong>against</strong> malaria has public health<br />

priority. With an increasing number of c<strong>and</strong>idate Plasmodium falciparum (Pf)<br />

vaccines <strong>and</strong> only a limited number of field trial sites available, human<br />

sporozoite challenges are used to assess preliminary vaccine efficacy before<br />

proceeding to phase IIb trials [1]. In such phase IIa challenge trials malaria-naïve<br />

volunteers are immunized with a c<strong>and</strong>idate vaccine <strong>and</strong> subsequently exposed<br />

to the bites of laboratory-reared Pf-infected mosquitoes. Traditionally, a<br />

comparison of the prepatent period (time from challenge until positive bloodslide)<br />

between controls <strong>and</strong> vaccinees provides an efficacy estimate. The<br />

development of molecular techniques (Q-PCR) allow for a detailed analysis of<br />

blood-stage parasite growth [2].<br />

Sporozoite challenge trials are thought suitable for testing pre-erythrocytic (liver<br />

stage) vaccines, because of the natural route of exposure (mosquito bite) <strong>and</strong><br />

the full pre-erythrocytic development of Pf parasites in volunteers. To date, 30<br />

reports of combined phaseI/IIa sporozoite challenge trials are available, of which<br />

three pre-erythrocytic vaccine c<strong>and</strong>idates induced full <strong>protection</strong> [1]. One of the<br />

three c<strong>and</strong>idates is currently in phase III clinical development [3], whereas<br />

disappointing preliminary efficacy data have halted the clinical development of<br />

others [4, 5]. This illustrates the importance of challenge trials in the clinical<br />

development path of pre-erythrocytic vaccines.<br />

Preliminary efficacy testing of asexual erythrocytic (blood-stage) stage vaccines<br />

is more complex, since vaccine efficacy can only be obtained by evaluating<br />

blood-stage parasite growth over a sufficient lengthy period of time. However,<br />

blood-stage parasitemia is terminated by curative anti-malarial treatment at<br />

0.0001% infected erythrocytes, limiting parasitemia to an interval of one to six<br />

days only (mean 1.7 multiplication cycles) [6]. Immunological effects have to<br />

exert significant parasite inhibition in this short period in order to ensure<br />

detectable vaccine efficacy, making the use of challenge trials for erythrocytic<br />

vaccine c<strong>and</strong>idates controversial. To date, only two erythrocytic malaria vaccine<br />

c<strong>and</strong>idates have been subjected to sporozoite challenge (Apical Membrane<br />

Antigen 1 <strong>and</strong> Merozoite Surface Protein 1) [7, 8]. Analysis of parasitological<br />

data from two of these trials revealed an apparent pre-erythrocytic inhibiting<br />

effect, but could not show blood-stage inhibition (S.H. Sheehy pers. comm.) [7].<br />

119


120 Chapter 6<br />

It is thus unclear whether blood-stage parasite growth inhibition can be<br />

determined with sufficient precision after sporozoite challenge.<br />

The advent of molecular techniques allows for quantification of submicroscopic<br />

parasitemia [2]. In addition to the number of protected volunteers, Q-PCR allows<br />

for estimations of liver stage parasite load (for pre-erythrocytic vaccines) or<br />

blood-stage multiplication rate (for erythrocytic vaccines), which may be<br />

particularly useful in vaccines that do not induce full <strong>protection</strong>, e.g. “leaky<br />

vaccines” [9]. Here we use Q-PCR data from 11 years of experience of sporozoite<br />

challenge trials at the Radboud University Nijmegen Medical Centre (RUNMC) to<br />

determine inter-individual variation in parasite kinetics <strong>and</strong> calculate the power<br />

of sporozoite challenge trials for both pre-erythrocytic <strong>and</strong> asexual erythrocytic<br />

vaccines.<br />

Methods<br />

Data <strong>and</strong> study population<br />

Data were available from 48 volunteers participating in seven different<br />

sporozoite challenge trials at the RUNMC from 1999 to 2010. The data set<br />

included subjects from immunological studies (N=20), infectivity controls from<br />

immunisation trials (N=16) <strong>and</strong> subjects from a malaria vaccine trial not showing<br />

any <strong>protection</strong> (N= 12). All volunteers were challenged by the bites of four to<br />

seven infected mosquitoes for ten minutes. Mosquitoes were laboratory-reared<br />

<strong>and</strong> infected with the NF54 strain of Pf [10]. Presence of sporozoites in<br />

mosquitoes was confirmed by salivary gl<strong>and</strong> dissection. Trial subjects were<br />

followed two to three times daily from day five after challenge until three days<br />

after start of antimalarial treatment. At every visit, blood samples were collected<br />

<strong>and</strong> assessed for presence of parasites by microscopy (threshold 4 Pf/µl) <strong>and</strong><br />

quantified by Q-PCR (threshold 20-100 Pf/ml) [2]. Antimalarial treatment was<br />

provided as soon parasites were detected by microscopy. Ethical approval was<br />

obtained for each trial separately (Review board numbers: 0011-0262,<br />

2001/203, 2002/170, 2004/129, 2006/207, NL14715.000.06, NL24193.091.09).<br />

Statistical analysis<br />

Analyses were performed using log-transformed data. We calculated geometric<br />

mean parasitemia for every cycle in individual volunteers as well as group<br />

geometric mean <strong>and</strong> st<strong>and</strong>ard deviation (SD). We determined individual parasite<br />

multiplication rate by division of parasite densities of two subsequent cycles <strong>and</strong>


Efficacy of pre-erythrocytic <strong>and</strong> blood-stage malaria vaccines can be assessed in small<br />

sporozoite challenge trials in human volunteers<br />

calculated the geometric mean multiplication rate <strong>and</strong> SD. Samples with a Q-PCR<br />

CT >45 were estimated at half the detection limit (10 or 50 Pf/ml).<br />

Geometric mean parasitemia in the first cycle <strong>and</strong> parasite multiplication rates<br />

showed a normal distribution, so power calculations were performed using a<br />

two-sample t test power analysis with α=0.05 <strong>and</strong> β=0.80. We separately<br />

determined the power using all three daily time points if available (8:00am,<br />

4:00pm <strong>and</strong> 10:00pm, n=33), two daily time points (8:00am <strong>and</strong> 4:00pm, n=48)<br />

or one daily time point (8:00am, n=48).<br />

Power calculations estimated the group size required to detect significant<br />

differences between vaccinees <strong>and</strong> controls. Vaccine <strong>and</strong> control group were<br />

assumed of equal size. Pre-erythrocytic vaccines were assumed to reduce the<br />

geometric mean parasitemia of the first multiplication cycle, but not alter the<br />

parasite multiplication rate. Erythrocytic stage vaccines were assumed to reduce<br />

the parasite blood-stage multiplication rate, reducing parasite load from the first<br />

cycle onwards. Vaccine effects were simulated by beta distribution with an<br />

assumed mean inhibition of 70, 80, 90 <strong>and</strong> 95% <strong>and</strong> SD of 5 or 10%. Vaccine<br />

inhibition was simulated 100 times for every individual subject <strong>and</strong> repeated ten<br />

times based on r<strong>and</strong>om sampling from the distribution. We assumed a<br />

continuous infectious dose of merozoites, as opposed to the actual release of<br />

discrete batches of merozoites from infected hepatocytes. Interindividual<br />

variation masked the discrete nature of <strong>infection</strong> in empirical situations with<br />

high numbers of infected hepatocytes. In simulations with low numbers of<br />

infected hepatocytes, where a discrete variable would give a binary outcome<br />

(i.e. protected or non-protected), the resulting blood-stage parasitemia was<br />

always below the Q-PCR detection limit. In such cases, values were corrected to<br />

a st<strong>and</strong>ard value of half the Q-PCR detection limit. The number of subjects with<br />

simulated parasitemia ≤ 1Pf/ml or multiplication rate ≤1 was counted. For some<br />

subjects second cycle data were missing because they had already received<br />

antimalarial treatment for safety reasons: eight volunteers in the group with<br />

samples thrice daily (24%) <strong>and</strong> nine volunteers in the group with samples twice<br />

daily (19%). The group size for erythrocytic vaccines was corrected for these<br />

missing data by addition of the respective proportion of volunteers. Similarly,<br />

the power for pre-erythrocytic vaccines was corrected for the number of<br />

volunteers with the first cycle data points below the detection limit: two<br />

volunteers in the group with samples once daily (4%).<br />

121


122 Chapter 6<br />

Figure 1. (A) Parasite density in blood measured by quantitative PCR in 48 volunteers<br />

participating in seven sporozoite challenge trials at the Radboud University Medical<br />

Centre Nijmegen. Grey lines are measurements for individual volunteers, the black line<br />

shows geometric mean parasitemia. Q-PCR cycle threshold above 45 was plotted as zero.<br />

(B) Geometric mean parasite density per multiplication cycle for individual volunteers.<br />

(C) Multiplication rate for every individual by taking the ratio of geometric mean<br />

parasitemia from two cycles. Shown are the ratio of multiplication cycle 2 with cycle 1,<br />

cycle 3 with cycle 2 <strong>and</strong> square root of the ratio of cycle 3 with cycle 1. Lines indicate<br />

geometric mean of ratios.<br />

Results<br />

Parasite densities showed a consistent cyclical pattern of blood-stage parasite<br />

growth in all volunteers (Figure 1A). The following multiplication cycle intervals<br />

were identified: first cycle from day 6.5 until day 8.5, second cycle from day 8.5<br />

until day 10.5 <strong>and</strong> third cycle from day 10.5 until day 12.5. All individuals deve-


Efficacy of pre-erythrocytic <strong>and</strong> blood-stage malaria vaccines can be assessed in small<br />

sporozoite challenge trials in human volunteers<br />

Assumed<br />

mean<br />

Assumed<br />

inhibition<br />

Corresponding Corresponding Group size (N)<br />

subjects with<br />

1st cycle mean<br />

subjects with<br />

multiplication<br />

Pre-erythrocytic<br />

vaccine<br />

Asexual erythrocytic<br />

vaccine<br />

inhibition (%) s.d. (%) ≤ 1 Pf/ml (%) rate ≤ 1 (%) Once Twice Thrice Once Twice Thrice<br />

70 5 2 9 24 21 20 12 12 11<br />

80 5 5 20 13 12 12 7 7 6<br />

90 5 16 47 6 6 6 4 4 4<br />

95 5 40 77 4 3 3 3 3 3<br />

70 10 3 11 22 20 19 12 11 11<br />

80 10 7 24 12 11 11 7 7 7<br />

90 10 29 59 6 5 5 4 4 4<br />

95 10 65 82 3 3 3 3 3 3<br />

Table 1. Sporozoite challenge group size required for determining parasite inhibition<br />

induced by pre-erythrocytic <strong>and</strong> asexual erythrocytic malaria c<strong>and</strong>idate vaccines when<br />

measuring parasitemia by PCR once, twice or three times a day. For pre-eythrocytic<br />

vaccines a threshold of 1Pf/ml was set somewhat arbitrarily to estimate the percentage<br />

of subjects considered protected. For erythrocytic vaccines, the percentage of subjects<br />

with multiplication rate ≤ 1 is provided. Power calculations were based on data from 48<br />

volunteers participating in seven sporozoite challenge trials at the Radboud University<br />

Medical Centre Nijmegen.<br />

loped microscopically detectable parasitemia (range day 7-12.3). No individuals<br />

were lost to follow-up.<br />

Parasite densities in the first cycle varied from 23 to 5273 Pf/ml (1.37 to 3.17<br />

log), with a geometric mean of 500 Pf/ml (2.70 log, SD 0.60, n=48, Figure 1B).<br />

Group geometric mean parasite density in the first cycle was not significantly<br />

different between the seven trials (ANOVA p=0.5), so data were pooled for<br />

analysis. In the second <strong>and</strong> third cycle, geometric mean parasite density<br />

increased to 4485 Pf/ml (3.65 log, SD 0.62, n=40) <strong>and</strong> 11369 Pf/ml (4.06 log, SD<br />

0.48, n=9) respectively. Only limited data were available for the third<br />

multiplication cycle, because the majority had crossed the blood-slide threshold<br />

for antimalarial treatment. Volunteers for whom data were available in the third<br />

cycle started with a lower parasite load in the first cycle (geometric mean<br />

density first cycle: 126 Pf/ml, t-test p=0.001). Generally, multiplication rates<br />

between volunteers were similar, as is clear from the parallel growth in Figure<br />

1B.<br />

Blood-stage multiplication rate was calculated by dividing the mean parasitemia<br />

in the second cycle by the first cycle (subtraction of log-transformed values) for<br />

every individual volunteer. The group geometric mean multiplication rate was<br />

10.9 (1.04 log, SD 0.33, n=40). Similarly, the ratio of the third with the second<br />

123


124 Chapter 6<br />

cycle gave a multiplication rate of 10.8 (1.03 log, SD 0.21, n=9) <strong>and</strong> the square<br />

root of third with the first cycle ratio gave an estimate of 8.9 (0.94 log, SD 0.25,<br />

n=9), Figure 1C. Multiplication rates were not significantly different between<br />

trials (ANOVA p=0.1) nor between multiplication cycles (ANOVA p=0.6).<br />

Results for power calculations are shown in Table 1. The vaccine effect SD hardly<br />

affected group size. The power of small sporozoite challenge trials to detect<br />

vaccine inhibition was slightly higher for erythrocytic vaccines as compared to<br />

pre-erythrocytic vaccines, due to smaller inter-individual variation in blood-stage<br />

multiplication rate. Sporozoite challenge trials with a group size of 6-7 subjects<br />

could be powered to detect 80% inhibiting erythrocytic <strong>and</strong> pre-erythrocytic<br />

vaccines. Drawing additional blood samples up to thrice daily enhanced the<br />

sensitivity of sporozoite challenge trials, allowing a reduced group size<br />

particularly in less efficacious vaccines.<br />

In order to translate biological effects into clinical efficacy, we estimated the<br />

threshold of inhibition necessary to give clinical <strong>protection</strong>. Clinical <strong>protection</strong><br />

was defined as volunteers that do not develop microscopic blood-stage<br />

parasitemia. For erythrocytic vaccines, we estimate that 90% inhibition results in<br />

a multiplication rate of ≤1 [6]. Depending on variation in vaccine effect, 20-25%<br />

of volunteers will cross this threshold in a trial with seven volunteers per group.<br />

Similarly, we hypothesize that parasitemia will have to be reduced to a<br />

somewhat arbitrary value 1 Pf/ml in pre-erythrocytic vaccines for clinical<br />

<strong>protection</strong>; the remaining low blood-stage parasite loads may possibly induce<br />

erythrocytic immunity [11]. Depending on the variation of vaccine effect, 16-30%<br />

of volunteers will cross this threshold in trials with seven volunteers per group<br />

(Table 1).<br />

Discussion<br />

The current data show that small sporozoite challenge trials, typically including<br />

seven subjects per group, are powered to detect biological effects induced by<br />

both pre-erythrocytic <strong>and</strong> erythrocytic c<strong>and</strong>idate vaccines that may herald<br />

clinical <strong>protection</strong>. We thus show that sporozoite challenge trials are a very<br />

sensitive tool to evaluate preliminary efficacy of any pre-erythrocytic or<br />

erythrocytic c<strong>and</strong>idate malaria vaccine.


Efficacy of pre-erythrocytic <strong>and</strong> blood-stage malaria vaccines can be assessed in small<br />

sporozoite challenge trials in human volunteers<br />

The decreasing malaria incidence in several endemic areas [12] has increased<br />

the size <strong>and</strong> costs of Phase IIb malaria vaccine field trials. Sporozoite challenges<br />

can be used for preliminary efficacy selection of c<strong>and</strong>idates. Efficacy goals set by<br />

the Malaria Vaccine Roadmap <strong>and</strong> the target product profile of RTS,S, the only<br />

c<strong>and</strong>idate currently in phase III clinical development, dictate a protective<br />

efficacy of more than 80% <strong>against</strong> clinical disease [13] <strong>and</strong> a 50% reduction of<br />

clinical attacks due to Pf respectively [14]. So called all-or-nothing vaccines or<br />

very effective “leaky” vaccines will induce full <strong>protection</strong> <strong>and</strong> can thus easily be<br />

tested by controlled <strong>infection</strong>. However, in addition to these promising vaccines,<br />

vaccine developers may be interested in less efficacious vaccines when<br />

combining antigens. Such approach is currently followed, for example, for the<br />

malaria vaccine ME-TRAP [15]. We show that in these vaccines, Q-PCR may be<br />

useful to improve the power of sporozoite challenge trials to detect 80-90%<br />

parasite inhibition. These biological effects correspond with clinical <strong>protection</strong> in<br />

a smaller proportion of subjects.<br />

We show that the sensitivity of sporozoite challenge trials is limited by variation<br />

in vaccine effect, inter-individual variation in parasite development <strong>and</strong> the<br />

limited time window during which blood-stage parasitemia can be assessed.<br />

Inter-individual variation is the most important determinant. Three factors may<br />

account for this variation: the parasite inoculum size, parasite fitness <strong>and</strong> human<br />

(innate) immune factors. St<strong>and</strong>ardizations of these points would likely decrease<br />

variance. For this purpose, clinical trials testing the needle administration of a<br />

predefined number of viable sporozoites are currently planned (NCT01086917).<br />

As an alternative, challenge trials using low numbers of erythrocytic stage<br />

parasites are conducted, allowing for a longer time window of follow-up [16].<br />

Careful analyses of Q-PCR data from several of such trials are needed to quantify<br />

their benefit.<br />

We performed calculations using simplified methodology based on crude<br />

empirical data, taking into account a limited number of variables. The power of<br />

sporozoite challenge trials may be higher if inhibiting effects transcend parasite<br />

developmental stage when antigens are shared or combined between stages, or<br />

when inter-individual variation is limited. More complicated statistical models,<br />

such as previously described by Hermsen et al. [6] <strong>and</strong> Bejon et al. [17], take into<br />

account more variables <strong>and</strong> may subsequently generate data with higher<br />

precision <strong>and</strong> power, but also require more assumptions <strong>and</strong> may be less<br />

reliable.<br />

125


126 Chapter 6<br />

The community is in urgent need of criteria for selection of vaccine c<strong>and</strong>idates<br />

that warrant clinical efficacy trials in the field. Knowing that sporozoite challenge<br />

trials can not only evaluate pre-erythrocytic vaccines, but can also confidently<br />

assess asexual erythrocytic malaria vaccine c<strong>and</strong>idates is an assuring<br />

consummation to the malaria vaccine development community.<br />

Acknowledgements<br />

We acknowledge the support of the team from the Clinical Centre for Malaria<br />

Studies <strong>and</strong> the volunteers participating in the challenge trials. We would like to<br />

thank Teun Bousema for his critical review of the manuscript.


Efficacy of pre-erythrocytic <strong>and</strong> blood-stage malaria vaccines can be assessed in small<br />

sporozoite challenge trials in human volunteers<br />

References<br />

1. Sauerwein RW, Roestenberg M, Moorthy VS. <strong>Experimental</strong> human challenge<br />

<strong>infection</strong>s can accelerate clinical malaria vaccine development. Nat Rev<br />

Immunol 2011; 11:57-64.<br />

2. Hermsen CC, Telgt DS, Linders EH, et al. Detection of Plasmodium falciparum<br />

malaria parasites in vivo by real-time quantitative PCR. Mol Biochem Parasitol<br />

2001; 118:247-251.<br />

3. Olotu A, Lusingu J, Leach A, et al. Efficacy of RTS,S/AS01E malaria vaccine <strong>and</strong><br />

exploratory analysis on anti-circumsporozoite antibody titres <strong>and</strong> <strong>protection</strong> in<br />

children aged 5-17 months in Kenya <strong>and</strong> Tanzania: a r<strong>and</strong>omised controlled<br />

trial. Lancet Infect Dis 2011; 11:102-109.<br />

4. Cummings JF, Spring MD, Schwenk RJ, et al. Recombinant Liver Stage Antigen-1<br />

(LSA-1) formulated with AS01 or AS02 is safe, elicits high titer antibody <strong>and</strong><br />

induces IFN-gamma/IL-2 CD4+ T cells but does not protect <strong>against</strong> experimental<br />

Plasmodium falciparum <strong>infection</strong>. Vaccine 2010; 28:5135-5144.<br />

5. Genton B, D'Acremont V, Lurati-Ruiz F, et al. R<strong>and</strong>omized double-blind<br />

controlled Phase I/IIa trial to assess the efficacy of malaria vaccine PfCS102 to<br />

protect <strong>against</strong> challenge with P. falciparum. Vaccine 2010; 28:6573-6580.<br />

6. Hermsen CC, de Vlas SJ, van Gemert GJ, Telgt DS, Verhage DF, Sauerwein RW.<br />

Testing vaccines in human experimental malaria: statistical analysis of<br />

parasitemia measured by a quantitative real-time polymerase chain reaction.<br />

Am J Trop Med Hyg 2004; 71:196-201.<br />

7. Spring MD, Cummings JF, Ockenhouse CF, et al. Phase 1/2a study of the malaria<br />

vaccine c<strong>and</strong>idate apical membrane antigen-1 (AMA-1) administered in<br />

adjuvant system AS01B or AS02A. PLoS One 2009; 4:e5254.<br />

8. Heppner DG, Jr., Kester KE, Ockenhouse CF, et al. Towards an RTS,S-based,<br />

multi-stage, multi-antigen vaccine <strong>against</strong> falciparum malaria: progress at the<br />

Walter Reed Army Institute of Research. Vaccine 2005; 23:2243-2250.<br />

9. White MT, Griffin JT, Drakeley CJ, Ghani AC. Heterogeneity in malaria exposure<br />

<strong>and</strong> vaccine response: implications for the interpretation of vaccine efficacy<br />

trials. Malar J 2010; 9:82.<br />

10. Ponnudurai T, Lensen AH, van Gemert GJ, Bensink MP, Bolmer M, Meuwissen<br />

JH. Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes.<br />

Parasitology 1989; 98 Pt 2:165-173.<br />

11. Pombo DJ, Lawrence G, Hirunpetcharat C, et al. Immunity to malaria after<br />

administration of ultra-low doses of red cells infected with Plasmodium<br />

falciparum. Lancet 2002; 360:610-617.<br />

12. World Malaria Report. Available at: http://www.who.int/malaria. Accessed 28<br />

July 2011.<br />

13. Malaria Vaccine Roadmap. Available at:<br />

http://www.malariavaccine.org/malvac-roadmap.php. Accessed 28 July 2011.<br />

14. Target Product Profile RTS`S. Available at: http://www.who.int/vaccinesdocuments/DocsPDF04/www773.pdf.<br />

Accessed 28 July 2011.<br />

127


128 Chapter 6<br />

15. Dunachie SJ, Walther M, Epstein JE, et al. A DNA prime-modified vaccinia virus<br />

ankara boost vaccine encoding thrombospondin-related adhesion protein but<br />

not circumsporozoite protein partially protects healthy malaria-naive adults<br />

<strong>against</strong> Plasmodium falciparum sporozoite challenge. Infect Immun 2006;<br />

74:5933-5942.<br />

16. S<strong>and</strong>erson F, Andrews L, Douglas AD, Hunt-Cooke A, Bejon P, Hill AV. Bloodstage<br />

challenge for malaria vaccine efficacy trials: a pilot study with discussion<br />

of safety <strong>and</strong> potential value. Am J Trop Med Hyg 2008; 78:878-883.<br />

17. Bejon P, Andrews L, Andersen RF, et al. Calculation of liver-to-blood inocula,<br />

parasite growth rates, <strong>and</strong> preerythrocytic vaccine efficacy, from serial<br />

quantitative polymerase chain reaction studies of volunteers challenged with<br />

malaria sporozoites. J Infect Dis 2005; 191:619-626.


Chapter 7<br />

NF135.C10: a new Plasmodium falciparum<br />

clone for controlled human malaria<br />

<strong>infection</strong>s<br />

Anne C. Teirlinck 1 *, Meta Roestenberg 1 * ± , Marga van de Vegte-Bolmer 1 , Anja<br />

Scholzen 1 , Moniek J.L. Heinrichs 1 , Rianne Siebelink-Stoter 1 , Wouter Graumans 1 ,<br />

Geert-Jan van Gemert 1 , Karina Teelen 1 , Martijn W. Vos 1 , Krystelle Nganou-<br />

Makamdop 1 , Steffen Borrmann 2± , Yol<strong>and</strong>a P.A. Rozier 3 , Marianne A.A. Erkens 3 ,<br />

Adrian J.F. Luty 1± , Cornelus C. Hermsen 1 , B. Kim Lee Sim 6 , Lisette van Lieshout 3,4 ,<br />

Stephen L. Hoffman 6 , Leo G. Visser 5 , Robert W. Sauerwein 1<br />

*The authors contributed equally to this work<br />

1<br />

Radboud University Nijmegen Medical Centre, Department of Medical<br />

Microbiology, Nijmegen, The Netherl<strong>and</strong>s<br />

2<br />

Department of Infectious Diseases, Heidelberg University School of Medicine,<br />

Heidelberg, Germany<br />

3<br />

Department of Medical Microbiology (Clinical Microbiology Laboratory), Leiden<br />

University Medical Centre, Leiden, The Netherl<strong>and</strong>s<br />

4<br />

Department of Parasitology, Leiden University Medical Centre, Leiden, The<br />

Netherl<strong>and</strong>s<br />

5<br />

Department of Infectious Diseases, Leiden University Medical Centre, Leiden, The<br />

Netherl<strong>and</strong>s<br />

6 Sanaria Inc., Rockville, Maryl<strong>and</strong>, United States of AmericaJ.<br />

J.Infect. Dis 2012


130 Chapter 7<br />

Abstract<br />

The portfolio of Plasmodium falciparum (Pf) strains for controlled human<br />

malaria <strong>infection</strong>s (CHMI) primarily consists of NF54 <strong>and</strong> its clone 3D7.<br />

Availability of additional strains will allow better evaluation of efficacy of<br />

experimental vaccines <strong>and</strong> drugs, <strong>and</strong> underst<strong>and</strong>ing of immunological<br />

<strong>protection</strong> mechanisms.<br />

NF135 was isolated from a Dutch patients who acquired malaria whilst travelling<br />

in Cambodia. Parasites were adapted to continuous culture <strong>and</strong> cloned. Clone<br />

NF135.C10 consistently produced gametocytes <strong>and</strong> generated substantial<br />

numbers of sporozoites in Anopheles mosquitoes. NF135.C10 was tested in a<br />

CHMI trial in comparison with NF54 for parasitological, immunological <strong>and</strong><br />

clinical parameters. Two groups of five malaria-naive volunteers were exposed<br />

to bites of five NF54 or NF135.C10 infected mosquitoes.<br />

NF135.C10 parasites diverged from NF54 parasites by drug sensitivity <strong>and</strong><br />

genetic marker profiles. Both NF135.C10 <strong>and</strong> NF54 were infectious to humans.<br />

Three out of five volunteers challenged with NF135.C10 <strong>and</strong> four out of five<br />

challenged with NF54 developed parasitemia after seven to eleven days as<br />

detected by microscopy. The two strains showed great similarity in prepatent<br />

period, kinetics of parasitemia, clinical features <strong>and</strong> cellular immunological<br />

responses.<br />

In conclusion, we successfully established a new field strain of Pf that can be<br />

used in CHMI <strong>and</strong> vaccine studies.


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

Introduction<br />

Malaria caused an estimated 216 million cases <strong>and</strong> around one million deaths in<br />

2010 [1, 2], mainly in sub-Saharan Africa where most cases are caused by<br />

Plasmodium falciparum (Pf). Development of vaccines <strong>and</strong> new drugs, <strong>and</strong><br />

better underst<strong>and</strong>ing of immunological processes are essential in order to tackle<br />

this immense problem. Controlled Human Malaria Infection (CHMI), in which<br />

healthy volunteers are exposed to bites of Pf-infected mosquitoes, is a powerful<br />

tool to address questions regarding Pf drug <strong>and</strong> vaccine efficacy, clinical<br />

properties, parasite kinetics <strong>and</strong> human immunology. Since the first CHMI by<br />

mosquitoes fed on cultures of Pf [3], more than 1300 healthy volunteers have<br />

been exposed to CHMI [4] with mainly either the Nijmegen Falciparum strain<br />

NF54 [5] or its clone 3D7 [6]. Strain NF54 stably produces sexual stages required<br />

for production of infectious mosquitoes. Parasites have been adapted to<br />

laboratory conditions by continuous in vitro culture for more than three<br />

decades. In the field, Pf isolates display a wide genetic diversity, which is<br />

currently not represented by the available laboratory strains for CHMI. Other<br />

strains, such as the South American 7G8 Pf clone [7, 8] of the Brazilian isolate<br />

IMTM22 [9] have been sporadically used in limited number of volunteers [10-<br />

12]. We therefore aimed to identify <strong>and</strong> test an additional Pf strain that can be<br />

used in CHMIs, <strong>and</strong> developed several qualification criteria: i) The strain must<br />

consistently produce gametocytes <strong>and</strong> sporozoites. ii) The strain should be<br />

cloned to create a single genetically homogenous parasite population <strong>and</strong><br />

should be genetically characterized. iii) The clone should be sensitive to<br />

commonly administered antimalarials. iv) It should be of non-African origin in<br />

order to be geographically <strong>and</strong> genetically distinct from the NF54 strain, an<br />

airport strain that probably originates from Africa [13, 14]. Here we report the<br />

generation, characterization <strong>and</strong> first CHMI with NF135.C10, a new Cambodian<br />

clone, including drug sensitivity, microsatellite profile, kinetics of parasitemia,<br />

clinical features <strong>and</strong> immunological properties in a direct comparison with NF54.<br />

Methods<br />

Culturing of NF135.C10 <strong>and</strong> NF54 parasites<br />

Parasites were cultured as described previously [15]. Briefly, NF135.C10 <strong>and</strong><br />

NF54 Pf asexual blood-stage parasites, regularly screened for mycoplasma<br />

contamination, were grown in RPMI-1640 medium containing 10% human A +<br />

131


132 Chapter 7<br />

serum at 5% haematocrit in a semi-automated suspension culture system, in the<br />

absence of antibiotics <strong>and</strong> in an atmosphere containing 4% CO2 <strong>and</strong> 3% O2.<br />

Cloning of NF135 was performed in 96-wells plates by limiting dilution [16].<br />

Characterization of NF135.C10 <strong>and</strong> NF54<br />

Genetic identity of NF135.C10 <strong>and</strong> NF54 was defined by PCR <strong>and</strong> microsatellite<br />

mapping. The polymorphic regions of three Pf antigen genes were assessed in a<br />

method adapted from Snounou et al. [17]. Briefly, parasite DNA was isolated<br />

using QIAamp DNA Blood Mini Kit (Qiagen) <strong>and</strong> amplified with thermoperfect<br />

taq polymerase using specific primers for GLURP, MSP1 K1, MSP1 MAD20 <strong>and</strong><br />

MSP2 IC (all primers from Invitrogen). For microsatellite mapping, the repetitive<br />

element rif MS (pfRRM), a polymorphic microsatellite marker [18] was used to<br />

compare NF135.C10 with NF54. The NF135.C10 <strong>and</strong> NF54 genomic DNA used<br />

was derived from respective master cell banks produced in compliance with<br />

cGMPs at Sanaria. PCR amplification was performed using fluorescently labelled<br />

forward primer 5’-TACGTTACATTATGTTTTA-3’ <strong>and</strong> reverse primer 5’-<br />

ATATGTATTGCGCTTTTA-3’. PCR products generated from each sample were<br />

separated by capillary electrophoresis on an Applied Biosystems 3100 Genetic<br />

Analyzer. A spectral image was generated with the Genemapper software V4.0.<br />

with each individual peak in the spectral image representing a PCR product. The<br />

base pair (bp) size of the amplified sequence products gave a pattern<br />

(fingerprint) that was unique to each malaria strain [18, 19]. Sensitivity of<br />

NF135.C10 <strong>and</strong> NF54 to dihydroartemisinin (DHA; SigmaTau), chloroquine<br />

diphosphate salt (Sigma-Aldrich), proguanil (British <strong>Pharma</strong>copia), atovaquone<br />

(GSK) <strong>and</strong> lumefantrine (Novartis) was tested by the Malaria SYBR Green I-<br />

Based Fluorescence Assay in triplicate experiments [20].<br />

Production of NF135.C10 <strong>and</strong> NF54 infected mosquitoes<br />

Gametocyte suspensions of NF135.C10 or NF54 were fed to Anopheles stephensi<br />

mosquitoes reared according to st<strong>and</strong>ard operating procedures as described<br />

previously [21]. To assess the rate of <strong>infection</strong>, salivary gl<strong>and</strong>s of ten mosquitoes<br />

were dissected for each strain to confirm the presence of sporozoites.<br />

Inclusion, <strong>infection</strong> <strong>and</strong> clinical follow-up of volunteers<br />

Volunteers, aged 18-35, were recruited public by advertisement <strong>and</strong> screened at<br />

the Leiden University Medical Centre (LUMC) for eligibility based on medical <strong>and</strong><br />

family history, physical examination <strong>and</strong> general haematological <strong>and</strong><br />

biochemical tests including HIV, hepatitis B <strong>and</strong> hepatitis C serology, urine


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

NF135.C10<br />

Session<br />

# mosquitoes<br />

offered<br />

# mosquitoes<br />

with infected<br />

bites<br />

1060-09 s1 5 5<br />

total 5 5<br />

1065-10 s1 5 3<br />

s2 2 1<br />

s3 1 1<br />

total 8 5<br />

1083-15 s1 5 4<br />

s2 1 0<br />

s3 1 1<br />

total 7 5<br />

1109-19 s1 5 4<br />

s2 1 1<br />

total 6 5<br />

1114-21 s1 5 4<br />

s2 1 1<br />

total 6 5<br />

NF54<br />

1004-03 s1 5 5<br />

total 5 5<br />

1089-18 s1 5 5<br />

total 5 5<br />

1094-16 s1 5 4<br />

s2 1 1<br />

total 6 5<br />

1095-25 s1 5 5<br />

total 5 5<br />

1100-26 s1 5 5<br />

total 5 5<br />

Supplementary Table 1. Feeding sessions on volunteers by mosquitoes infected<br />

with either NF135.C10 or NF54 parasites. Sessions for every individual volunteer<br />

are displayed for either NF135.C10 (upper panel) or NF54 (lower panel) groups.<br />

Sessions were repeated with a smaller number of mosquitoes until precisely five<br />

infected mosquitoes per volunteer had bitten.<br />

toxicology <strong>and</strong> pregnancy. Main exclusion criteria were residence in a malaria<br />

endemic area within the previous six months, positive Pf serology [22] or an<br />

estimated ten year risk of >5% of developing a cardiac event as estimated by the<br />

Systematic Coronary Evaluation system. All volunteers gave written informed<br />

consent prior to inclusion.<br />

Ten Dutch malaria-naïve volunteers were included <strong>and</strong> after r<strong>and</strong>omization two<br />

groups of five volunteers each were exposed to bites of Anopheles stephensi<br />

mosquitoes infected with either NF54 or NF135.C10 for ten minutes at the<br />

133


134 Chapter 7<br />

insectary of the Radboud University Nijmegen Medical Centre. Feeding sessions<br />

were repeated when necessary, with a smaller number of mosquitoes until each<br />

volunteer had been exposed to exactly five mosquitoes that took a blood meal<br />

<strong>and</strong> had Pf sporozoites in their salivary gl<strong>and</strong>s. One feeding session was<br />

sufficient for one <strong>and</strong> four volunteers in the NF135.C10 <strong>and</strong> NF54 groups<br />

respectively, two <strong>and</strong> one volunteers required two sessions <strong>and</strong> three feeding<br />

sessions were needed in three volunteers of the NF135.C10 group<br />

(Supplementary Table 1). Starting from day five post-<strong>infection</strong>, volunteers were<br />

subjected to intensive follow-up with up to thrice daily visits to the LUMC outpatient<br />

clinical research department. All signs <strong>and</strong> symptoms (solicited <strong>and</strong><br />

unsolicited) were recorded <strong>and</strong> graded by the attending physician as follows:<br />

mild (easily tolerated), moderate (interferes with normal activity), or severe<br />

(prevents normal activity), or in case of fever grade 1 (>37.5°C – 38.0°C), grade 2<br />

(>38.0°C – 39.0°C) or grade 3 (>39.0°C). Haematological <strong>and</strong> biochemical<br />

parameters were monitored daily. Because of a previously reported serious<br />

cardiac adverse event after a malaria challenge <strong>infection</strong> in a separate study<br />

[23], particular attention was paid to markers of coagulation or cardiac damage<br />

with daily follow-up of highly sensitive troponin, platelets, d-dimer <strong>and</strong> lactate<br />

dehydrogenase during the period of expected blood stage parasitemia.<br />

Whenever abnormal, blood samples were checked for the presence of<br />

fragmentocytes <strong>and</strong> von Willebr<strong>and</strong> cleaving protease activity. Promptly after<br />

identification of a positive blood smear, volunteers were treated with a curative<br />

regimen of four tablets of 250/100mg atovaquone/proguanil once daily for three<br />

days. Volunteers whose blood smears remained free of parasites until day 21<br />

after challenge presumptively received the same curative treatment with followup<br />

to the end of the study at day 28. Complete cure was always confirmed by<br />

two consecutive parasite-negative blood smears. The trial was performed in<br />

accordance with Good Clinical Practice <strong>and</strong> approved by the Central Committee<br />

for Research Involving Human Subjects of The Netherl<strong>and</strong>s (CCMO<br />

NL30350.058.09). Clinicaltrials.gov identifier: NCT01002833.<br />

Parasitological Outcomes<br />

Thick blood smears were examined by microscopy twice daily on days five <strong>and</strong><br />

six post-challenge, thrice daily on days seven to eleven, twice daily on days 12-<br />

15 <strong>and</strong> once daily on days 16-21 post-challenge. 15µl of EDTA-anti-coagulated<br />

blood was spread over the st<strong>and</strong>ardised surface of one well of a 3-well glass<br />

slide (CEL-LINE Diagnostic Microscope Slides, 30-12A-black-CE24). After drying,<br />

wells were stained with Giemsa for 30 minutes. Slides were read at 1000x


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

magnification by assessing 123 high-power fields, equal to approximately 0.5µl<br />

of blood. A smear was considered positive if two unambiguously identifiable<br />

parasites were found. Pre-patent period was defined as the period between<br />

exposure to infected mosquitoes <strong>and</strong> first positive blood smear. Additionally,<br />

parasitemia was measured by real-time quantitative PCR (qPCR), performed<br />

retrospectively on all samples collected after challenge, as described previously<br />

[24] with minor changes: using the MGB probe AAC AAT TGG AGG GCA AG<br />

instead of the turbo TaqMan probe sequence.<br />

In vitro immunological assays<br />

For antigen preparation, asynchronous asexual-stage cultures of NF135.C10 <strong>and</strong><br />

NF54 parasites were harvested at parasitemias of approximately 5-10% <strong>and</strong><br />

mature asexual stages were purified by centrifugation on a 27% <strong>and</strong> 63% Percoll<br />

density gradient [25] resulting in preparations of 80-90% parasitemia with >95%<br />

schizonts/mature trophozoites. Preparations of parasitized red blood cells<br />

(PfRBC) were washed twice in PBS <strong>and</strong> cryopreserved at 150x10 6 /ml in 15%<br />

glycerol/PBS in aliquots for use in individual stimulation assays. Mock-cultured<br />

uninfected erythrocytes (uRBC) were obtained similarly <strong>and</strong> served as controls.<br />

For cellular immunology, venous whole blood was collected into citrated<br />

vacutainer CPT cell preparation tubes (Becton <strong>and</strong> Dickinson) on the day prior to<br />

challenge (C-1), on days 5, 35 <strong>and</strong> 140 after challenge <strong>and</strong> on the first day of<br />

treatment (DT). Peripheral blood mononuclear cells (PBMCs) were isolated by<br />

density gradient centrifugation, washed twice in PBS, enumerated, frozen in<br />

foetal-calf serum containing 10% dimethylsulfoxide <strong>and</strong> stored in liquid nitrogen.<br />

After thawing, 500.000 PBMCs were cultured in the presence of NF135.C10 or<br />

NF54 PfRBC at a 1:2 (PBMC:PfRBC) ratio for 24 hours with addition of Brefeldin A<br />

(Sigma-Aldrich) for the last four hours. Cells were harvested <strong>and</strong> subsequently<br />

stained for viability (Live/Dead fixable dead cell stain kit Aqua, Invitrogen) <strong>and</strong><br />

surface markers; either 1) CD4 PE (SK3, BioLegend), CD45RO ECD (UCHL1,<br />

Beckman-Coulter), CD3 PerCP (UCHT1, BioLegend), CD62L PeCy7 (DREG-56,<br />

eBioscience) <strong>and</strong> CD8a Alexa Fluor 700 (HIT8A, BioLegend) or 2) anti-TCR Pan<br />

γ/δ-PE (IMMU510, Beckman-Coulter), CD45RO ECD, CD3 PerCP, CD4 PECy7<br />

(RPA-T4, BioLegend), Biotin CD56 (HCD56, BioLegend) <strong>and</strong> CD8a Alexa Fluor 700.<br />

Cells were washed <strong>and</strong> the second staining panel was incubated with<br />

Streptavidin eFluor450 (eBioscience). After washing, all cells were incubated<br />

with fixation Medium A (Caltag) <strong>and</strong> subsequently stained with either 1) IFNγ<br />

FITC (4S.B3, eBioscience), TNF Pacific Blue (MAb11, BioLegend), IL-2 APC (MQ1-<br />

135


136 Chapter 7<br />

NF135.C10 NF54<br />

Re-started cultures until CHMI 7 306<br />

Period 2009-2010<br />

Infection 74% (62-87) 86% (78-94)<br />

Oocysts 12 (7.3-16) 27 (22-33)<br />

Sporozoites/mosquito x10 3 39 (18-60) 99 (74-124)<br />

CHMI (April 2010)<br />

Infection 100% 100%<br />

Oocysts 5.6 17<br />

Sporozoites/mosquito x10 3 12.5 69<br />

Gametocyte male: female ratio 1:5 1:3<br />

Drug sensitivity (IC50)<br />

dihydroartemisinin 3.4 nM 9.9 nM<br />

lumefantrine 89 nM 78 nM<br />

proguanil 21 µM 27 µM<br />

atovaquone 0.3 nM 0.6 nM<br />

chloroquine 201 nM 24 nM<br />

Table 1. Mosquito <strong>infection</strong> <strong>and</strong> drug sensitivity profile of NF135.C10 <strong>and</strong> NF54 in<br />

the period 2009-2010 <strong>and</strong> for the specific batches used in this CHMI. Data are<br />

displayed for the period 2009-2010 (mean (95% CI)) after 26 <strong>and</strong> 39 st<strong>and</strong>ard<br />

dissections respectively, from 10 mosquitoes per dissection. Half-inhibitory<br />

concentrations (IC50) are means from three independent experiments.<br />

17H12, eBioscience )) or 2) IFNγ FITC, in permeabilization Medium B (Caltag).<br />

Cells were read on a CyAn ADP 9-color flow cytometer (Beckman-Coulter) <strong>and</strong><br />

analysed using FlowJo software (Tree Star, Inc.) version 9.2. Gating of cytokinepositive<br />

cells was performed based on the Median Fluorescent Intensity (MFI) of<br />

cytokine negative PBMCs for each volunteer, time point <strong>and</strong> stimulus.<br />

Statistical analysis<br />

Data analysis was performed using GraphPad Prism5 software. Differences in<br />

parasite kinetics between subjects in the NF135.C10 <strong>and</strong> NF54 group were<br />

analysed using the non-parametric Mann-Whitney test. A two-sided P value of<br />

less than 0.05 was considered statistically significant.<br />

Results<br />

Generation <strong>and</strong> characterization of NF135.C10<br />

Clinical isolates of asexual Pf parasites were obtained by culturing blood from Pfmalaria<br />

patients from hospitals in The Netherl<strong>and</strong>s. We adapted 74 different


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

Figure 1. Genetic characterization of Plasmodium falciparum parasite isolates.<br />

PCR was performed to assess allelic variation on (A) MSP1 K1 <strong>and</strong> MSP1 MAD20, (B)<br />

MSP2 IC <strong>and</strong> (C) GLURP for the Pf strains NF135.C10 <strong>and</strong> NF54. Rif repetitive element<br />

(PfRR) were characterized by the microsatellite identity test on the genomic DNA of the Pf<br />

parasite strains (D) NF135.C10 <strong>and</strong> (E) NF54. The spectral image generated in the Gene<br />

Mapper showing a strain-specific microsatellite fingerprint comprising a unique peak<br />

pattern.<br />

field strains to culture, of which 21 produced gametocytes <strong>and</strong> 16 were able to<br />

successfully infect mosquitoes. Based on their ability to stably produce mature<br />

gametocytes, exflagellate <strong>and</strong> to be transmitted to mosquitoes, seven strains<br />

were cloned <strong>and</strong> two strains produced at least five oocysts <strong>and</strong> 30 000<br />

sporozoites in more than 70% of mosquitoes after feeding. Clone NF135.C10 was<br />

isolated by limiting dilution from isolate NF135, which was obtained from a<br />

Dutch traveller to Cambodia diagnosed with Pf malaria in February 1993. Culture<br />

characteristics of NF135.C10 <strong>and</strong> NF54 are displayed in Table 1.<br />

137


138 Chapter 7<br />

Figure 2. Parasite kinetics of strains NF135.C10 <strong>and</strong> NF54 assessed by<br />

thick smear <strong>and</strong> quantitative real-time PCR. Volunteers were infected by<br />

bites of mosquitoes infected with either NF135.C10 or NF54. (A) Percentage<br />

of volunteers infected with NF135.C10 (red, n=5) or NF54 (black, n=5)<br />

becoming thick smear positive, followed up up to 21 days after <strong>infection</strong>.<br />

(B) Parasitemia of volunteers until thick smear positivity, measured by<br />

qPCR, is shown as geometric mean <strong>and</strong> 95% confidence interval for<br />

volunteers infected with NF135.C10 (red) <strong>and</strong> NF54 (black), <strong>and</strong> historical<br />

controls infected with NF54 (grey area, n=48).<br />

The drug sensitivity profile of NF135.C10 is similar to NF54 for atovaquone,<br />

proguanil, DHA <strong>and</strong> lumefantrine, but NF135.C10 is more than 8-fold less sensitive<br />

to chloroquine than NF54. Comparison of NF135.C10 <strong>and</strong> NF54 genotypes


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

ANY ADVERSE<br />

EVENT<br />

NF135.C10(n=3) NF54(n=4)<br />

frequency<br />

Mean duration<br />

days (stdev)<br />

frequency<br />

Mean duration<br />

days (stdev)<br />

Smear negative(n=3)<br />

frequency<br />

Mean duration<br />

days (stdev)<br />

Abdominal pain<br />

Arthralgia<br />

Chills<br />

2 0 (0.0)<br />

Fatigue 3 2.4 (1.9) 1 3.0 -- 2 13.5 (9.3)<br />

Fever 1 0.2 -- 2 0.7 (0.8)<br />

Headache 3 1.5 (2.4) 4 2.3 (2.0) 3 4.6 (3.8)<br />

Itching<br />

4 3.1 (1.5) 2 5.2 (0.3)<br />

Malaise<br />

4 2.5 (3.3)<br />

Myalgia 1 2.7 -- 3 1.3 (1.4) 2 2.7 (2.4)<br />

Nausea 1 0.1 -- 3 2 (2.0) 1 0.0 --<br />

Vomiting<br />

1 0.4 --<br />

Any 3 1.3 (1.7) 4 2.2 (1.9) 3 5.7 (5.8)<br />

GRADE 3<br />

ADVERSE EVENT<br />

Headache<br />

1 4.6 --<br />

Malaise<br />

1 0.4 --<br />

Vomiting<br />

1 0.4 --<br />

Any<br />

1 1.8 (2.4)<br />

Table 2. Reported solicited adverse events that were considered possibly, probably, or<br />

definitely related to the trial procedures. Data is displayed as frequency per event (n)<br />

with mean duration in days (n (stdev)), separately for volunteers infected with NF135.C10<br />

(first column) or NF54 (second column) or volunteers that did not became positive by<br />

thick smear <strong>and</strong> PCR (third column).<br />

using PCR <strong>and</strong> rifin microsatellite mapping showed distinct genetic differences<br />

between the two strains (Figure 1).<br />

Controlled human malaria <strong>infection</strong><br />

Ten Dutch malaria-naive volunteers were exposed to bites of mosquitoes<br />

infected with either NF135.C10 (n=5) or NF54 (n=5). Daily follow-up until day 21<br />

revealed positive thick smears in three out of five volunteers infected with<br />

NF135.C10 <strong>and</strong> four out of five volunteers infected with NF54 parasites. The<br />

remaining three smear-negative volunteers were presumptively treated 21 days<br />

post-<strong>infection</strong> (Figure 2A).<br />

All blood samples from these three smear negative volunteers were also<br />

negative for Pf as retrospectively assessed by qPCR. In Pf positive volunteers,<br />

kinetics of parasitemia of both strains were comparable to historical controls<br />

(n=48) infected with NF54 (grey area represents 95% CI, Figure 2B [26]). Patent<br />

139


140 Chapter 7<br />

parasitemia for NF135.C10 seemed to occur slightly earlier than for NF54 as<br />

measured by thick smear (median of 7.0 days after <strong>infection</strong>; range 7.0-9.0<br />

versus median of 10.6; range 10.6-11; p=0.05; Mann-Whitney test) <strong>and</strong> qPCR<br />

(median day 7.0; range 6.3-7.0 versus median day 7.3; range 7.0-7.3; p=0.1<br />

Mann-Whitney test, Figure 2B). Particularly the peak of the first cycle seemed<br />

higher for NF135.C10 (Geometric Mean (GM) 1.2; 95% CI 0.61-2.4 parasites/µL)<br />

than NF54 (GM 0.16; 95% CI 0.055-0.46; p=0.06 Mann-Whitney test). The GM of<br />

peak parasitemia was 11 parasites/µL (95% CI 1.8-73) <strong>and</strong> 30 parasites/µL (95%<br />

CI 7.7-120) (p=0.4) for the positive volunteers of the NF135.C10 <strong>and</strong> NF54<br />

infected groups, respectively. PCR identity of both strains was confirmed by<br />

culture of smear-positive samples from several r<strong>and</strong>omly selected infected<br />

volunteers.<br />

Safety<br />

All volunteers, including the smear-negative volunteers, reported solicited<br />

adverse events (AEs) that were considered possibly or probably related to the<br />

trial procedures (Table 2). All volunteers reported headache with a mean<br />

duration of 1.5 (NF135.C10) versus 2.3 (NF54) days <strong>and</strong> most volunteers<br />

experienced fatigue, myalgia <strong>and</strong> nausea. One volunteer infected with NF54<br />

reported grade three adverse events (malaise, headache <strong>and</strong> vomiting).<br />

Duration <strong>and</strong> frequency of AEs were not different between NF135.C10 <strong>and</strong> NF54<br />

infected volunteers in this limited number of volunteers. One infected volunteer<br />

of the NF135.C10 group showed a decreased platelet count of 146x10 9 /L at day<br />

3 after treatment (cut-off 150x10 9 /L), which returned to normal values at<br />

routine check-up on day 28. D-dimers did not increase in any of the volunteers<br />

before thick smear positivity. As can be expected, four volunteers had increased<br />

d-dimers (range 632-1100 ng/mL) during treatment, that decreased to normal<br />

levels after treatment. Highly sensitive troponin T values were always below<br />

0.05 µg/L.<br />

Cellular immune responses<br />

We next compared the induction of cellular recall immune responses between<br />

volunteers infected with NF135.C10 or NF54. In 24-hour in vitro stimulation<br />

assays, T-lymphocytes of volunteers successfully infected with either NF135.C10<br />

or NF54 showed similarly increased IFNγ, TNF <strong>and</strong> IL-2 responses 35 days after<br />

<strong>infection</strong> (Figure 3A-C).<br />

Furthermore, cellular responses showed the same kinetics for both homologous<br />

(NF135.C10 infected/NF135.C10 stimulated, NF54 infected/NF54 stimulated)


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

Figure 3. Production of IFNγ, TNF <strong>and</strong> IL-2 by CD3+ T-lymphocytes after in vitro restimulation<br />

with NF135.C10 or NF54 determined before <strong>and</strong> after challenge of<br />

volunteers. Time points displayed are: before challenge (C-1), five days after challenge<br />

(C+5), on day of treatment (DT) <strong>and</strong> 35 <strong>and</strong> 140 days after challenge. PBMCs were<br />

stimulated for 24-hour with asexual stage parasites (PfRBC) of NF135.C10 or NF54.<br />

Numbers of (A,D,G) IFNγ (B,E,H) TNF <strong>and</strong> (C,F,I) IL-2 producing cells are depicted as<br />

percentages of total T-lymphocytes. (A-C) Production of cytokines after homologous<br />

stimulation of cells taken before <strong>and</strong> after challenge. Cells of volunteers infected with<br />

NF135.C10 (open red circles) were stimulated with NF135.C10 PfRBC. Cells of volunteers<br />

infected with NF54 (closed black circles) were stimulated with NF54 PfRBC. (D-F)<br />

Production of cytokines after homologous (NF135.C10, closed red circles) or<br />

heterologous (NF54, open grey circles) stimulation of PBMCs of volunteers before <strong>and</strong><br />

after challenge with NF135.C10. (G-I) Production of cytokines after homologous (NF54,<br />

closed black circles) or heterologous (NF135.C10, open grey circles) stimulation of PBMCs<br />

of volunteers before <strong>and</strong> after challenge with NF54. Symbols indicate individual values<br />

from volunteers (A-C) or represent group medians with interquartile range (D-I).<br />

141


142 Chapter 7<br />

Figure 4. Contribution of different cell types to the total number of IFNγ-producing<br />

cells upon stimulation with either homologous or heterologous PfRBC. Mean<br />

percentage contribution to the total response are displayed for PBMCs of volunteers<br />

successfully infected with NF135.C10 (n=3) or NF54 (n=4) when re-stimulated in<br />

vitro with either NF135.C10 or NF54 PfRBC. Columns show mean percentage<br />

contribution on day 35 post-challenge of (A) NK cells (CD3 - CD56 + ), NK-T γδ-T + cells<br />

(CD3 + γδ-T + CD56 + ), NK-T cells (CD3 + γδ-T - CD56 + ), γδ-T cells (CD3 + γδ-T + CD56 - ), αβ-T<br />

CD4 cells (CD3 + γδ-T - CD4 + ), <strong>and</strong> αβ-T CD8 cells (CD3 + γδ-T - CD8 + ) <strong>and</strong> (B) effector<br />

memory (EM) cells (CD3 + , CD45RO + , CD62L - ), central memory (CM) cells (CD3 + ,<br />

CD45RO + , CD62L + ) <strong>and</strong> naive T-lymphocytes (CD3 + , CD45RO - ). Responses to<br />

uninfected red blood cells are subtracted from responses to PfRBC per contributing<br />

cell type.


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

<strong>and</strong> heterologous (NF54 infected/NF135.C10 stimulated, NF135.C10<br />

infected/NF54 stimulated) in vitro re-stimulation (Figure 3D-I). IL-2<br />

responses seemed slightly higher when stimulated with NF135.C10 but this was<br />

similar for NF135.C10 <strong>and</strong> NF54 infected volunteers. IFNγ-producing cells were<br />

found in both the innate compartment (γδ-T, NK, NK-T) <strong>and</strong> the adaptive<br />

compartment (CD4 <strong>and</strong> CD8) <strong>and</strong> mainly displayed an effector memory<br />

phenotype (Figure 4). Despite inter-individual differences in relative contribution<br />

of various subsets, the composition of responding cells was highly consistent<br />

over time for every individual (data not shown). Upon homologous restimulation<br />

of cells on C+35, the vast majority of responding lymphocytes were<br />

single positive for IFNγ (mean 71%), TNF (9.4%) or double positive for IFNγ <strong>and</strong><br />

TNF (18%). Additional responding lymphocytes were single positive for IL-2<br />

(0.40%), double positive IFNγ + IL-2 + (0.21%) or IL-2 + TNF + (0.61%) or triple<br />

positive (0.50%) (data not shown).<br />

Discussion<br />

We identified <strong>and</strong> characterized NF135.C10 as the first Pf clone of Asian origin<br />

for successful <strong>infection</strong> of malaria-naive human volunteers by CHMI. Clone<br />

NF135.C10 consistently produced gametocytes in culture, <strong>and</strong> was able to<br />

generate <strong>infection</strong>s in laboratory-reared mosquitoes with high yields of<br />

sporozoites. NF135.C10 parasites were clearly distinct from NF54 parasites by<br />

drug sensitivity <strong>and</strong> established genetic marker profiles. Clinical presentation<br />

after CHMI <strong>and</strong> characteristics of PfRBC-specific recall (T-)lymphocyte responses<br />

in vitro were similar to NF54.<br />

Selection <strong>and</strong> identification of field strain parasites for CHMI poses technical<br />

difficulties, because of insufficient <strong>and</strong> unstable production of infectious sexual<br />

<strong>and</strong> sporogonic stages. For manufacturing purposes, cultures should ideally<br />

produce gametocytes that consistently infect at least 75% of the mosquitoes<br />

with at least ten oocysts resulting in 10-30 thous<strong>and</strong> sporozoites/mosquito. Only<br />

after extensive effort on more than seventy isolates were we able to successfully<br />

identify a parasite clone, NF135.C10, that met these criteria. Until now, the<br />

culture of NF135.C10 has only been re-started (using previous cryopreserved<br />

batches) seven times, as compared to 306 times for the NF54, <strong>and</strong> is therefore<br />

more closely related to the original clone. Also the geographical <strong>and</strong> molecular<br />

divergence from the NF54 strain makes this new clone an attractive c<strong>and</strong>idate<br />

for use in CHMI trials. The vast majority of CHMI trials have been carried out<br />

143


144 Chapter 7<br />

with either the NF54 strain or its clone 3D7 (~1300 volunteers [4, 27]) as<br />

opposed to only 42 volunteers challenged with strain 7G8 of South American<br />

origin [7, 8, 27]. 3D7 is derived from NF54 <strong>and</strong> cannot be distinguished from<br />

NF54 by drug sensitivity testing, simple genetic markers, or microsatellite<br />

mapping (Sim et al., unpublished). Thus, clinical trials with more genetically<br />

distinct parasite strains including 7G8 or NF135.C10 will be important to<br />

complement current knowledge on heterologous Pf strains. Parasitological <strong>and</strong><br />

clinical findings after CHMI with NF54 or 3D7 were recently compared in two<br />

meta-analyses (Roestenberg et al. PLoS One, in press)[28]. Here we show that<br />

clinical signs <strong>and</strong> symptoms induced by NF135.C10 or NF54 do not show any<br />

differences in successfully infected volunteers. We found slight differences in<br />

infectivity of the parasites; in these small groups of volunteers, NF135.C10<br />

showed a marginally shorter prepatent period, but parasite kinetics of both<br />

NF135.C10 <strong>and</strong> NF54 were both within the limits of historical NF54 controls.<br />

Notably, not all volunteers exposed to NF54 infected mosquitoes became<br />

parasitemic, as assessed by both microscopy <strong>and</strong> qPCR in contrast to 22 previous<br />

CHMI trials infecting 128 naive volunteers with NF54 parasites (Roestenberg et<br />

al. PLoS One, in press). Unsuccessful <strong>infection</strong> after bites of five mosquitoes has<br />

been described previously for 3D7 [29, 30]. Although the exact reason is unclear,<br />

this might due to the unusual low NF135.C10 <strong>and</strong> NF54 oocyst <strong>and</strong> sporozoite<br />

counts per mosquito obtained for this trial compared to our routinely obtained<br />

counts (table 1). A technical disturbance in our cultures, leading to suboptimal<br />

culture circumstances prior to this study was most likely the reason for this<br />

relatively low mosquito <strong>infection</strong> with both strains. However, a formal<br />

relationship between sporozoite counts <strong>and</strong> mosquito infectivity has never been<br />

established [31] <strong>and</strong> only small percentages of sporozoites in salivary gl<strong>and</strong>s are<br />

injected during a blood meal of a mosquito [32-34]. Surprisingly, all three<br />

unsuccessfully infected volunteers reported AEs that were considered possibly,<br />

probably, or definitely related to the trial procedures. Symptoms reported by<br />

these volunteers might have been the result of over-reporting in an intense<br />

follow-up schedule, concurring with our findings from a previous study, in which<br />

volunteers reported malaria-related symptoms after exposure to bites of only<br />

uninfected mosquitoes [35].<br />

Previously, we reported sustained IFNγ re-call responses in vitro in both αβ-T<br />

cells <strong>and</strong> γδ-T cells upon homologous PfRBC re-stimulation with NF54 asexual<br />

stage parasites after a single CHMI, suggestive of cross talk between innate <strong>and</strong><br />

adaptive compartments with induction of memory [36, 37]. Here we show<br />

similar kinetics <strong>and</strong> composition of IFNγ responses upon homologous Pf54 <strong>and</strong>


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

Pf135.C10 re-stimulation. Notably, responses to heterologous re-stimulation<br />

were very similar to homologous responses, both in kinetics <strong>and</strong> magnitude.<br />

Considerable genetic diversity has been found in a number of important malaria<br />

antigens <strong>and</strong> vaccine c<strong>and</strong>idates, especially in blood stages [38], but also in preerythrocytic<br />

stages [39]. Specific (conserved) antigens may be responsible for<br />

induction <strong>and</strong> maintenance of heterologous memory responses <strong>against</strong> Pf [40,<br />

41]; conserved epitopes have indeed been found in many immunogenic proteins<br />

[42-44], although some being cryptic <strong>and</strong> therefore not available for the immune<br />

system [45]. Albeit tested in only a small number of volunteers, (partial)<br />

<strong>protection</strong> has previously been reported after heterologous challenge <strong>infection</strong><br />

following immunisation with radiation-attenuated sporozoites [10, 46, 47] or a<br />

previous experimental <strong>infection</strong> [48]. Whether the heterologous T-lymphocyte<br />

responses observed in our volunteers also translate into or represent crossstrain<br />

protective immunity in vivo remains to be investigated.<br />

The availability of NF135.C10 increases the portfolio of Pf parasites that can be<br />

used in CHMI. Obtaining additional multiple genetically distinct, fully<br />

characterized <strong>and</strong> sequenced Pf clones will be important in the evaluation<br />

process of diversity covering sub-unit vaccines or whole-parasite based vaccine<br />

approaches [4], intended to protect <strong>against</strong> all Pf parasite strains in nature [49].<br />

Additionally, if immunization with whole sporozoite vaccines based on one<br />

single Pf strain does not fully protect <strong>against</strong> CHMI with heterologous Pf<br />

parasites, it will be necessary to determine whether immunization with<br />

sporozoites from a combination of clones are required to achieve such<br />

<strong>protection</strong>. Finally, to take advantage of our recent demonstration that CHMIs<br />

can also be successfully conducted by needle <strong>and</strong> syringe inoculation of aseptic,<br />

purified, cryopreserved Pf sporozoites called PfSPZ Challenge (Roestenberg et<br />

al., submitted), we have established the master <strong>and</strong> working cell banks, <strong>and</strong><br />

manufacturing process required to produce PfSPZ Challenge using NF135.C10<br />

parasites (Sim et al., unpublished).<br />

In conclusion, increasing the portfolio of new Pf parasite strains, as achieved<br />

here for NF135.C10, will accelerate the evaluation of malaria vaccines<br />

c<strong>and</strong>idates by facilitating the downstream selection process for further clinical<br />

vaccine development. Although more trials will be necessary to fine-tune the<br />

heterologous CHMI model with strain NF135.C10, the current results will boost<br />

the continued application of CHMIs as a crucial tool for malaria vaccine<br />

development.<br />

145


146 Chapter 7<br />

Acknowledgements<br />

We thank the volunteers for their enthusiastic participation in this trial <strong>and</strong> Kitty<br />

Suijk for her nursing support. We thank Laura Pelser, Jol<strong>and</strong>a Klaassen, Astrid<br />

Pouwelsen <strong>and</strong> Jacqueline Kuhnen for their work in the culturing <strong>and</strong> dissection<br />

of mosquitoes. We are indebted to all the slide readers in Leiden: Jan Kromhout,<br />

Jaco Verweij, Meriam Beljon, Jol<strong>and</strong>a van Schie, Jaqueline Schelfaut, Jeanette<br />

van der Slot, Heleen Gerritsma, Fons van der S<strong>and</strong>e, Eric Brienen, <strong>and</strong> Els van<br />

Oorschot. We thank Adriana Ahumada at Protein Potential, Jianbing Mu <strong>and</strong> Xinzhuan<br />

Su at Laboratory of Malaria <strong>and</strong> Vector Research, NIAID, NIH for the<br />

microsatellite mapping studies. Moreover, we thank Chris Janse, Shahid Khan<br />

<strong>and</strong> the malaria team for their hospitality in their laboratory in Leiden. We thank<br />

safety monitors S<strong>and</strong>ra Arend <strong>and</strong> Mark de Boer, <strong>and</strong> independent physician<br />

Frank Kroon for their continuing support.<br />

Funding<br />

This work was supported by of Top Institute <strong>Pharma</strong> [grant number T4-102]. ACT<br />

was funded by the European Malaria Vaccine Development Association, AS by a<br />

long term EMBO fellowship <strong>and</strong> KN by NWO Mozaiek [grant number<br />

017.005.011].


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

References<br />

1. WHO. World malaria report : 2011: World Health Organization, 2011.<br />

2. Murray CJ, Rosenfeld LC, Lim SS, et al. Global malaria mortality between 1980<br />

<strong>and</strong> 2010: a systematic analysis. Lancet 2012; 379:413-31.<br />

3. Chulay JD, Schneider I, Cosgriff TM, et al. Malaria transmitted to humans by<br />

mosquitoes infected from cultured Plasmodium falciparum. Am J Trop Med Hyg<br />

1986; 35:66-8.<br />

4. Sauerwein RW, Roestenberg M, Moorthy VS. <strong>Experimental</strong> human challenge<br />

<strong>infection</strong>s can accelerate clinical malaria vaccine development. Nat Rev<br />

Immunol 2011; 11:57-64.<br />

5. Ponnudurai T, Leeuwenberg AD, Meuwissen JH. Chloroquine sensitivity of<br />

isolates of Plasmodium falciparum adapted to in vitro culture. Trop Geogr Med<br />

1981; 33:50-4.<br />

6. Walliker D, Quakyi IA, Wellems TE, et al. Genetic analysis of the human malaria<br />

parasite Plasmodium falciparum. Science 1987; 236:1661-6.<br />

7. Egan JE, Hoffman SL, Haynes JD, et al. Humoral immune responses in volunteers<br />

immunized with irradiated Plasmodium falciparum sporozoites. Am J Trop Med<br />

Hyg 1993; 49:166-73.<br />

8. Heppner DG, Gordon DM, Gross M, et al. Safety, immunogenicity, <strong>and</strong> efficacy<br />

of Plasmodium falciparum repeatless circumsporozoite protein vaccine<br />

encapsulated in liposomes. J Infect Dis 1996; 174:361-6.<br />

9. Burkot TR, Williams JL, Schneider I. Infectivity to mosquitoes of Plasmodium<br />

falciparum clones grown in vitro from the same isolate. Trans R Soc Trop Med<br />

Hyg 1984; 78:339-41.<br />

10. Hoffman SL, Goh LM, Luke TC, et al. Protection of humans <strong>against</strong> malaria by<br />

immunization with radiation-attenuated Plasmodium falciparum sporozoites. J<br />

Infect Dis 2002; 185:1155-64.<br />

11. Jeffery GM, Young MD, Burgess RW, Eyles DE. Early activity in sporozoiteinduced<br />

Plasmodium falciparum <strong>infection</strong>s. Ann Trop Med Parasitol 1959;<br />

53:51-8.<br />

12. Rieckmann KH, Carson PE, Beaudoin RL, Cassells JS, Sell KW. Letter: Sporozoite<br />

induced immunity in man <strong>against</strong> an Ethiopian strain of Plasmodium falciparum.<br />

Trans R Soc Trop Med Hyg 1974; 68:258-9.<br />

13. Drakeley CJ, Duraisingh MT, Povoa M, Conway DJ, Targett GA, Baker DA.<br />

Geographical distribution of a variant epitope of Pfs48/45, a Plasmodium<br />

falciparum transmission-blocking vaccine c<strong>and</strong>idate. Mol Biochem Parasitol<br />

1996; 81:253-7.<br />

147


148 Chapter 7<br />

14. Conway DJ, Machado RL, Singh B, et al. Extreme geographical fixation of<br />

variation in the Plasmodium falciparum gamete surface protein gene Pfs48/45<br />

compared with microsatellite loci. Mol Biochem Parasitol 2001; 115:145-56.<br />

15. Ponnudurai T, Lensen AH, Leeuwenberg AD, Meuwissen JH. Cultivation of fertile<br />

Plasmodium falciparum gametocytes in semi-automated systems. 1. Static<br />

cultures. Trans R Soc Trop Med Hyg 1982; 76:812-8.<br />

16. Rosario V. Cloning of naturally occurring mixed <strong>infection</strong>s of malaria parasites.<br />

Science 1981; 212:1037-8.<br />

17. Snounou G, Zhu X, Siripoon N, et al. Biased distribution of msp1 <strong>and</strong> msp2 allelic<br />

variants in Plasmodium falciparum populations in Thail<strong>and</strong>. Trans R Soc Trop<br />

Med Hyg 1999; 93:369-74.<br />

18. Su XZ, Carucci DJ, Wellems TE. Plasmodium falciparum: parasite typing by using<br />

a multicopy microsatellite marker, PfRRM. Exp Parasitol 1998; 89:262-5.<br />

19. Liu S, Mu J, Jiang H, Su XZ. Effects of Plasmodium falciparum mixed <strong>infection</strong>s on<br />

in vitro antimalarial drug tests <strong>and</strong> genotyping. Am J Trop Med Hyg 2008;<br />

79:178-84.<br />

20. Johnson JD, Dennull RA, Gerena L, Lopez-Sanchez M, Roncal NE, Waters NC.<br />

Assessment <strong>and</strong> continued validation of the malaria SYBR green I-based<br />

fluorescence assay for use in malaria drug screening. Antimicrob Agents<br />

Chemother 2007; 51:1926-33.<br />

21. Ponnudurai T, Lensen AH, Van Gemert GJ, Bensink MP, Bolmer M, Meuwissen<br />

JH. Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes.<br />

Parasitology 1989; 98 Pt 2:165-73.<br />

22. Bousema JT, Roeffen W, van der Kolk M, et al. Rapid onset of transmissionreducing<br />

antibodies in javanese migrants exposed to malaria in Papua,<br />

Indonesia. Am J Trop Med Hygiene 2006; 74:425-31.<br />

23. Nieman AE, de Mast Q, Roestenberg M, et al. Cardiac complication after<br />

experimental human malaria <strong>infection</strong>: a case report. Malar J 2009; 8:277.<br />

24. Hermsen CC, Telgt DS, Linders EH, et al. Detection of Plasmodium falciparum<br />

malaria parasites in vivo by real-time quantitative PCR. Mol Biochem Parasitol<br />

2001; 118:247-51.<br />

25. Rivadeneira EM, Wasserman M, Espinal CT. Separation <strong>and</strong> concentration of<br />

schizonts of Plasmodium falciparum by Percoll gradients. J Protozool 1983;<br />

30:367-70.<br />

26. Roestenberg M, de Vlas SJ, Nieman AE, Sauerwein RW, Hermsen CC. Efficacy of<br />

pre-erythrocytic <strong>and</strong> blood-stage malaria vaccines can be assessed in small<br />

sporozoite challenge trials in human volunteers. J Infect Dis 2012.<br />

27. Moorthy VS, Diggs C, Ferro S, et al. Report of a consultation on the optimization<br />

of clinical challenge trials for evaluation of c<strong>and</strong>idate blood stage malaria<br />

vaccines, 18-19 March 2009, Bethesda, MD, USA. Vaccine 2009; 27:5719-25.


NF135.C10: a new Plasmodium falciparum clone for controlled human malaria<br />

<strong>infection</strong>s<br />

28. Epstein JE, Rao S, Williams F, et al. Safety <strong>and</strong> clinical outcome of experimental<br />

challenge of human volunteers with Plasmodium falciparum-infected<br />

mosquitoes: an update. J Infect Dis 2007; 196:145-54.<br />

29. Edelman R, Hoffman SL, Davis JR, et al. Long-term persistence of sterile<br />

immunity in a volunteer immunized with X-irradiated Plasmodium falciparum<br />

sporozoites. J Infect Dis 1993; 168:1066-70.<br />

30. Kester KE, McKinney DA, Tornieporth N, et al. Efficacy of recombinant<br />

circumsporozoite protein vaccine regimens <strong>against</strong> experimental Plasmodium<br />

falciparum malaria. J Infect Dis 2001; 183:640-7.<br />

31. Rickman LS, Jones TR, Long GW, et al. Plasmodium falciparum-infected<br />

Anopheles stephensi inconsistently transmit malaria to humans. Am J Trop Med<br />

Hyg 1990; 43:441-5.<br />

32. Frischknecht F, Baldacci P, Martin B, et al. Imaging movement of malaria<br />

parasites during transmission by Anopheles mosquitoes. Cell Microbiol 2004;<br />

6:687-94.<br />

33. Rosenberg R, Wirtz RA, Schneider I, Burge R. An estimation of the number of<br />

malaria sporozoites ejected by a feeding mosquito. Trans R Soc Trop Med Hyg<br />

1990; 84:209-12.<br />

34. Ponnudurai T, Lensen AH, van Gemert GJ, Bolmer MG, Meuwissen JH. Feeding<br />

behaviour <strong>and</strong> sporozoite ejection by infected Anopheles stephensi. Trans R Soc<br />

Trop Med Hyg 1991; 85:175-80.<br />

35. Roestenberg M, McCall M, Hopman J, et al. Protection <strong>against</strong> a malaria<br />

challenge by sporozoite inoculation. N Engl J Med 2009; 361:468-77.<br />

36. McCall MB, Roestenberg M, Ploemen I, et al. Memory-like IFN-gamma response<br />

by NK cells following malaria <strong>infection</strong> reveals the crucial role of T cells in NK<br />

cell activation by P. falciparum. Eur J Immunol 2010; 40:3472-7.<br />

37. Teirlinck AC, McCall MB, Roestenberg M, et al. Longevity <strong>and</strong> Composition of<br />

Cellular Immune Responses Following <strong>Experimental</strong> Plasmodium falciparum<br />

Malaria Infection in Humans. PLoS Pathog 2011; 7:e1002389.<br />

38. Good MF, Stanisic D, Xu H, Elliott S, Wykes M. The immunological challenge to<br />

developing a vaccine to the blood stages of malaria parasites. Immunol Rev<br />

2004; 201:254-67.<br />

39. Yang C, Shi YP, Udhayakumar V, et al. Sequence variations in the non-repetitive<br />

regions of the liver stage-specific antigen-1 (LSA-1) of Plasmodium falciparum<br />

from field isolates. Mol Biochem Parasitol 1995; 71:291-4.<br />

40. Borrmann S, Matuschewski K. Protective immunity <strong>against</strong> malaria by 'natural<br />

immunization': a question of dose, parasite diversity, or both? Curr Opin<br />

Immunol 2011; 23:500-8.<br />

149


150 Chapter 7<br />

41. Douradinha B, Mota MM, Luty AJ, Sauerwein RW. Cross-species immunity in<br />

malaria vaccine development: two, three, or even four for the price of one?<br />

Infect Immun 2008; 76:873-8.<br />

42. Fidock DA, Gras-Masse H, Lepers JP, et al. Plasmodium falciparum liver stage<br />

antigen-1 is well conserved <strong>and</strong> contains potent B <strong>and</strong> T cell determinants. J<br />

Immunol 1994; 153:190-204.<br />

43. Lal AA, Hughes MA, Oliveira DA, et al. Identification of T-cell determinants in<br />

natural immune responses to the Plasmodium falciparum apical membrane<br />

antigen (AMA-1) in an adult population exposed to malaria. Infect Immun 1996;<br />

64:1054-9.<br />

44. Wu T, Black CG, Wang L, Hibbs AR, Coppel RL. Lack of sequence diversity in the<br />

gene encoding merozoite surface protein 5 of Plasmodium falciparum. Mol<br />

Biochem Parasitol 1999; 103:243-50.<br />

45. Bharadwaj A, Sharma P, Joshi SK, Singh B, Chauhan VS. Induction of protective<br />

immune responses by immunization with linear multiepitope peptides based on<br />

conserved sequences from Plasmodium falciparum antigens. Infect Immun<br />

1998; 66:3232-41.<br />

46. Clyde DF, McCarthy VC, Miller RM, Hornick RB. Specificity of <strong>protection</strong> of man<br />

immunized <strong>against</strong> sporozoite-induced falciparum malaria. Am J Med Sci 1973;<br />

266:398-403.<br />

47. Rieckmann KH. Human immunization with attenuated sporozoites. Bull World<br />

Health Organ 1990; 68 Suppl:13-6.<br />

48. Jeffery GM. Epidemiological significance of repeated <strong>infection</strong>s with<br />

homologous <strong>and</strong> heterologous strains <strong>and</strong> species of Plasmodium. Bull World<br />

Health Organ 1966; 35:873-82.<br />

49. Remarque EJ, Faber BW, Kocken CH, Thomas AW. A diversity-covering approach<br />

to immunization with Plasmodium falciparum apical membrane antigen 1<br />

induces broader allelic recognition <strong>and</strong> growth inhibition responses in rabbits.<br />

Infect Immun 2008; 76:2660-70.


Chapter 8<br />

Induction of malaria in volunteers by<br />

intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

Meta Roestenberg a,1 , Else M. Bijker a,1 , B. Kim Lee Sim b,c , Peter F. Billingsley b , Eric<br />

R. James b , Guido J.H. Bastiaens a , Anne C. Teirlinck a , Dunja Zimmerman a , Karina<br />

Teelen a , Theo Arens a , Pieter Beckers a , Annemieke Jansens a , Anja Scholzen a ,<br />

Adrian J.F. Luty a,* , Krystelle Nganou-Makamdop a , Jorien Wiersma a , André J.A.M.<br />

van der Ven d , Anusha Gunasekera b , Adam Richman b , Sumana Chakravarty b ,<br />

Soundarap<strong>and</strong>ian Velmurugan b , Tao Li b , Anita Manoj b , Abraham G. Eappen b ,<br />

Minglin Li c , Richard E. Stafford b,c , Cornelus C. Hermsen a , Robert W. Sauerwein a,2 ,<br />

Stephen L. Hoffman b,2 1 contributed equally to this study<br />

2 contributed equally to this study<br />

a<br />

Radboud University Nijmegen Medical Center, Department of Medical<br />

Microbiology, Nijmegen, The Netherl<strong>and</strong>s<br />

b<br />

Sanaria Inc., Rockville, USA<br />

c<br />

Protein Potential LLC, Rockville, USA<br />

d<br />

Radboud University Nijmegen Medical Center, Department of General Internal<br />

Medicine, Nijmegen, The Netherl<strong>and</strong>s<br />

*<br />

Current address: Institut de Recherche pour le Développement, UMR 216 Mère<br />

et enfant face aux <strong>infection</strong>s tropicales, Paris, France/Faculté de <strong>Pharma</strong>cie,<br />

Université Paris Descartes, Sorbonne Paris Cité, France.<br />

Am. J. Trop. Med. Hyg. 2012; nov.13


152 Chapter 8<br />

Abstract<br />

Vaccines <strong>and</strong> new drugs are needed to prevent the approximately one million<br />

deaths <strong>and</strong> hundreds of millions of cases caused by Plasmodium falciparum (Pf)<br />

malaria annually. To test these vaccines <strong>and</strong> drugs, volunteers are infected by<br />

the bites of Pf sporozoite (SPZ)-infected mosquitoes under controlled conditions.<br />

Such <strong>infection</strong>s are limited to the few centres with production of PfSPZ-infected<br />

mosquitoes. We assessed the capacity to infect volunteers by intradermal (ID)<br />

injection of aseptic, purified, vialed, cryopreserved PfSPZ (PfSPZ Challenge).<br />

Dutch volunteers received ID injections of PfSPZ that had been cryopreserved<br />

for 27-30 months. In a dose escalation study volunteers (N=6/group) received<br />

2,500, 10,000, or 25,000 PfSPZ. Detection of blood-stage parasites by<br />

microscopy was the primary outcome. Kinetics of parasitemia were<br />

retrospectively assessed by quantitative PCR. Fifteen of eighteen volunteers<br />

(84%) developed Pf parasitemia, 5/6 volunteers at each dose. Infection was safe.<br />

There were no differences between groups in time until parasitemia by<br />

microscopy <strong>and</strong> PCR, parasite kinetics, clinical symptoms <strong>and</strong> signs or laboratory<br />

values.<br />

Aseptic, purified, vialed, cryopreserved PfSPZ manufactured in compliance with<br />

regulatory st<strong>and</strong>ards <strong>and</strong> administered ID successfully infected volunteers.<br />

Following optimization of administration, such PfSPZ could be used to assess<br />

efficacy of vaccines <strong>and</strong> drugs in clinical trial centres worldwide. In addition<br />

PfSPZ Challenge can also be used for immunization strategies <strong>against</strong> malaria.<br />

For example, 100% <strong>protection</strong> <strong>against</strong> Pf <strong>infection</strong> in volunteers immunized by<br />

exposure to PfSPZ-infected mosquitoes while taking chloroquine was recently<br />

demonstrated. PfSPZ Challenge could be used to translate this artificial<br />

immunization protocol into an implementable vaccine.


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

Introduction<br />

Malaria caused by Plasmodium falciparum [Pf] causes approximately one million<br />

deaths <strong>and</strong> 250 million clinical cases annually [1-2]. Implementation of<br />

insecticide impregnated bednets, residual insecticide spraying, <strong>and</strong><br />

combinations of antimalarial drugs, has reduced malaria associated morbidity<br />

<strong>and</strong> mortality in many areas [1]. Questions related to sustainability of this effort,<br />

however, have led to a recent delineation of requirements for new tools [3]. A<br />

safe, long-acting anti-malarial drug <strong>and</strong> a highly effective malaria vaccine would<br />

be powerful tools for control <strong>and</strong> elimination of Pf malaria.<br />

Progress has been facilitated by the capacity to infect volunteers under<br />

controlled conditions in order to test new vaccines <strong>and</strong> drugs. Volunteers are<br />

infected by exposure to laboratory-reared Anopheles spp. mosquitoes<br />

transmitting Pf sporozoites (SPZ) [4], which was first introduced for treatment of<br />

neurosyphilis in the 1920s [5]. The development of drugs such as chloroquine<br />

[6], primaquine [7], <strong>and</strong> atovaquone [8] were facilitated by these controlled<br />

human malaria <strong>infection</strong>s. The ability to culture Pf gametocytes [9-10] enhanced<br />

the capacity to produce infected mosquitoes for controlled human malaria<br />

<strong>infection</strong> (CHMI) studies. Although potentially serious or even lethal, Pf malaria<br />

can be radically cured at the earliest stages of blood <strong>infection</strong> when risks of<br />

complications are virtually absent. CHMIs are restricted to a few specialized<br />

centres that can produce PfSPZ-infected mosquitoes, where more than 1,300<br />

volunteers have been safely infected by the bite of PfSPZ-infected mosquitoes<br />

since 1986, primarily for clinical trials of malaria vaccines [4, 11-15], but also for<br />

trials of drugs [8] <strong>and</strong> diagnostic tests [16], <strong>and</strong> studying human immune<br />

responses to Pf [17].<br />

In addition to the use of CHMIs for testing vaccines <strong>and</strong> drugs, controlled<br />

<strong>infection</strong>s can also be used to immunize <strong>against</strong> malaria. For example,<br />

immunization with radiation-attenuated PfSPZ by bites of mosquitoes protects ><br />

90% of volunteers [18-20]. And recently 100% <strong>protection</strong> <strong>against</strong> CHMI was<br />

achieved by immunization of volunteers with PfSPZ by mosquito bites while<br />

taking chloroquine [21-22].<br />

These highly protective immunization strategies have not been translated into<br />

an implementable vaccine, because they require inoculation of SPZ by mosquito<br />

bites. Inoculation of SPZ by injection would be a more feasible method <strong>and</strong> was<br />

performed through the early 1950s. The SPZ preparations used, however, were<br />

153


154 Chapter 8<br />

contaminated with bacteria <strong>and</strong> rates of <strong>infection</strong> with frozen <strong>and</strong> thawed SPZ<br />

were highly variable [23-26]. A contemporary approach to production of SPZ for<br />

vaccination requires generating aseptic SPZ-infected mosquitoes, purifying SPZ<br />

from mosquito tissues, vialing, preserving, <strong>and</strong> administering the SPZ by needle<br />

<strong>and</strong> syringe. Sanaria has met these requirements to produce an attenuated<br />

(irradiated) PfSPZ vaccine [27-28] <strong>and</strong> infectious (non-irradiated) aseptic,<br />

purified, vialed, cryopreserved PfSPZ (PfSPZ Challenge). Here we report <strong>infection</strong><br />

of volunteers with PfSPZ Challenge administered intradermally (ID) by needle<br />

<strong>and</strong> syringe.<br />

Methods<br />

Study population <strong>and</strong> study design<br />

This open label, Phase 1 clinical trial was performed at Radboud University<br />

Nijmegen Medical Centre, the Netherl<strong>and</strong>s, from October 2010 to July 2011.<br />

Volunteers aged 18-35 years were screened for eligibility by assessing medical<br />

history, physical examination, haematological, biochemical, urine toxicology, <strong>and</strong><br />

pregnancy tests, <strong>and</strong> malaria, HIV, hepatitis B <strong>and</strong> hepatitis C serology. The main<br />

exclusion criteria were pregnancy, residence in a malaria endemic area within<br />

the previous six months, positive Pf serology, symptoms, physical signs or<br />

laboratory test results suggestive of systemic disorders, <strong>and</strong> history of drug or<br />

alcohol abuse interfering with normal social function. All volunteers gave written<br />

informed consent.<br />

Eighteen healthy malaria-naïve volunteers were recruited for this trial. Groups of<br />

six volunteers were injected ID with 2,500, 10,000, or 25,000 PfSPZ Challenge.<br />

The sample size of six per group had a power of 75% to show a difference<br />

between 2 of 6 volunteers infected in the 2,500 PfSPZ group <strong>and</strong> 6/6 volunteers<br />

infected in the 25,000 PfSPZ group. Dose escalation was done at a minimum<br />

interval of 3.5 weeks.<br />

The trial was performed in accordance with Good Clinical Practice <strong>and</strong> an<br />

Investigational New Drug application filed with the U.S. Food <strong>and</strong> Drug<br />

Administration, <strong>and</strong> approved by the Central Committee for Research Involving<br />

Human Subjects of The Netherl<strong>and</strong>s (CCMO NL31858.091.10). Clinicaltrials.gov<br />

identifier: NCT 01086917.


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

Figure 1. Liver-stage Pf parasite expressing PfMSP-1 after six days in culture. PfSPZ<br />

from the lot of PfSPZ Challenge used in this clinical trial were used to infect a human<br />

hepatocyte line in vitro (see Table S1 for methods). Six days later the parasites were<br />

stained with an antibody to PfMSP-1. PfMSP-1 is the major protein on the surface of<br />

merozoites, <strong>and</strong> is required for merozoite invasion of erythrocytes. It is only<br />

expressed in mature liver stage parasites <strong>and</strong> blood stage parasites, <strong>and</strong> is required<br />

for the survival of blood stage parasites. Potency of lots of PfSPZ Challenge is<br />

assessed by expression of PfMSP-1 in vitro (see Table S1). Image captured with a<br />

Zeiss LSM510 META laser scanning confocal microscope.<br />

Study treatment (PfSPZ Challenge)<br />

PfSPZ Challenge contains aseptic, purified, cryopreserved PfSPZ isolated from<br />

salivary gl<strong>and</strong>s of aseptically reared mosquitoes [28]. A. stephensi mosquitoes<br />

were raised under aseptic conditions, then fed on cultured Stage V gametocytes<br />

of the NF54 strain of Pf [38]. Approximately two weeks later, mosquito salivary<br />

gl<strong>and</strong>s containing PfSPZ were dissected, <strong>and</strong> PfSPZ were purified, formulated,<br />

vialed (15,000 PfSPZ per vial) <strong>and</strong> cryopreserved in liquid nitrogen vapour phase<br />

at -140°C to -196°C [28]. PfSPZ Challenge released for clinical use met quality<br />

control specifications including sterility (USP 71 compendial assay), purity (Fig.<br />

S1), <strong>and</strong> potency (Table S1). The latter was assessed by quantification of late<br />

liver stage parasites expressing Pf merozoite surface protein 1 (PfMSP-1) [39] in<br />

cultured human hepatocytes (HC-04 cells) six days after addition of PfSPZ (Figure<br />

1, Table S1). Membrane integrity of PfSPZ was used to assess cell viability (Table<br />

155


156 Chapter 8<br />

S1). The lot of PfSPZ Challenge used in this study had been cryopreserved for 27<br />

(dose of 2,500 PfSP)-30 months (dose of 25,000 PfSPZ) before administration.<br />

Immediately prior to use, a vial of PfSPZ Challenge was thawed <strong>and</strong> diluted with<br />

phosphate buffered saline (PBS) containing human serum albumin (HSA).<br />

Volunteers were injected within 30 minutes of thawing.<br />

Controlled human malaria <strong>infection</strong> (CHMI)<br />

Three groups of six volunteers each were injected ID with PfSPZ Challenge over<br />

the deltoid muscle, one injection in each upper arm. Each injection of 50 μl<br />

contained half the total dose. After injection, volunteers were observed for at<br />

least 60 minutes. Inoculations of volunteers were spaced 60 minutes apart. In<br />

each dose group, two volunteers were inoculated three days prior to the<br />

remaining four volunteers.<br />

Volunteers made at least one daily outpatient clinical visit beginning five days<br />

after inoculation of PfSPZ Challenge. All symptoms <strong>and</strong> signs (solicited <strong>and</strong><br />

unsolicited) were recorded <strong>and</strong> graded by the attending physician as follows:<br />

mild (easily tolerated), moderate (interferes with normal activity), or severe<br />

(prevents normal activity); fever was recorded as grade 1 (>37.5°C – 38.0°C),<br />

grade 2 (>38.0°C – 39.0°C) or grade 3 (>39.0°C). Haematological <strong>and</strong> biochemical<br />

parameters were monitored daily. Because of a previous cardiac related serious<br />

adverse event following CHMI with Pf <strong>infection</strong> [40], markers of cardiac damage<br />

<strong>and</strong> coagulation were assessed. Troponin, lactate dehydrogenase, platelets, <strong>and</strong><br />

d-dimer were assessed daily during the period when blood stage parasitemia<br />

was expected, <strong>and</strong> for three days after initiating curative treatment with<br />

atovaquone/proguanil. If d-dimer or LDH were abnormal, blood samples were<br />

tested for fragmentocytes <strong>and</strong> Von Willebr<strong>and</strong> cleaving protease activity, as<br />

markers for vascular endothelial cell activation [41]. Final follow-up visits were<br />

on days 35 <strong>and</strong> 140 after <strong>infection</strong>.<br />

As soon as parasites were detected by microscopic examination of blood smears,<br />

volunteers were treated with atovaquone/proguanil (1000/400 mg)<br />

administered orally once daily for three days. Complete cure was confirmed in<br />

all volunteers by two consecutive parasite-negative blood-slides after treatment.<br />

Volunteers who did not develop parasitemia by day 21 after challenge were<br />

presumptively treated with the same regimen.


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

Thick Smear qPCR<br />

Volunteer Pre patent Parasite qPCR positive Parasite Parasite<br />

code period (day) density at (day) density at first density by<br />

diagnosis<br />

day positive qPCR at time of<br />

(Pf/μl)<br />

(Pf/μl) diagnosis by<br />

thick smear<br />

(Pf/μl)<br />

Group 1 – 2,500PfSPZ<br />

696-18 12.3 4 9.6 0.08 5<br />

711-08 14.0 16 12 0.16 71<br />

795-06 N/A N/A N/A N/A N/A<br />

935-01 14.0 124 10.6 0.03 89<br />

937-20 12.3 6 10.6 0.12 43<br />

940-14 12.3 6 10.3 0.06 35<br />

Geom. mean 13.0 12 10.59 0.1 35<br />

# of positives 5/6<br />

Group 2 – 10,000 PfSPZ<br />

119-03 12.6 24 9.6 0.68 6<br />

603-11 13.0 8 11 0.17 2<br />

736-04 11.0 6 9.6 0.04 3<br />

783-25 13.3 6 10.6 0.03 15<br />

788-21 14.0 26 11 1.12 6<br />

925-26 N/A N/A N/A N/A N/A<br />

Geom. mean 12.7 11 10.34 0.2 5<br />

# of positives 5/6<br />

Group 3 – 25,000 PfSPZ<br />

647-30 14.0 512 9.3 0.32 759<br />

720-13 12.3 6 10.3 0.32 162<br />

789-15 N/A N/A N/A N/A N/A<br />

806-09 12.3 8 9 0.25 48<br />

909-29 14.3 48 11.3 0.13 102<br />

926-24 12.3 6 10 0.19 68<br />

Geom. mean 13.0 23 9.95 0.2 132<br />

Table 1. Parasitemia data by thick blood smear <strong>and</strong> quantitative polymerase chain<br />

reaction (Q-PCR)<br />

N/A: not applicable; thick-smear negative volunteers were presumptively treated on day<br />

21 after <strong>infection</strong>.<br />

Outcomes<br />

The primary outcome was occurrence of Pf parasitemia detected by microscopic<br />

examination of blood smears. Sampling was done twice daily on days 5 <strong>and</strong> 6<br />

post-inoculation, thrice daily on days 7-11, twice daily on days 12-15, once daily<br />

on days 16-21, <strong>and</strong> for two days after initiation of treatment for positive smears.<br />

To make thick blood smears, 15 μL of EDTA-anti-coagulated blood was spread on<br />

one well of a 3-well glass slide (CEL-LINE Diagnostic Microscope Slides, 30-12A-<br />

157


158 Chapter 8<br />

Figure 2. Parasite density as measured by Q-PCR in the 2,500 (A), 10,000 (B) <strong>and</strong> 25,000<br />

(C) PfSPZ Challenge dose groups. Panels A, B <strong>and</strong> C show geometric mean parasite<br />

density of positive volunteers per group with confidence intervals (N=5 for all groups)<br />

from day of inoculation through last day of positivity after initiation of treatment. Panel<br />

D shows an overlay of geometric mean parasite densities of positive volunteers in each<br />

group.<br />

black-CE24). After drying, wells were stained with Giemsa for 45 minutes, <strong>and</strong><br />

examined at 1000× magnification to assess 0.5 μL of blood. The smear was<br />

scored as positive if two unambiguous parasites were found. Thus, volunteers<br />

could be diagnosed with as few as 4 parasites/μL of blood. Pre-patent period<br />

was defined as the period between inoculation of PfSPZ Challenge <strong>and</strong><br />

appearance of first positive blood smear.<br />

Retrospectively, parasitemias were determined by real-time quantitative PCR<br />

(qPCR), performed on all samples collected after challenge, as previously<br />

described [42]. The sensitivity of qPCR was 20 parasites/mL of blood.<br />

Statistical analysis<br />

Data analysis was performed using SPSS software version 16.0. Q-PCR results<br />

were assessed by ANOVA on log-transformed data.


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

Figure 3. Number of possibly, probably, or definitely related solicited <strong>and</strong> unsolicited<br />

adverse events reported over time in the 2,500 (black ), 10,000 (red ) <strong>and</strong> 25,000 PfSPZ<br />

Challenge dose (green ) groups.<br />

Results<br />

Parasitemia after injection of PfSPZ Challenge<br />

Thirty-six healthy, malaria-naïve volunteers were screened <strong>and</strong> eighteen were<br />

included. All volunteers completed follow-up (Figure S2). After ID injection of<br />

PfSPZ Challenge, 15 of the 18 volunteers developed a positive blood smear for<br />

Pf, five of six volunteers from each group (Table 1). The slide-negative volunteers<br />

in each group were presumptively treated with atovaquone/proguanil at 21 days<br />

post-<strong>infection</strong>.<br />

Blood slides were first positive 11 to 14.3 days after administration of PfSPZ<br />

Challenge. The geometric mean (GM) pre-patent period was similar for all<br />

groups i.e. 13.0, 12.7, <strong>and</strong> 13.0 days for the groups receiving 2,500, 10,000, <strong>and</strong><br />

25,000 PfSPZ Challenge, respectively (ANOVA p=0.92). The GM parasite densities<br />

by microscopy at the time of diagnosis were 12.4, 11.2, <strong>and</strong> 23.4 parasites/μL<br />

blood (ANOVA p=0.69 on log-transformed data) (Table 1).<br />

Quantitative PCRs (qPCRs) were first positive 9⋅0 to 12 days after challenge<br />

(Table 1). Volunteers in the 2,500, 10,000, <strong>and</strong> 25,000 PfSPZ Challenge groups<br />

had similar GM times to first detection of parasites by qPCR of 10.6, 10.3, <strong>and</strong><br />

9.9 days (ANOVA p=0.486) at a GM parasite density of 0.07, 0.2 <strong>and</strong> 0.2<br />

parasites/μL blood (ANOVA p=0.24), respectively. The GM parasite densities by<br />

PCR at the time of thick smear diagnosis were 35, 5, <strong>and</strong> 132 parasites/μL<br />

(ANOVA p=0⋅23). Thick smear <strong>and</strong> qPCR were negative throughout the 21-day<br />

159


160 Chapter 8<br />

Adverse-event<br />

Number of<br />

volunteers<br />

2,500 (n=6) 10,000 (n=6) 25,000 (n=6)<br />

Mean<br />

Duration<br />

±SD (days)<br />

Number of<br />

volunteers<br />

Mean<br />

Duration<br />

±SD (days)<br />

Number of<br />

volunteers<br />

Mean<br />

Duration<br />

±SD (days)<br />

Abdominal pain 1 2.9 1 0.04 2 0.3±0.1<br />

Arthralgia 0 N/A 0 N/A 0 N/A<br />

Chest pain 1 0.04 0 N/A 0 N/A<br />

Chills 1 2.0 2 0.3±0.2 2 0.9±0.6<br />

Diarrhea 0 N/A 0 N/A 1 0.8<br />

Fatigue 5 2.9±3.3 3 2.5±1.7 5 3.0±3.9<br />

Fever 3 1.6±1.5 2 1.8±0.6 4 0.8±0.4<br />

Headache 6 1.1±1.1 6 1.5±1.6 6 1.4±2.6<br />

Malaise 2 2.2±2.4 5 1.8±1.4 1 0.7<br />

Myalgia 2 3.7±3.2 2 1.3±0.5 2 0.8±0.1<br />

Nausea 3 1.7±1.3 5 0.9±0.9 3 1.0±0.9<br />

Vomiting 0 N/A 2 0.01±0.0 0 N/A<br />

Any 6 2.0±1.4 6 1.1±0.8 6 1.1±1.0<br />

Grade 3 adverse event<br />

Fatigue 0 N/A 0 N/A 1 2.2<br />

Fever 0 N/A 1 1.2 0 N/A<br />

Headache 2 3.0±0.4 0 N/A 0 N/A<br />

Malaise 1 4.8 0 N/A 1 0.1<br />

Vomiting 0 N/A 2 0.01±0.0 0 N/A<br />

Any 2 3.9±0.2 3 0.6±0.0 2 1.2±0.0<br />

Table 2. Numbers of volunteers reporting solicited adverse events possibly, probably, or<br />

definitely related to administration of PfSPZ Challenge, with mean duration of events<br />

N/A: not applicable<br />

follow-up for the three slide-negative volunteers. Parasite growth was cyclical,<br />

<strong>and</strong> was similar in all dose groups (Figure 2).<br />

Safety.<br />

All volunteers, including the three volunteers who did not develop parasitemia,<br />

reported solicited adverse events (AEs) considered possibly, probably, or<br />

definitely related to the trial procedures (clinical malaria) (Table 2). Headache<br />

was the most frequently reported AE, <strong>and</strong> occurred in all volunteers including<br />

the three who did not develop parasitemia. There were no significant<br />

differences among the groups in solicited AEs, which were most frequently<br />

reported between days 12 <strong>and</strong> 18 post-injection. The total number of solicited<br />

<strong>and</strong> unsolicited AEs reported over time is shown in Figure 3.<br />

Routine daily laboratory tests showed no clinically significant abnormalities<br />

before initiation of anti-malarial treatment. Three or four days after receiving<br />

the first dose of atovaquone/proguanil, four volunteers had thrombocyte levels<br />

in the range 78-95 × 10 9 /L, which was below the lower limit of normal (120 ×


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

10 9 /L). In thirteen volunteers, D-dimers were above 500 ng/mL, the upper limit<br />

of normal (ULN), at one or two days after initiation of anti-malarial treatment<br />

(range of peaks: 540 to 10,200 ng/mL). In all volunteers, D-dimer levels<br />

normalized without complications. One volunteer had abnormal liver function<br />

tests at day 2 post atovaquone/proguanil initiation. Maximum values were 526<br />

units/L ASAT (ULN 40 units/L), 745 units/L ALAT (ULN 45 units/L), 777 units/L<br />

LDH (ULN 450 units/L), <strong>and</strong> 74 units/L γGT (ULN 50 units/L). Bilirubin <strong>and</strong> alkaline<br />

phosphatase were normal. Abnormal values had returned to baseline levels at<br />

day 100 after <strong>infection</strong>.<br />

One serious adverse event (SAE) occurred in a volunteer who reported chest<br />

pain one day after the first dose of atovaquone/proguanil. Based on medical<br />

history, the chest pain was initially considered possibly consistent with angina<br />

pectoris. Pain resolved within one hour without treatment. The volunteer was<br />

admitted to the cardiac care unit for monitoring for 6.5 hours. The first<br />

electrocardiogram (ECG) had a negative T-wave in V2, which was absent at time<br />

of study initiation. All subsequent ECGs, beginning 2.5 hours after the first ECG,<br />

were comparable to baseline, with a negative T in V1 only. Troponin T levels<br />

were normal at the time of chest pain, 6 <strong>and</strong> 17 hours later, daily for the next<br />

three days <strong>and</strong> at trial days 28 <strong>and</strong> 35. As per protocol, the trial was put on hold,<br />

<strong>and</strong> the event was reported to the Safety Monitoring Committee (SMC) <strong>and</strong><br />

regulatory authorities. The SMC concurred with the PI’s attribution of the chest<br />

pain as “possibly related” to participation in the trial. The SMC concluded that<br />

while the cause of chest pain was not clear, the clinical data suggested that the<br />

SAE was not a serious cardiac event, <strong>and</strong> recommended resumption of the trial<br />

within three days of the event. The regulatory authorities concurred.<br />

Discussion<br />

We report for the first time that healthy, malaria-naïve volunteers can be<br />

infected with P. falciparum malaria by injection of aseptic, purified,<br />

cryopreserved PfSPZ manufactured in compliance with regulatory st<strong>and</strong>ards.<br />

Five of six volunteers became infected when 2500, 10,000 or 25,000 PfSPZ were<br />

inoculated ID. AEs were comparable with those in mosquito bite challenge trials<br />

[29-31].<br />

In the 1950s Jeffery <strong>and</strong> colleagues were able to infected volunteers with<br />

cryopreserved PfSPZ [20]. They cryopreserved PfSPZ-infected salivary gl<strong>and</strong>s <strong>and</strong><br />

161


162 Chapter 8<br />

inoculated the thawed salivary gl<strong>and</strong>s from 2.5 to 15 mosquitoes intravenously<br />

<strong>and</strong> achieved Pf parasitemia in 13 of 14 volunteers. The only volunteer who did<br />

not develop Pf parasitemia received salivary gl<strong>and</strong>s from 2.5 PfSPZ-infected<br />

mosquitoes, the lowest dose. Because the preparations were likely highly<br />

contaminated with bacteria they administered penicillin concomitantly with<br />

PfSPZ-infected salivary gl<strong>and</strong> preparations.<br />

Since 1986 CHMIs have been performed by exposing volunteers to bites of<br />

laboratory-reared mosquitoes infected by feeding on Pf gametocyte-infected<br />

erythrocytes grown in culture [11]. When volunteers are challenged by bites of<br />

five PfSPZ-infected mosquitoes, essentially all develop Pf parasitemia [4, 11, 29,<br />

31]. When numbers are reduced to one or two mosquitoes success rates drop to<br />

50% [30, 32]. Recently, 5 of 6 volunteers became infected when bitten by one<br />

aseptic, PfSPZ-infected mosquito produced in compliance with regulatory<br />

st<strong>and</strong>ards [33]. ID inoculation of 2,500 cryopreserved PfSPZ Challenge in the<br />

current study was at least as effective as the bites of 1-2 infected mosquitoes in<br />

infecting volunteers.<br />

The capacity to infect volunteers with PfSPZ Challenge is a function of the<br />

infectiousness of the cryopreserved PfSPZ <strong>and</strong> the efficiency of administration.<br />

We use in vitro assays of potency <strong>and</strong> viability to estimate infectiousness of fresh<br />

vs. cryopreserved SPZ. These in vitro assays predicted a maximum difference of<br />

25-30% (Table S1). Rodent model in vivo data, however, suggested that the in<br />

vitro assays underestimate the reduction in infectivity associated with SPZ<br />

cryopreservation (Table S2).<br />

Increasing the dose of PfSPZ Challenge from 2,500 PfSPZ to 25,000 PfSPZ<br />

administered ID neither increased the percentage of infected volunteers nor<br />

reduced time until blood stage parasites were detectable. Parasite density at<br />

diagnosis was lower in the 10,000 PfSPZ group compared to both the lower <strong>and</strong><br />

the higher dose groups. This is probably a reflection of natural variability<br />

between the volunteers. Apparently, increasing the dose did not result in higher<br />

numbers of PfSPZ developing in hepatocytes, <strong>and</strong> invading erythrocytes. A<br />

possible explanation for this lack of dose response may be trapping of PfSPZ at<br />

the inoculation site. Mosquitoes deposit SPZ in the dermis <strong>and</strong> subcutaneous<br />

tissues in much smaller volumes (< 0.5 μL) than the inocula used in this study (50<br />

μL). This interpretation is consistent with recent studies that showed that<br />

approximately 90% of P. yoelii sporozoites did not reach the liver when<br />

administered ID [34]. Additionally, mosquitoes also inoculate SPZ directly into


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

the circulation [35]. The fact that Jeffery et al. achieved <strong>infection</strong> in 13 of 14<br />

volunteers by intravenous injection of PfSPZ-infected salivary gl<strong>and</strong>s [25], <strong>and</strong><br />

the only negative case was at the lowest does of salivary gl<strong>and</strong>s, supports this<br />

interpretation. To closer mimic the SPZ delivery of mosquitoes, administration of<br />

PfSPZ Challenge will be optimized by modifying route of administration (e.g. ID,<br />

subcutaneous, intramuscular, intravenous), inoculation volume, numbers of<br />

inoculations, <strong>and</strong> sites of injection. Our sample sizes of 6 per group, in this firstin-humans<br />

study, were not powered to be able show a modest difference<br />

between groups. Thus, another explanation for the lack of difference in<br />

infectivity or prepatent period between as the dose was increased 10-fold from<br />

2,500 to 25,000 PfSPZ is that that sample sizes were too small to detect small<br />

differences between groups. However, they should have been adequate to<br />

detect a 10-fold difference, if the PfSPZ actually reached the bloodstream. Since<br />

there was no difference as we increased the dose, it is possible that 2,500 PfSPZ<br />

was above the threshold necessary to achieve <strong>infection</strong>, <strong>and</strong> even lower doses<br />

might be equally successful.<br />

Once administration of PfSPZ Challenge is optimized, the global capacity to<br />

conduct CHMIs can be further exp<strong>and</strong>ed including sites in malaria endemic<br />

areas. This will be critical for meeting the dem<strong>and</strong> to assess the increasing<br />

numbers of malaria vaccine c<strong>and</strong>idates <strong>and</strong> anti-malarial drugs in development<br />

[36-37]. Comparative analysis of CHMI by mosquito bite has shown that<br />

variability of parasites may lead to significant variation in the primary outcome<br />

variables between sites. By using needle administration of defined quantities of<br />

PfSPZ Challenge from the same lot, variation in infectivity in CHMI will be<br />

reduced, allowing comparisons of parallel <strong>and</strong> sequential clinical trials at<br />

multiple sites, including in malaria endemic areas.<br />

In addition to its use in CHMI, needle <strong>and</strong> syringe administration of<br />

cryopreserved SPZ is critical for development of whole SPZ vaccines. PfSPZ<br />

protective efficacy in humans was originally established by controlled exposure<br />

of volunteers to bites of irradiated PfSPZ-infected mosquitoes [18-20]. The<br />

potential impact of a radiation-attenuated PfSPZ vaccine was reappraised <strong>and</strong><br />

significant progress has been made in manufacturing <strong>and</strong> clinically testing such a<br />

vaccine [27-28]. The aseptic, purified, cryopreserved PfSPZ Vaccine is<br />

manufactured identically to PfSPZ Challenge, except that the parasites in the<br />

vaccine are attenuated by irradiation <strong>and</strong> cannot fully develop in the liver. In the<br />

first clinical trial the PfSPZ Vaccine was administered ID or subcutaneously. It<br />

163


164 Chapter 8<br />

was safe <strong>and</strong> well tolerated, but immunogenicity <strong>and</strong> protective efficacy were<br />

not nearly to the levels found after mosquito bite immunization [28]. The data<br />

reported herein on PfSPZ Challenge administered ID demonstrated that the<br />

manufacturing process produces viable <strong>and</strong> potent SPZ. These data, along with<br />

non-human primate immunization studies with PfSPZ Vaccine, informed the<br />

design of the next clinical trial of the PfSPZ Vaccine, which is being administered<br />

by intravenous injection [28].<br />

Another approach to whole SPZ vaccination was recently established [21-22].<br />

Healthy malaria-naïve volunteers were protected for 28 months <strong>against</strong> CHMI<br />

after being immunized by exposure to bites of PfSPZ-infected mosquitoes while<br />

taking the antimalarial drug chloroquine. This <strong>protection</strong> was induced using ><br />

20-fold fewer PfSPZ-infected mosquitoes [36 to 45] than needed for <strong>protection</strong><br />

with radiation attenuated PfSPZ (> 1000) [21-22]. PfSPZ Challenge can replace<br />

mosquito bites, allowing for rapid translation of this experimental research<br />

finding into a potential vaccine. We are planning the first clinical trials of PfSPZ<br />

Challenge administered to volunteers taking chloroquine for 2012.<br />

In summary, we have established that aseptic, purified, vialed, cryopreserved<br />

PfSPZ (PfSPZ Challenge) are infectious to humans for at least 2.5 years after<br />

cryopreservation. These data provide the rationale <strong>and</strong> foundation for a clinical<br />

trials program aimed at significantly increasing the scale of CHMI in clinical trials<br />

of malaria vaccines <strong>and</strong> new drugs, <strong>and</strong> producing, testing, <strong>and</strong> licensing a highly<br />

effective, long-acting PfSPZ malaria vaccine.<br />

Acknowledgements<br />

We thank the trial volunteers, the staff from the Clinical Research Centre<br />

Nijmegen <strong>and</strong> the staff from the RUNMC <strong>Pharma</strong>cy who made this study<br />

possible; Wendy Arts, Nanny Huiberts, Chantal Siebes, Marlou Kooreman, Paul<br />

Daemen <strong>and</strong> Ella Driessen for reading many thick smears; <strong>and</strong> Dr. Gheorghe Pop<br />

for his cardiac monitoring of the trial volunteers. We thank Bas van Haren for his<br />

support with the ADAMTS13 measurements <strong>and</strong> Jody van den Ouwel<strong>and</strong> for the<br />

performance of the Troponin T measurements. We thank the members of the<br />

Safety Monitoring Committee, Dr. Barney Graham, Dr. David Diemert <strong>and</strong> Dr.<br />

Alex<strong>and</strong>er Rennings for their participation <strong>and</strong> for their guidance <strong>and</strong> safety<br />

recommendations throughout the trial. We thank the entire Sanaria<br />

Manufacturing Team including Gametocyte Production: Yonas Abebe, Asha Patil,<br />

Yeab Getachew, Mark Loyvesky, Bingbing Deng; Mosquito Production: Steve


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

Matheny, Yingda Wen, Keith Nelson, James Overby, Virak Pich, Thomas Mitchell;<br />

Dissection: Richard Fan; Purification: Lixin Gao, Ray Xu; Formulation <strong>and</strong><br />

Cryopreservation: Adam Ruben, Aderonke Awe, Quality Assurance <strong>and</strong> Quality<br />

Control: Irina Belyakova, Mary King; Assays: Chinnamma Chakiath; Rana<br />

Chattopadhyay, Charles Anderson, Solomon Conteh, Gwynne Roth; Regulatory<br />

<strong>and</strong> Clinical Affairs: Tooba Murshedkar. We are grateful for Sanaria’s Operations<br />

<strong>and</strong> Legal teams, especially Robert Thompson, Alex<strong>and</strong>er Hoffman, <strong>and</strong> David<br />

Dolberg. We thank the entire Protein Potential Team including, Adriana<br />

Ahumada, Maria Socorro Orozco, Bing Jiang, Emily Chen. We also thank MSource<br />

(Belgium, now CromSource, Italy) for clinical monitoring.<br />

Funding<br />

Manufacturing of PfSPZ Challenge <strong>and</strong> this clinical trial were funded by<br />

<strong>TI</strong><strong>Pharma</strong>, grant number T4-102. ACT is funded by the European Malaria Vaccine<br />

Development Association, KNM was funded by a Mozaiek grant from the<br />

Netherl<strong>and</strong>s Organisation for Scientific Research. The development <strong>and</strong><br />

manufacturing of PfSPZ Challenge was also supported by Small Business<br />

Innovation Research (SBIR) grants R44AI058375-03,04,05,05S1 from NIAID/NIH<br />

<strong>and</strong> through Agreement 07984 from the PATH Malaria Vaccine Initiative.<br />

165


166 Chapter 8<br />

References<br />

1. World Malaria Report, 2011. (http://www.who.int/malaria)<br />

2. Murray CJ, Rosenfeld LC, Lim SS, et al. Global malaria mortality between 1980<br />

<strong>and</strong> 2010: a systematic analysis. Lancet 2012; 379:413-431.<br />

3. Najera JA, Gonzalez-Silva M, Alonso PL. Some lessons for the future from the<br />

Global Malaria Eradication Programme (1955-1969). PLoS Med 2011;<br />

8:e1000412.<br />

4. Sauerwein RW, Roestenberg M, Moorthy VS. <strong>Experimental</strong> human challenge<br />

<strong>infection</strong>s can accelerate clinical malaria vaccine development. Nat Rev<br />

Immunol 2011; 11:57-64.<br />

5. Covell G, Nicol WD. Clinical, chemotherapeutic <strong>and</strong> immunological studies on<br />

induced malaria. Br Med Bull 1951; 8:51-5.<br />

6. Coggeshall LT & Craige B Old <strong>and</strong> new plasmodicides. Malariology: a<br />

comprehensive survey of all aspects of this group of diseases from a global<br />

st<strong>and</strong>point, ed Boyd MF (W. B. Saunders, Philadelphia), 1949; Vol II:1071-1114.<br />

7. Alving AS, Arnold J, Hockwald RS, et al. Potentiation of the curative action of<br />

primaquine in vivax malaria by quinine <strong>and</strong> chloroquine. J. Lab. Clin. Med. 1955;<br />

46:301-306.<br />

8. Shapiro TA, Ranasinha CD, Kumar N, & Barditch-Crovo P Prophylactic activity of<br />

atovaquone <strong>against</strong> Plasmodium falciparum in humans. Am. J. Trop. Med. Hyg.<br />

1999; 60:831-836.<br />

9. Ifediba T, V<strong>and</strong>erberg JP. Complete in vitro maturation of Plasmodium<br />

falciparum gametocytes. Nature 1981; 294:364-6.<br />

10. Campbell CC, Collins WE, Nguyen Dinh P, Barber A, Broderson JR. Plasmodium<br />

falciparum gametocytes from culture in vitro develop to sporozoites that are<br />

infectious to primates. Science 1982; 217:1048-50.<br />

11. Chulay JD, Schneider I, Cosgriff TM, et al. Malaria transmitted to humans by<br />

mosquitoes infected from cultured Plasmodium falciparum. Am J Trop Med Hyg<br />

1986;35:66-8.<br />

12. Gordon DM, McGovern TW, Krzych U, et al. Safety, immunogenicity, <strong>and</strong><br />

efficacy of a recombinantly produced Plasmodium falciparum circumsporozoite<br />

protein-hepatitis B surface antigen subunit vaccine. J Infect Dis 1995; 171:1576-<br />

85.<br />

13. Stoute JA, Slaoui M, Heppner DG, et al. A preliminary evaluation of a<br />

recombinant circumsporozoite protein vaccine <strong>against</strong> Plasmodium falciparum<br />

malaria. N Engl J Med 1997; 336:86-91.


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

14. Thompson FM, Porter DW, Okitsu SL, et al. Evidence of blood stage efficacy with<br />

a virosomal malaria vaccine in a phase IIa clinical trial. PLoS One 2008; 3:e1493.<br />

15. Kester KE, Cummings JF, Ofori-Anyinam O, et al. R<strong>and</strong>omized, Double-Blind,<br />

Phase 2a Trial of falciparum Malaria Vaccines RTS,S/AS01B <strong>and</strong> RTS,S/AS02A in<br />

Malaria-Naive Adults: Safety, Efficacy, <strong>and</strong> Immunologic Associates of<br />

Protection. J Infect Dis 2009; 200:337-46.<br />

16. Beadle C, Long GW, Weiss WR, et al. Diagnosis of malaria by detection of<br />

Plasmodium falciparum HRP-2 antigen with a rapid dipstick antigen-capture<br />

assay. Lancet 1994; 343:564-568.<br />

17. McCall MB, Netea MG, Hermsen CC, et al. Plasmodium falciparum <strong>infection</strong><br />

causes proinflammatory priming of human TLR responses. J Immunol 2007;<br />

179:162-71.<br />

18. Clyde DF, McCarthy VC, Miller RM, Woodward WE. Immunization of man<br />

<strong>against</strong> falciparum <strong>and</strong> vivax malaria by use of attenuated sporozoites. Am J<br />

Trop Med Hyg 1975; 24:397-401.<br />

19. Rieckmann KH, Carson PE, Beaudoin RL, Cassells JS, Sell KW. Sporozoite induced<br />

immunity in man <strong>against</strong> an Ethiopian strain of Plasmodium falciparum. Trans R<br />

Soc Trop Med Hyg 1974; 68:258-9.<br />

20. Hoffman SL, Goh LM, Luke TC, et al. Protection of humans <strong>against</strong> malaria by<br />

immunization with radiation-attenuated Plasmodium falciparum sporozoites. J<br />

Infect Dis 2002; 185:1155-64.<br />

21. Roestenberg M, McCall M, Hopman J, et al. Protection <strong>against</strong> a malaria<br />

challenge by sporozoite inoculation. N Engl J Med 2009; 361:468-77.<br />

22. Roestenberg M, Teirlinck AC, McCall MB, et al. Long-term <strong>protection</strong> <strong>against</strong><br />

malaria after experimental sporozoite inoculation: an open-label follow-up<br />

study. Lancet 2011; 377:1770-6.<br />

23. Maynes B. The injection of mosquito sporozoites in malaria therapy. Pub Health<br />

Rep 1933;48:909-13.<br />

24. Mayne B, Young M. The technique of induced malaria as used in the South<br />

Carolina State Hospital. Journal of Venereal Disease Information 1941; 22:271-<br />

6.<br />

25. Jeffery GM, Rendtorff RC. Preservation of viable human malaria sporozoites by<br />

low-temperature freezing. Exp Parasitology 1955; 4:445-54.<br />

26. Glynn JR, Bradley DJ. Inoculum size, incubation period <strong>and</strong> severity of malaria.<br />

Analysis of data from malaria therapy records. Parasitology 1995; 110:7-19.<br />

27. Hoffman SL, Billingsley PF, James E, et al. Development of a metabolically active,<br />

non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria.<br />

Hum Vaccin 2010; 6:97-106.<br />

167


168 Chapter 8<br />

28. Epstein JE, Tewari K, Lyke KE, et al. Live attenuated malaria vaccine designed to<br />

protect through hepatic CD8 T cell immunity. Science 2011; 334:475-480.<br />

29. Church LW, Le TP, Bryan JP, et al. Clinical manifestations of Plasmodium<br />

falciparum malaria experimentally induced by mosquito challenge. J Infect Dis<br />

1997; 175:915-20.<br />

30. Verhage DF, Telgt DS, Bousema JT, et al. Clinical outcome of experimental<br />

human malaria induced by Plasmodium falciparum-infected mosquitoes. Neth J<br />

Med 2005; 63:52-8.<br />

31. Epstein JE, Rao S, Williams F, et al. Safety <strong>and</strong> clinical outcome of experimental<br />

challenge of human volunteers with Plasmodium falciparum-infected<br />

mosquitoes: an update. J Infect Dis 2007; 196:145-54.<br />

32. Rickman LS, Jones TR, Long GW, et al. Plasmodium falciparum-infected<br />

Anopheles stephensi inconsistently transmit malaria to humans. Am J Trop Med<br />

Hyg 1990; 43:441-5.<br />

33. Lyke KE, Laurens M, Adams M, et al. Plasmodium falciparum malaria challenge<br />

by the bite of aseptic Anopheles stephensi mosquitoes: results of a r<strong>and</strong>omized<br />

infectivity trial. PLoS One 2010; 5:e13490.<br />

34. Inoue M & Culleton RL The intradermal route for inoculation of sporozoites of<br />

rodent malaria parasites for immunological studies. Parasite Immunol. 2011;<br />

33:137-142.<br />

35. Fairley NH. Sidelights on malaria in man obtained by subinoculation<br />

experiments. Trans R Soc Trop Med Hyg 1947; 40:621-76.<br />

36. Alonso PL, Djimde A, Kremsner P, et al. A research agenda for malaria<br />

eradication: drugs. PLoS Med 2011;8:e1000402.<br />

37. WHO (2011) Malaria Vaccine Rainbow Tables.<br />

http://www.who.int/vaccine_research /links/Rainbow/en/index.html.<br />

38. Ponnudurai T, Lensen AHW, Van Gemert GJ, Bensink MPE, Bolmer M,<br />

Meuwissen JH. Infectivity of cultured Plasmodium falciparum gametocytes to<br />

mosquitoes. Parasitol 1989; 98:165-73.<br />

39. Holder AA. The carboxy-terminus of merozoite surface protein 1: structure,<br />

specific antibodies <strong>and</strong> immunity to malaria. Parasitology 2009; 136:1445-56.<br />

40. Nieman AE, de Mast Q, Roestenberg M, et al. Cardiac complication after<br />

experimental human malaria <strong>infection</strong>: a case report. Malar J 2009; 8:277.<br />

41. de Mast Q, Groot E, Lenting PJ, et al. Thrombocytopenia <strong>and</strong> release of<br />

activated von Willebr<strong>and</strong> Factor during early Plasmodium falciparum malaria. J<br />

Infect Dis 2007; 196:622-8.<br />

42. Hermsen CC, Telgt DS, Linders EH, et al. Detection of Plasmodium falciparum<br />

malaria parasites in vivo by real-time quantitative PCR. Mol. Biochem.<br />

Parasitol.2001;118:247-251.


Induction of malaria in volunteers by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites<br />

Supplementary Data<br />

Time point Potency<br />

(# of parasites expressing<br />

PfMSP-1/well)<br />

% Viability<br />

(sporozoite membrane<br />

integrity assay)<br />

Fresh 27 ± 4.6 ND*<br />

Release 20 ± 1.7 83.3% ± 6.5%<br />

6 Month 18 ± 2.1 86.6% ± 1.9%<br />

9 Month 20 ± 2.1 83.7% ± 8.4%<br />

12 Month 21 ± 1.5 84.8% ± 3.0%<br />

18 Month 20 ± 0.6 83.7% ± 4.2%<br />

24 Month 18 ± 1.0 86.0% ± 1.5%<br />

Pre-1st clinical dose (26<br />

Month)<br />

17 ± 0.6 79.4% ± 6.5%<br />

Post-last clinical dose (30<br />

Month)<br />

16 ± 2.6 87.4% ± 1.9%<br />

Table S1. Results of potency <strong>and</strong> sporozoite membrane integrity assays on<br />

the lot of PfSPZ<br />

* Not done<br />

Date Status of PySPZ Viability<br />

(SMIA)<br />

Number of PySPZ<br />

Inoculated (IV)<br />

ID50 (Number of<br />

PySPZ)<br />

Oct 2009 fresh 96.3% 24-12-6-3 8.9<br />

Dec 2009 cryopreserved 72.7% 200-100-50-25 33.1<br />

Dec 2009 cryopreserved 68.3% 200-100-50-25 62.1<br />

Jan 2010 cryopreserved 67.7% 400-200-100-50-25 103.8<br />

Feb 2010 cryopreserved 67.1% 400-200-100-50-25 55.2<br />

Feb 2010 cryopreserved 71.6% 400-200-100-50-25 107<br />

Feb 2010 cryopreserved 73.9% 400-200-100-50-25 34.5<br />

Mean cryopreserved 70.2% 66.0<br />

Difference between fresh<br />

<strong>and</strong> cryopreserved PySPZ<br />

26.1% 7.4-fold<br />

Table S2. Effect of cryopreservation on sporozoite membrane integrity <strong>and</strong> infectivity<br />

in mice inoculated intravenously with the same lot of P. yoelii sporozoites. Infectivity<br />

was the number of sporozoites required to infect 50% of BALB/c mice.<br />

169


170 Chapter 8<br />

Before Purification (14 x 10 6 PfSPZ/ml) After Purification (1.6 x 10 6 PfSPZ/ml)<br />

Figure S1. PfSPZ before <strong>and</strong> after purification. Photomicrograph (200 x) of the PfSPZ in<br />

the lot of PfSPZ Challenge used in the clinical trial prior to purification <strong>and</strong> after<br />

purification. The purification process reduced the amount of salivary gl<strong>and</strong> material in<br />

the PfSPZ samples by greater than 99.9%.<br />

Figure S2: Clinical trial profile


Section 3<br />

Whole parasite inoculation


Chapter 9<br />

Protection <strong>against</strong> a Malaria Challenge by<br />

Sporozoite Inoculation<br />

Meta Roestenberg 1 *, Matthew McCall 1 *, Joost Hopman 1 , Jorien Wiersma 1 ,<br />

Adrian J.F. Luty 1 , Geert Jan van Gemert 1 , Marga van de Vegte-Bolmer 1 , Ben van<br />

Schaijk 1 , Karina Teelen 1 , Theo Arens 1 , Lopke Spaarman 1 , Quirijn de Mast 2 , Will<br />

Roeffen 1 , Georges Snounou 3 , Laurent Rénia 4 , Andre van der Ven 2 , Cornelus C.<br />

Hermsen 1 , Robert Sauerwein 1<br />

*These authors contributed equally<br />

1<br />

Department of Medical Microbiology Radboud University Nijmegen Medical<br />

Center, Nijmegen, the Netherl<strong>and</strong>s<br />

2<br />

Department of General Internal Medicine, Radboud University Nijmegen<br />

Medical Center, Nijmegen, the Netherl<strong>and</strong>s<br />

3<br />

Department of Parasitology, INSERM Unité 511, Hôpital Pitié– Salpêtrière, <strong>and</strong><br />

the Université Pierre et Marie Curie, Paris, France<br />

4<br />

Laboratory of Malaria Immunobiology, Singapore Immunology Network,<br />

Agency for Technology <strong>and</strong> Research, Biopolis, Singapore [L.R.).<br />

N Engl J Med 2009; 361:468-77.


174 Chapter 9<br />

Abstract<br />

An effective vaccine for malaria is urgently needed. Naturally acquired immunity<br />

to malaria develops slowly, <strong>and</strong> induction of <strong>protection</strong> in humans can be<br />

achieved artificially by the inoculation of radiation-attenuated sporozoites by<br />

means of more than 1000 infective mosquito bites.<br />

We exposed 15 healthy volunteers — with 10 assigned to a vaccine group <strong>and</strong> 5<br />

assigned to a control group — to bites of mosquitoes once a month for 3 months<br />

while they were receiving a prophylactic regimen of chloroquine. The vaccine<br />

group was exposed to mosquitoes that were infected with Plasmodium<br />

falciparum, <strong>and</strong> the control group was exposed to mosquitoes that were not<br />

infected with the malaria parasite. One month after the discontinuation of<br />

chloroquine, <strong>protection</strong> was assessed by homologous challenge with five<br />

mosquitoes infected with P. falciparum. We assessed humoral <strong>and</strong> cellular<br />

responses before vaccination <strong>and</strong> before the challenge to investigate correlates<br />

of <strong>protection</strong>.<br />

All 10 subjects in the vaccine group were protected <strong>against</strong> a malaria challenge<br />

with the infected mosquitoes. In contrast, patent parasitemia (i.e., parasites<br />

found in the blood on microscopic examination) developed in all five control<br />

subjects. Adverse events were mainly reported by vaccinees after the first<br />

immunization <strong>and</strong> by control subjects after the challenge; no serious adverse<br />

events occurred. In this model, we identified the induction of parasite-specific<br />

pluripotent effector memory T cells producing interferon-γ tumour necrosis<br />

factor α, <strong>and</strong> interleukin-2 as a promising immunologic marker of <strong>protection</strong>.<br />

In conclusion, <strong>protection</strong> <strong>against</strong> a homologous malaria challenge can be<br />

induced by the inoculation of intact sporozoites.


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 175<br />

Introduction<br />

Malaria is responsible for a significant burden of morbidity <strong>and</strong> mortality in the<br />

developing world <strong>and</strong> an effective vaccine <strong>against</strong> this disease is urgently<br />

required [1]. Despite decades of research a licensed vaccine is still not available,<br />

largely because immunity to Plasmodium falciparum malaria is considered<br />

difficult to acquire, whether through natural exposure or artificially through<br />

vaccination. A further critical factor is our incomplete underst<strong>and</strong>ing of precisely<br />

what constitutes protective anti-malarial immunity in humans.<br />

The possibility of vaccinating humans <strong>against</strong> P. falciparum malaria was raised<br />

originally by the success of the radiation-attenuated sporozoite model<br />

developed several decades ago [2,3]. Irradiation of infectious mosquitoes<br />

disrupts the gene expression of sporozoites, which remain capable of<br />

hepatocyte invasion but no longer of complete liver-stage maturation or<br />

progression to the pathogenic blood-stage [4]. Infection of human volunteers<br />

with irradiated sporozoites thus exposes them to liver-stage antigens <strong>and</strong><br />

generates pre-erythrocytic immunity. However, the requirement of a minimum<br />

of one thous<strong>and</strong> bites by irradiated mosquitoes, over five or more immunization<br />

sessions, in order to successfully induce sterile immunity in humans [5],<br />

precludes this method for routine immunization.<br />

Research turned thus to developing a subunit vaccine based on antigens<br />

expressed by pre-erythrocytic, intra-erythrocytic or sexual stages of the parasite.<br />

Unfortunately, results of many such subunit vaccines in humans have been<br />

disappointing <strong>and</strong> to date only one c<strong>and</strong>idate, based on the circum-sporozoite<br />

protein (CSP) <strong>and</strong> known as RTS,S, has progressed to phase III field trials. The<br />

<strong>protection</strong> induced by this vaccine is encouraging but the ultimate success of<br />

this approach remains to be determined [6-9].<br />

It has been shown that, in rodent models, sterile <strong>protection</strong> <strong>against</strong> malaria can<br />

also be achieved by inoculation of intact sporozoites whilst treating the animals<br />

concomitantly with chloroquine (CQ) [10], a drug that kills asexual blood-stage<br />

but not pre-erythrocytic parasites [11], with an efficacy significantly higher than<br />

that of the radiation-attenuated sporozoite model [12]. We therefore designed a<br />

proof-of-concept study in malaria-naive volunteers to investigate whether<br />

<strong>protection</strong> can be equally effectively induced by this approach in humans <strong>and</strong> to<br />

explore the immune responses elicited.


176 Chapter 9<br />

Figure 1. Study design <strong>and</strong> enrolment<br />

Immunological assessment was performed 1 day before the first immunization<br />

(day I-1) <strong>and</strong> 1 day before the challenge <strong>infection</strong> (day C-1).A final challenge<br />

with infectious mosquito bites was performed 28 days after the<br />

discontinuation of chloroquine prophylaxis.<br />

Methods<br />

Volunteers<br />

Fifteen healthy volunteers were recruited, aged 18-45 years old, without history<br />

of malaria or of residence in a malaria endemic area in the six months prior to<br />

the trial onset. Only one volunteer had ever been in an endemic area, several<br />

years earlier. All volunteers underwent routine physical examination,<br />

haematology <strong>and</strong> biochemistry screening at the Clinical Research Centre<br />

Nijmegen. Serology for HIV, hepatitis B <strong>and</strong> C <strong>and</strong> asexual P. falciparum parasites<br />

was negative in all volunteers. All volunteers gave written informed consent<br />

prior to inclusion. The trial was approved by the Institutional Review Board of


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 177<br />

the Radboud University Nijmegen Medical Centre (CMO 2006/207).<br />

Clinicaltrials.gov identifier: NCT00442377.<br />

Protocol<br />

Volunteers were r<strong>and</strong>omized double blind into two groups (Figure 1), 10 vera<br />

<strong>and</strong> 5 controls. Chloroquine(CQ) was provided to all volunteers in a st<strong>and</strong>ard<br />

prophylactic regimen of 300mg base weekly, starting with 600mg in two days,<br />

for a total duration of 13 weeks. Whilst under CQ prophylaxis, vera were<br />

exposed on three occasions, at four-weekly intervals, to bites of 12-15 P.<br />

falciparum-infected mosquitoes per session, for a total exposure of 36-45<br />

infected mosquitoes per volunteer. Controls received bites from an equal<br />

number of uninfected mosquitoes on the same occasions. Anopheles stephensi<br />

mosquitoes were reared according to st<strong>and</strong>ard procedures at our insectary.<br />

Infected mosquitoes were obtained by feeding on gametocytes of NF54, a<br />

chloroquine sensitive strain of P. falciparum, as described previously [13]. NF54<br />

is genetically homogeneous, but has not been formally cloned. Only the<br />

insectary technicians preparing the mosquitoes were aware of the infectivity<br />

status of the mosquitoes allocated to the volunteers. Blood-engorged<br />

mosquitoes were dissected to confirm the presence of sporozoites. If necessary,<br />

feeding sessions were repeated until precisely the predefined number of<br />

infected mosquitoes had fed. However, a single feeding session was sufficient in<br />

49/60 of all instances of immunization or challenge, whereas a second session<br />

was required in just 10 instances <strong>and</strong> a third session in only 1 instance.<br />

From day 6-10 after each mosquito exposure, all volunteers were followed on an<br />

outpatient basis <strong>and</strong> blood was drawn for st<strong>and</strong>ard whole blood counts <strong>and</strong> daily<br />

thick smears. Any signs <strong>and</strong> symptoms were recorded by the attending physician<br />

as follows: mild (easily tolerated), moderate (interferes with normal activity), or<br />

severe (prevents normal activity).<br />

Eight weeks after the last immunization dose <strong>and</strong> 4 weeks after discontinuation<br />

of CQ prophylaxis, all 15 volunteers were challenged by exposure to the bites of<br />

five homologous NF54 strain P. falciparum-infected mosquitoes. This period was<br />

considered to be sufficient for CQ levels to drop below levels which might be<br />

inhibitory to parasite multiplication [14]. Volunteers were checked daily on an<br />

outpatient basis from day 5 to day 21 for symptoms <strong>and</strong> signs of malaria,<br />

haematological parameters <strong>and</strong> thick smears.<br />

As soon as thick smear positive, volunteers were treated with a st<strong>and</strong>ard<br />

curative regimen of artemether/lumefantrine (starting dose of 80/480 mg,


178 Chapter 9<br />

followed by five identical doses at 8, 24, 36, 48 <strong>and</strong> 60 hours) <strong>and</strong> followed<br />

closely for three days. Complete cure was confirmed by thick blood smears. All<br />

volunteers still negative by day 21 post-challenge were presumptively treated<br />

with artemether/lumefantrine.<br />

Haematological <strong>and</strong> biochemical parameters were determined in routine fashion<br />

at the hospital’s central clinical laboratory. Nucleic Acid Sequence Base<br />

Amplification (NASBA) <strong>and</strong> real-time QT-PCR to determine the densities of P.<br />

falciparum parasites have been described in detail [15,16]. Chloroquine levels<br />

were measured by liquid chromatography as previously described [17,18].<br />

Minimum therapeutic concentrations for plasma chloroquine levels maintained<br />

by the laboratory were 30 µg/l, in accordance with Rombo et al. [14].<br />

Immunological analysis<br />

Venous whole blood was collected into CPT vacutainers (Becton <strong>and</strong> Dickinson,<br />

Basel) prior to first immunization <strong>and</strong> again prior to challenge. Plasma was<br />

collected <strong>and</strong> stored at -70°C. Peripheral Blood Mononuclear Cells [PBMCs)<br />

were isolated by density gradient centrifugation, frozen down in 10% DMSO/FCS<br />

<strong>and</strong> stored in liquid nitrogen. Antibody titres were assessed by ELISA <strong>and</strong><br />

immunofluorescence assay according st<strong>and</strong>ard protocols as previously described<br />

[19-21]. Cellular responses to cryopreserved asexual parasites were assessed by<br />

24-hour in vitro PBMC cell stimulation assays as previously described [22],<br />

followed by intracellular cytokine staining (Fix & Perm kit, Caltag Laboratories)<br />

<strong>and</strong> flow-cytometry. A more detailed description of these immunological assays<br />

is provided in the web-only supplementary methods.<br />

Statistical analysis<br />

Flowcytometric analysis was performed using CellQuest <strong>and</strong> data analyzed in<br />

SPSS; differences in responses within volunteers between time points <strong>and</strong><br />

between vera <strong>and</strong> control volunteers were analyzed by non-parametric tests<br />

(Wilcoxon <strong>and</strong> Mann-Whitney-U, respectively), two-sided p-values


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 179<br />

Abdominal pain mild 2<br />

moderate<br />

severe<br />

Vera [n=10) Controls [n=5)<br />

Post immunization<br />

Post<br />

Post immunization<br />

challenge<br />

Post<br />

challenge<br />

I II III I II III<br />

Fatigue mild 1 1 7 1 2<br />

moderate 2 1 1 2<br />

severe 1<br />

Fever mild 2<br />

moderate 1<br />

severe 2 5<br />

Headache mild 4 7 2 3 2<br />

moderate 2 1 1 1 1 3<br />

severe 1 1<br />

Loss of appetite mild 1<br />

moderate<br />

severe<br />

Malaise mild 1 1 1<br />

moderate 2 1<br />

severe 1 1 5<br />

Myalgia mild 1 2 3<br />

moderate 2 1 4<br />

severe 1<br />

Nausea mild 3 1 1 1 1<br />

moderate 1 1 1 1<br />

severe 1<br />

Vomiting mild 1<br />

moderate 1<br />

severe<br />

Overall† mild 4<br />

[40%)<br />

moderate 3<br />

[30%)<br />

severe 2<br />

[20%)<br />

1<br />

[10%)<br />

1<br />

[10%)<br />

8<br />

[80%)<br />

1<br />

[10%)<br />

1 3<br />

[20%) [60%)<br />

1 2 1<br />

[20%) [40%) [20%)<br />

1<br />

[20%)<br />

5<br />

[100%)<br />

Table 1. Adverse events after the first, second, <strong>and</strong> third exposures to immunizing<br />

mosquito bites <strong>and</strong> after challenge with infectious mosquito bites.<br />

* Subjects could have more than one adverse event <strong>and</strong> reports of events could have<br />

been either solicited or unsolicited. Only adverse events possibly or probably related to<br />

the study are listed.† The highest-grade event is listed per subject per <strong>infection</strong>.† Highest<br />

grade event listed per volunteer per <strong>infection</strong>.


180 Chapter 9<br />

Figure 2. Parasitemia in the Vaccine Group <strong>and</strong> the Control Group.<br />

Panel A shows the mean number of Plasmodium falciparum parasites per millilitre, as<br />

measured by nucleic acid sequence–based amplification (NASBA), after each of three<br />

immunizations on days before <strong>infection</strong> <strong>and</strong> during expected blood-stage parasitemia<br />

(days 6 to 10) in the vaccine group. The numbers of subjects who had positive results<br />

on peripheral blood smears for malarial parasites (MPS) <strong>and</strong> NASBA are shown below<br />

the graph. Panel B shows the mean number of P. falciparum parasites in 5 subjects in<br />

the control group <strong>and</strong> 10 subjects in the vaccine group, as determined by real-time<br />

polymerase-chain-reaction assay before treatment with artemether–lumefantrine.<br />

The I bars denote st<strong>and</strong>ard errors.


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 181<br />

No parasites were seen in the thick blood-smears of any of the 10 vera following<br />

each of the three immunization sessions under CQ prophylaxis, but after the first<br />

immunization a brief sub-microscopic parasitemic episode was detected in all<br />

vera (Figure 2a). This was not unexpected, since CQ has no effect <strong>against</strong> either<br />

sporozoites, liver-stage parasites, or the early ring forms of the first generation<br />

of blood-stage parasites caused by merozoites released from mature hepatic<br />

schizonts [11]. Following each of the subsequent two immunizations, a<br />

progressively reduced incidence <strong>and</strong> burden of sub-microscopic parasitemia was<br />

seen.<br />

In line with these findings, all vera reported solicited or unsolicited adverse<br />

events at least once during the immunization phase. When excluding local<br />

itching after the mosquito bites, adverse events were most commonly reported<br />

after the first immunization (9/10 volunteers), with headache being the most<br />

frequent (7/10) (Table 1). Only few adverse events occurred subsequently (0/10<br />

<strong>and</strong> 2/10 volunteers after the second <strong>and</strong> third immunization respectively).<br />

Severe adverse events were reported by three vera: two experienced fever<br />

above 39°C after the first immunization, one reported severe malaise after the<br />

last immunization.<br />

Following challenge with homologous NF54 strain of P. falciparum, asexual<br />

blood-stage parasites were detected in thick blood smears of all 5 control<br />

volunteers between days 7 <strong>and</strong> 10.6 post-exposure (mean pre-patent period 9.2<br />

days). Real-time PCR analyses revealed the expected cyclical multiplication of<br />

blood-stage parasites (Figure 2b). Clinical course <strong>and</strong> kinetics of parasite<br />

multiplication were identical to those in previous studies involving naïve<br />

volunteers [23,24], with all controls reporting severe events, in particular fever<br />

above 39°C <strong>and</strong> malaise (Table 1). In marked contrast, there was no evidence of<br />

blood-stage parasites in any of the vera at any time during the post-challenge<br />

follow-up period till day 20, either by repeated microscopy of thick blood smears<br />

or by real-time PCR analyses (Figure 2b). Interestingly, 9/10 vera did report mild<br />

to moderate events in the week following challenge. No serious adverse events<br />

occurred during any part of the trial <strong>and</strong> all 15 volunteers completed follow-up<br />

according to protocol.<br />

Mean peak chloroquine <strong>and</strong> desbutyl-chloroquine levels measured 24 hours<br />

after administration were 76 µg/l (range 58-104 µg/l) <strong>and</strong> 13 µg/l (5-33 µg/l)<br />

respectively <strong>and</strong> did not differ between control <strong>and</strong> vera. The day before<br />

challenge plasma levels had dropped to 8 µg/l (range


182 Chapter 9<br />

Test Day I-1 Day C-1<br />

Vaccine group<br />

(N=10)<br />

Control group<br />

(N=5)<br />

Vaccine group<br />

(N=10)<br />

Control group<br />

(N=5)<br />

No. of Median<br />

No. of<br />

Antibody<br />

Median<br />

No. of<br />

Antibody<br />

Median<br />

Antibody<br />

No. of Median<br />

Antibody<br />

Subjects Titre Subjects Titre Subjects Titre Subjects Titre<br />

AU AU AU AU<br />

CSP 0


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 183<br />

Figure 3 The proportion of lymphocytes that produced interferon-γ (IFN-γ) <strong>and</strong> interleukin-2 (Panel A),<br />

tumour necrosis factor α (TNF-α) <strong>and</strong> interleukin-2 (Panel B), or IFN-γ, TNF-α, <strong>and</strong> interleukin-2 (Panel<br />

C) after in vitro stimulation with erythrocytes infected with the homologous strain of P. falciparum<br />

(PfRBC), with uninfected erythrocytes (uRBC), or with phytohemagglutinin (PHA) as a positive control<br />

are shown before immunization (day I-1) <strong>and</strong> before malaria challenge (day C-1). Dashed lines<br />

represent the proportion of positive cells in unstimulated wells (culture medium only). The geometric<br />

mean fluorescence intensity of cells producing IFN-γ (Panel D), TNF-α (Panel E), <strong>and</strong> interleukin-<br />

2(Panel F) that were isolated from vaccinees on day C-1 is shown after stimulation in vitro with<br />

infected erythrocytes. Cells are grouped according to their positivity or negativity for each of the<br />

other two cytokines. In Panels G, H, <strong>and</strong> I, the proportion of lymphocytes that produced IFN-γ <strong>and</strong><br />

interleukin-2 in response to infected erythrocytes are shown on day I-1 <strong>and</strong> day C-1 for lymphocyte<br />

phenotypes, including naive T cells (CD3+CD45RO−), memory T cells (CD3+CD45RO+), <strong>and</strong> non-T<br />

lymphocytes (CD3−CD45RO−) (Panel G); for T-cell phenotypes, including helper T cells (CD4+CD8−),<br />

cytotoxic T cells (CD4−CD8+), <strong>and</strong> other lymphocytes (CD4−CD8−) (Panel H); <strong>and</strong> for memory<br />

phenotypes, including naive T cells (CD62L+CD45RO−), central memory T cells (CD62L+CD45RO+),<br />

effector memory T cells (CD62L−CD45RO+), <strong>and</strong> other lymphocytes (CD62L−CD45RO−) (Panel I). The<br />

proportions of lymphocytes that produced IFN-γ <strong>and</strong> interleukin-2 after stimulation with uninfected<br />

erythrocytes were below 0.005% (not shown). All P values are for the comparison between the<br />

vaccine group <strong>and</strong> the control group <strong>and</strong> were calculated with the use of the Mann–Whitney test. The<br />

T bars represent st<strong>and</strong>ard errors..


184 Chapter 9<br />

strain P. falciparum-infected erythrocytes (PfRBC) or controls (Figure 3 <strong>and</strong><br />

supplementary figures 2+3). Whereas cellular responses to uninfected<br />

erythrocytes (uRBC) never differed in any experiment from those to culture<br />

medium alone, stimulation with PfRBC elicited small percentages of lymphocytes<br />

producing IFNγ or TNFα, but not IL-2, in both groups of volunteers prior to<br />

immunization (day I-1, Supplementary figure 2). Although no statistically<br />

significant increment was seen in the overall percentage of cells producing<br />

individual cytokines (IFNγ+ or TNFα+) in either group following immunization<br />

(day C-1), a marked <strong>and</strong> significant increase was observed in the percentages of<br />

cells producing multiple cytokines in response to PfRBC in vera (IFNγ+IL-2+<br />

p=0.026, Figure 3a; TNFα+IL-2+ p=0.046, Figure 3b; IFNγ+TNFα+IL-2+ p=0.032,<br />

Figure 3c). The importance of these pluripotent lymphocytes in acquired<br />

immune <strong>protection</strong> is suggested by their higher cytokine content <strong>and</strong> possibly as<br />

a consequence a better effector function (Figure 3d-f). The major contributors to<br />

this increase in PfRBC-responding pluripotent lymphocytes were<br />

[CD3+CD45RO+) memory-like T cells (p=0.025 vs. day I-1, Figure 3g <strong>and</strong><br />

supplementary figure 3), in particular CD4+CD8- cells (p=0.005 vs. day I-1, Figure<br />

3h). Most noticeably, these new pluripotent lymphocytes were predominantly of<br />

the effector memory (CD62L-CD45RO+) phenotype (p=0.005 vs. day I-1),<br />

although there was also a small but significant (p=0.017) increase in the<br />

numbers of responding central memory (CD62L+CD45RO+) cells in vera (Figure<br />

3i).<br />

Discussion<br />

Our study shows that sterile <strong>protection</strong> to an homologous challenge with P.<br />

falciparum malaria can be much more efficiently induced in comparison to<br />

irradiation-attenuated sporozoite immunization. In the endemic situation, nonsterile<br />

semi-immunity is acquired only after years of repeated natural exposure.<br />

The improved efficiency of our approach we believe to be due to a critical<br />

balance of exposure to pre- <strong>and</strong> intra-erythrocytic antigens. In contrast to<br />

irradiated sporozoites that arrest early during liver-stage development [4], intact<br />

sporozoites under CQ cover mature fully <strong>and</strong> develop into a first generation of<br />

blood-stage parasites [11], thus presenting to the host’s immune system a<br />

markedly broader palette of pre-erythrocytic <strong>and</strong> additionally, albeit at relatively<br />

low-dose, erythrocytic-stage antigens. The contribution of intra-erythrocytic<br />

antigens to the development of protective immunity is suggested by Pombo et<br />

al., who reported that repeated intravenous injection of ultra-low densities of


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 185<br />

blood-stage parasites followed by drug-cure with atovaquone/proguanil induced<br />

<strong>protection</strong> in human volunteers <strong>against</strong> a similarly low-dose blood-stage<br />

challenge [25]. However, caution needs to be exercised when interpreting the<br />

latter results, since residual anti-malarial drug concentrations may partially or<br />

even fully have accounted for the observed <strong>protection</strong> [26].<br />

In the field, in contrast, patent parasitemia typically develops before patients<br />

seek treatment. In these individuals, acute blood-stage <strong>infection</strong> may suppress<br />

the induction of protective pre-erythrocytic immunity, as has been shown in<br />

rodent models [27]. Indeed, parasitemic episodes or febrile malaria attacks in<br />

Kenyan children are prospectively associated with a poorer induction <strong>and</strong> more<br />

rapid attrition of cellular ex vivo <strong>and</strong> memory responses to a pre-erythrocytic P.<br />

falciparum antigen [28]. Thus, patent parasitemia <strong>and</strong> probably also chronic subpatent<br />

parasitemia, as are experienced regularly by children in endemic areas,<br />

appear to induce inhibitory mechanisms that delay the generation of protective<br />

anti-parasite immunity. Meta-analysis of intermittent preventative therapy in<br />

infancy (IPTi) studies has decreased concerns of a rebound-effect <strong>and</strong> in some<br />

cases even indicates sustained <strong>protection</strong> following discontinuation of<br />

prophylaxis [29], thus further indicating that the acquisition <strong>and</strong> maintenance of<br />

protective immunity is not dependent on chronic blood-stage exposure. The<br />

salient feature of our approach we believe therefore to be the exposure of the<br />

immune system to the full palette of pre-<strong>and</strong> intra-erythrocytic antigens, whilst<br />

restricting the development of symptomatic <strong>and</strong> potentially immunosuppressive<br />

parasitemia [30]. Since NF54 is known to be a CQ-sensitive strain in vitro [31],<br />

we cannot formally exclude a synergistic effect of residual sub-therapeutic<br />

chloroquine levels on immunological parasite clearance. However, chloroquine<br />

levels prior to challenge approached or fell below the limit of detection <strong>and</strong> had<br />

no measureable parasitocidal effect in control volunteers. Of more importance,<br />

the longevity of immunological responses, both naturally-acquired <strong>and</strong> vaccineinduced,<br />

remains a critical issue in malaria <strong>and</strong> follow-up studies are planned to<br />

address this issue.<br />

We have identified pluripotent effector memory T cell responses as associated<br />

with <strong>protection</strong>. Undefined lymphocyte subsets with the same cytokine profile<br />

have been associated with the induction <strong>and</strong> maintenance of antigen-specific T<br />

cell memory in individuals immunized with pre-erythrocytic malaria vaccine<br />

c<strong>and</strong>idates, but this study did not explore associations with <strong>protection</strong> [32]. The<br />

potent effector function of pluripotent cells, as suggested by their high cytokine<br />

content, has however been noted in other investigations demonstrating their


186 Chapter 9<br />

protective role in other infectious diseases [33,34]. Further detailed<br />

investigations will be necessary to ascertain the longevity of this immunological<br />

response, its association with central memory-type T cell activity <strong>and</strong> its ability<br />

to serve as a true correlate of <strong>protection</strong>.<br />

Since the magnitude of the first wave of parasitemia is thought to directly reflect<br />

the burden of erupting mature liver schizonts, the step-wise decrease of such<br />

following each subsequent immunizing <strong>infection</strong>, culminating in the total<br />

absence of PCR-detectable parasitemia following challenge, would suggest that<br />

the <strong>protection</strong> in our model is primarily due to pre-erythrocytic immunity.<br />

However, a component of blood-stage immunity, i.e. the inhibition of<br />

erythrocyte invasion <strong>and</strong> maturation of sub-PCR liver-derived merozoite inocula,<br />

is also possible. Indeed we found cellular responses to asexual blood-stage<br />

parasites [PfRBC) prior to challenge to be an excellent discriminative marker of<br />

exposure <strong>and</strong> <strong>protection</strong> in our volunteers <strong>and</strong> similar immune responses may<br />

have contributed to <strong>protection</strong> in the rodent model [12]. It must be borne in<br />

mind, however, that many of the best-studied P. falciparum antigens conferring<br />

protective immunity are shared between sporozoite, liver-stage <strong>and</strong> blood-stage<br />

parasites [35,36]. Thus it is plausible that our findings represent the response to<br />

a broad antigenic repertoire that transcends parasite developmental stages [37],<br />

making a dichotomy into pre-erythrocytic or intra-erythrocytic immunity<br />

inappropriate. At present the stage-specificity of the protective immune<br />

response must thus remain formally unresolved, although one way to further<br />

address this issue in future studies would be a blood-stage challenge.<br />

Whereas the methodology described here does not itself represent a widely<br />

implementable vaccine strategy, the induction of sterile <strong>protection</strong> <strong>against</strong> an<br />

homologous malaria challenge suggests that the concept of a whole parasite<br />

malaria vaccine warrants further consideration. In addition, this model allows<br />

the nature of protective immune responses <strong>against</strong> malaria, both stage- <strong>and</strong><br />

antigen-specific, to be further investigated.<br />

Acknowledgements<br />

Foremost, we are indebted to the study volunteers for their participation. We<br />

thank K. Nganou Makamdop for help with RT-PCR, J. Bakkers & W. Melchers for<br />

parasite genotyping, W. Arts, N. Huibers & P. Beckers for blood slide reading, P.<br />

Houze & D. Mazier for chloroquine measurements <strong>and</strong> J. Klaassen, L. Pelser-<br />

Posthumus, J. Kuhnen & A. Pouwelsen for technical assistance generating


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 187<br />

infected mosquitoes. We acknowledge the safety monitors on this study, B.<br />

Schouwenberg & P. Smits <strong>and</strong> independent physician A. Brouwer.<br />

Funding<br />

Supported by the Dioraphte Foundation, by a fellowship from the European<br />

Union FP6 Network of Excellence (to Dr. McCall), <strong>and</strong> by grants from the<br />

NutsOhra Foundation (to Dr. van der Ven) <strong>and</strong> Agence Nationale de la Recherche<br />

in France (to Dr. Snounou).


188 Chapter 9<br />

References<br />

1. Greenwood BM, Fidock DA, Kyle DE et al. Malaria: progress, perils, <strong>and</strong><br />

prospects for eradication. J Clin Invest 2008; 118:1266-76.<br />

2. Clyde DF, Most H, McCarthy VC, V<strong>and</strong>erberg JP. Immunization of man <strong>against</strong><br />

sporozite-induced falciparum malaria. Am J Med Sci 1973; 266:169-77.<br />

3. Nussenzweig RS, V<strong>and</strong>erberg J, Most H, Orton C. Protective immunity produced<br />

by the injection of x-irradiated sporozoites of plasmodium berghei. Nature<br />

1967; 216:160-2.<br />

4. Mellouk S, Lunel F, Sedegah M, Beaudoin RL, Druilhe P. Protection <strong>against</strong><br />

malaria induced by irradiated sporozoites. Lancet 1990; 335:721.<br />

5. Hoffman SL, Goh LM, Luke TC et al. Protection of humans <strong>against</strong> malaria by<br />

immunization with radiation-attenuated Plasmodium falciparum sporozoites. J<br />

Infect Dis 2002; 185:1155-64.<br />

6. Alonso PL, Sacarlal J, Aponte JJ et al. Duration of <strong>protection</strong> with RTS,S/AS02A<br />

malaria vaccine in prevention of Plasmodium falciparum disease in Mozambican<br />

children: single-blind extended follow-up of a r<strong>and</strong>omised controlled trial.<br />

Lancet 2005; 366:2012-8.<br />

7. Abdulla S, Oberholzer R, Juma O et al. Safety <strong>and</strong> immunogenicity of<br />

RTS,S/AS02D malaria vaccine in infants. N Engl J Med 2008; 359:2533-44.<br />

8. Bejon P, Lusingu J, Olotu A et al. Efficacy of RTS,S/AS01E vaccine <strong>against</strong> malaria<br />

in children 5 to 17 months of age. N Engl J Med 2008; 359:2521-32.<br />

9. Aponte JJ, Aide P, Renom M et al. Safety of the RTS,S/AS02D c<strong>and</strong>idate malaria<br />

vaccine in infants living in a highly endemic area of Mozambique: a double blind<br />

r<strong>and</strong>omised controlled phase I/IIb trial. Lancet 2007; 370:1543-51.<br />

10. Beaudoin RL, Strome CP, Mitchell F, Tubergen TA. Plasmodium berghei:<br />

immunization of mice <strong>against</strong> the ANKA strain using the unaltered sporozoite as<br />

an antigen. Exp Parasitol 1977; 42:1-5.<br />

11. Yayon A, V<strong>and</strong>e Waa JA, Yayon M, Geary TG, Jensen JB. Stage-dependent effects<br />

of chloroquine on Plasmodium falciparum in vitro. J Protozool 1983; 30:642-7.<br />

12. Belnoue E, Costa FT, Frankenberg T et al. Protective T cell immunity <strong>against</strong><br />

malaria liver stage after vaccination with live sporozoites under chloroquine<br />

treatment. J Immunol 2004; 172:2487-95.<br />

13. Ponnudurai T, Lensen AH, van Gemert GJ, Bensink MP, Bolmer M, Meuwissen<br />

JH. Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes.<br />

Parasitology 1989; 98 Pt 2:165-73.<br />

14. Rombo L, Bergqvist Y, Hellgren U. Chloroquine <strong>and</strong> desethylchloroquine<br />

concentrations during regular long-term malaria prophylaxis. Bull World Health<br />

Organ 1987; 65:879-83.


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 189<br />

15. Hermsen CC, Telgt DS, Linders EH et al. Detection of Plasmodium falciparum<br />

malaria parasites in vivo by real-time quantitative PCR. Mol Biochem Parasitol<br />

2001; 118:247-51.<br />

16. Schneider P, Wolters L, Schoone G et al. Real-time nucleic acid sequence-based<br />

amplification is more convenient than real-time PCR for quantification of<br />

Plasmodium falciparum. J Clin Microbiol 2005; 43:402-5.<br />

17. Houze P, de RA, Baud FJ, Benatar MF, Pays M. Simultaneous determination of<br />

chloroquine <strong>and</strong> its three metabolites in human plasma, whole blood <strong>and</strong> urine<br />

by ion-pair high-performance liquid chromatography. J Chromatogr 1992;<br />

574:305-12.<br />

18. Lejeune D, Souletie I, Houze S et al. Simultaneous determination of<br />

monodesethylchloroquine, chloroquine, cycloguanil <strong>and</strong> proguanil on dried<br />

blood spots by reverse-phase liquid chromatography. J Pharm Biomed Anal<br />

2007; 43:1106-15.<br />

19. Bousema JT, Roeffen W, van der KM et al. Rapid onset of transmission-reducing<br />

antibodies in javanese migrants exposed to malaria in papua, indonesia. Am J<br />

Trop Med Hyg 2006; 74:425-31.<br />

20. Hermsen CC, Verhage DF, Telgt DS et al. Glutamate-rich protein [GLURP)<br />

induces antibodies that inhibit in vitro growth of Plasmodium falciparum in a<br />

phase 1 malaria vaccine trial. Vaccine 2007; 25:2930-40.<br />

21. Remarque EJ, Faber BW, Kocken CH, Thomas AW. A diversity-covering approach<br />

to immunization with Plasmodium falciparum apical membrane antigen 1<br />

induces broader allelic recognition <strong>and</strong> growth inhibition responses in rabbits.<br />

Infect Immun 2008; 76:2660-70.<br />

22. McCall MB, Netea MG, Hermsen CC et al. Plasmodium falciparum <strong>infection</strong><br />

causes proinflammatory priming of human TLR responses. J Immunol 2007;<br />

179:162-71.<br />

23. Verhage DF, Telgt DS, Bousema JT et al. Clinical outcome of experimental<br />

human malaria induced by Plasmodium falciparum-infected mosquitoes. Neth J<br />

Med 2005; 63:52-8.<br />

24. Hermsen CC, de Vlas SJ, van Gemert GJ, Telgt DS, Verhage DF, Sauerwein RW.<br />

Testing vaccines in human experimental malaria: statistical analysis of<br />

parasitemia measured by a quantitative real-time polymerase chain reaction.<br />

Am J Trop Med Hyg 2004; 71:196-201.<br />

25. Pombo DJ, Lawrence G, Hirunpetcharat C et al. Immunity to malaria after<br />

administration of ultra-low doses of red cells infected with Plasmodium<br />

falciparum. Lancet 2002; 360:610-7.<br />

26. Edstein MD, Kotecka BM, Anderson KL et al. Lengthy antimalarial activity of<br />

atovaquone in human plasma following atovaquone-proguanil administration.<br />

Antimicrob Agents Chemother 2005; 49:4421-2.


190 Chapter 9<br />

27. Orjih AU. Acute malaria prolongs susceptibility of mice to Plasmodium berghei<br />

sporozoite <strong>infection</strong>. Clin Exp Immunol 1985; 61:67-71.<br />

28. Bejon P, Mwacharo J, Kai O et al. The induction <strong>and</strong> persistence of T cell IFNgamma<br />

responses after vaccination or natural exposure is suppressed by<br />

Plasmodium falciparum. J Immunol 2007; 179:4193-201.<br />

29. Grobusch MP, Egan A, Gosling RD, Newman RD. Intermittent preventive<br />

therapy for malaria: progress <strong>and</strong> future directions. Curr Opin Infect Dis 2007;<br />

20:613-20.<br />

30. Sutherl<strong>and</strong> CJ, Drakeley CJ, Schellenberg D. How is childhood development of<br />

immunity to Plasmodium falciparum enhanced by certain antimalarial<br />

interventions? Malar J 2007; 6:161.<br />

31. Davis JR, Cortese JF, Herrington DA et al. Plasmodium falciparum: in vitro<br />

characterization <strong>and</strong> human infectivity of a cloned line. Exp Parasitol 1992;<br />

74:159-68.<br />

32. Bejon P, Keating S, Mwacharo J et al. Early gamma interferon <strong>and</strong> interleukin-2<br />

responses to vaccination predict the late resting memory in malaria-naive <strong>and</strong><br />

malaria-exposed individuals. Infect Immun 2006; 74:6331-8.<br />

33. Darrah PA, Patel DT, De Luca PM et al. Multifunctional TH1 cells define a<br />

correlate of vaccine-mediated <strong>protection</strong> <strong>against</strong> Leishmania major. Nat Med<br />

2007; 13:843-50.<br />

34. Precopio ML, Betts MR, Parrino J et al. Immunization with vaccinia virus induces<br />

polyfunctional <strong>and</strong> phenotypically distinctive CD8[+) T cell responses. J Exp Med<br />

2007; 204:1405-16.<br />

35. Hogh B, Thompson R, Zakiuddin IS, Boudin C, Borre M. Glutamate rich<br />

Plasmodium falciparum antigen [GLURP). Parassitologia 1993; 35 Suppl:47-50.<br />

36. Silvie O, Franetich JF, Charrin S et al. A role for apical membrane antigen 1<br />

during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol<br />

Chem 2004; 279:9490-6.<br />

37. Krzych U, Lyon JA, Jareed T et al. T lymphocytes from volunteers immunized<br />

with irradiated Plasmodium falciparum sporozoites recognize liver <strong>and</strong> blood<br />

stage malaria antigens. J Immunol 1995; 155:4072-7.


Protection <strong>against</strong> a Malaria Challenge by Sporozoite Inoculation 191<br />

Supplementary methods<br />

Serology<br />

To assess antibody titres, asexual ELISAs were performed as previously described<br />

[1]. CSP ELISAs were performed with (NANP)6 NA (kind gift from G. Corradin)<br />

according to published methods [2]. AMA-1 <strong>and</strong> GLURP ELISAs were performed<br />

according to a st<strong>and</strong>ard in-house protocol [2,3]. In all ELISAs a plasma pool of 8<br />

healthy malaria-naïve Dutch volunteers was used as a negative control to define<br />

sample positivity (above mean+2SD of negative control). A plasma pool of<br />

Tanzanian adults (n=100) living in a highly malaria endemic area was used as<br />

reference positive control, defined to contain 100 arbitrary units (AU).<br />

Immunofluorescence assays were performed by incubating plasma at 1/320<br />

dilution with air-dried whole sporozoites or at 1/40 dilution with asynchronous<br />

cultures of NF54 strain asexual P. falciparum parasites, as previously described<br />

[3].<br />

Cellular immunology<br />

Cellular immune responses were assessed by in vitro stimulation assays. For use<br />

in these assays, Percoll-purified asynchronous asexual-stage cultures of NF54<br />

strain parasites of 80-90% parasitemia, consisting of more than 95%<br />

schizonts/mature trophozoites, were washed, aliquoted <strong>and</strong> cryopreserved in<br />

advance. Mock-cultured uninfected erythrocytes (uRBC) were obtained similarly<br />

<strong>and</strong> served as control. Cryopreserved PBMCs were thawed immediately prior to<br />

use in in vitro stimulation assays, washed <strong>and</strong> resuspended in RPMI 1640 culture<br />

medium containing 2mM glutamine, 1mM pyruvate, 50μg/mL gentamicine <strong>and</strong><br />

10% pooled human AB+ serum (Sanquin, Nijmegen, NL), for a final concentration<br />

of 2.5x10 6 /mL. PBMCs were transferred into 96-well round-bottom plates<br />

(5x10 5 /well) <strong>and</strong> stimuli were added to duplicate wells. Stimuli included<br />

cryopreserved PfRBC (5x10 6 /mL), uRBC, PHA (10 μg/mL) or RPMI only. PBMCs<br />

were stimulated for 24 hours at 37°C/5%CO2. 4 hours prior to cell harvest,<br />

100μL/well supernatant was collected <strong>and</strong> replaced with 100 μL/well fresh<br />

culture medium containing 20 μg/mL brefeldin A (Sigma). PBMCs from both time<br />

points per volunteer were tested simultaneously <strong>and</strong> cells from two vaccinees<br />

<strong>and</strong> one control were always tested together.<br />

Following 24 hour in vitro stimulation, PBMCs were harvested, washed once in<br />

FACS buffer (0.5% BSA/PBS) <strong>and</strong> incubated for 15’ with fluorescent mAbs <strong>against</strong><br />

cell surface markers. Cells were washed again <strong>and</strong> incubated for 15’ in fixation


192 Chapter 9<br />

medium A (Caltag Laboratories, Carlsbad, CA) according to the manufacturer’s<br />

instructions, washed again <strong>and</strong> incubated for 15’ with fluorescent mAbs <strong>against</strong><br />

intracellular cytokines in permeabilization medium B. After a final wash step,<br />

cells were resuspended in FACS buffer <strong>and</strong> read on a FACScalibur flowcytometer.<br />

The following fluorescent mAbs were used: CD3-PerCP, CD4-PerCP, CD8-PE (BD,<br />

Breda, NL), IFNγ-FITC, TNFα-PE, IL-2-APC, CD45RO-PE, CD62L-PeCy7, mouse<br />

IgG1-FITC & IgG2a-PE <strong>and</strong> rat IgG2a-APC isotype controls (all Ebioscience,<br />

Uithoorn, NL).<br />

References<br />

1. Bousema JT, Roeffen W, van der KM et al. Rapid onset of transmissionreducing<br />

antibodies in javanese migrants exposed to malaria in Papua,<br />

Indonesia. Am J Trop Med Hyg 2006; 74:425-31.<br />

2. Remarque EJ, Faber BW, Kocken CH, Thomas AW. A diversity-covering<br />

approach to immunisation with Plasmodium falciparum AMA1 induces broader<br />

allelic recognition <strong>and</strong> growth inhibition responses in rabbits. Infect Immun<br />

2008.<br />

3. Hermsen CC, Verhage DF, Telgt DS et al. Glutamate-rich protein (GLURP)<br />

induces antibodies that inhibit in vitro growth of Plasmodium falciparum in a<br />

phase 1 malaria vaccine trial. Vaccine 2007; 25:2930-40.


Chapter 10<br />

Long-term <strong>protection</strong> <strong>against</strong> malaria after<br />

experimental sporozoite inoculation<br />

Meta Roestenberg 1 , Anne C. Teirlinck 1 , Matthew B.B. McCall 1 , Karina Teelen 1 ,<br />

Krystelle Nganou Makamdop 1 , Jorien Wiersma 1 , Theo Arens 1 , Pieter Beckers 1 ,<br />

GeertJan van Gemert 1 , Marga van de Vegte-Bolmer 1 , André J.A.M. van der Ven 2 ,<br />

Adrian J.F. Luty 1 , Cornelus C. Hermsen 1 , Robert W. Sauerwein 1<br />

1<br />

Radboud University Nijmegen Medical Centre, Department of Medical<br />

Microbiology, Nijmegen, The Netherl<strong>and</strong>s<br />

2<br />

Radboud University Nijmegen Medical Centre, Department of Internal<br />

Medicine, Nijmegen, The Netherl<strong>and</strong>s [A J A M van der Ven)<br />

Lancet 2011; 377:1770-1776


194 Chapter 10<br />

Abstract<br />

Induction of long-lived immunity to Plasmodium falciparum (Pf) is a major<br />

obstacle to malaria vaccine development. Recently we showed that immunity to<br />

Pf can be induced experimentally in 10/10 of malaria-naïve volunteers through<br />

immunisation by bites of Pf-infected mosquitoes whilst simultaneously<br />

preventing disease by chloroquine prophylaxis. This immunity was associated<br />

with parasite-specific production of IFNγ <strong>and</strong> IL-2 by pluripotent effector<br />

memory cells in vitro. Here we explore the persistence of <strong>protection</strong> <strong>and</strong><br />

immune responses in the same volunteers.<br />

In an open-label study conducted 28 months after immunisation, six previously<br />

immune volunteers were re-challenged by the bites of five Pf-infected<br />

mosquitoes. Five naive volunteers served as <strong>infection</strong> controls. The primary<br />

outcome was detection of blood-stage parasitemia by microscopy. The kinetics<br />

of parasitemia were assessed by real-time quantitative PCR (Q-PCR) <strong>and</strong> clinical<br />

signs <strong>and</strong> symptoms were recorded. In vitro production of IFNγ <strong>and</strong> IL-2 by<br />

effector memory T cells was studied following stimulation with sporozoites <strong>and</strong><br />

Pf-infected red blood cells. This study is registered with clinicaltrials.gov, number<br />

NCT00757887.<br />

Four of six immune volunteers were fully protected <strong>against</strong> re-challenge. Q-PCRbased<br />

detection of blood-stage parasites in these individuals was negative<br />

throughout follow-up. Patency in the remaining two immunised volunteers was<br />

markedly delayed. In vitro assays revealed the long-term persistence of parasitespecific<br />

pluripotent effector memory T cell responses in protected volunteers.<br />

We demonstrate that immunity to Pf in human volunteers can persist for more<br />

than two years using an experimental immunisation protocol. Immunity was<br />

paralleled by maintenance of Pf-specific pluripotent effector memory T cells.<br />

These findings are unprecedented <strong>and</strong> demonstrate that artificially induced<br />

immunity may be longer-lasting than generally observed after natural exposure.<br />

These results open a novel avenue for research into mechanisms of malaria<br />

immunity.


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 195<br />

Introduction<br />

In 2009, an estimated 225 million cases of Plasmodium falciparum malaria lead<br />

to 781,000 deaths worldwide, of which about 85% were children under the age<br />

of five (http://www.who.int/malaria/world_malaria_report_2010/en/index.<br />

html). Malaria is responsible for an enormous economic burden due to medical<br />

expenses, missed education <strong>and</strong> lost productivity. It has been estimated that the<br />

GDP of countries endemic for malaria grows at 1.3% per year slower than<br />

countries which are malaria-free [1]. Reducing the burden of malaria has been<br />

identified as a specific target within the United Nations Millennium<br />

Development Goal 4: reduce Child Mortality (www.un.org/millenniumgoals).<br />

Control programs have been successful in decreasing the incidence of malaria by<br />

more than 50% in 11 countries of the WHO African region over the past decade<br />

<strong>and</strong> are highly cost-effective [2]. The recent increase of malaria cases in some<br />

other African countries illustrates the fragility of current malaria control<br />

(http://www.who.int/malaria/world_malaria_report_2010/en/index.html). The<br />

urgent need for an effective malaria vaccine to complement these control<br />

programs is thus widely acknowledged. A major hurdle in vaccine development<br />

is the complex parasite-host interaction that seems to preclude the<br />

establishment of long-lasting, sterile anti-parasitic immunity [3]. This notion is<br />

supported by the lack of sustained <strong>protection</strong> acquired through natural malaria<br />

exposure. When lifelong residents move away from an endemic area, immunity<br />

is thought to wane quickly, rendering them susceptible to re-<strong>infection</strong> <strong>and</strong><br />

disease when re-exposed. [4] Moreover, naturally acquired immunity controls<br />

parasitemia <strong>and</strong> prevents clinical symptoms but neither eliminates parasites<br />

completely from the blood nor prevents re-<strong>infection</strong>, <strong>and</strong> is therefore not<br />

sterilizing [4].<br />

Immunity to Plasmodium falciparum (Pf) malaria in humans can be induced<br />

artificially by vaccination with subunit vaccine c<strong>and</strong>idates or by repeated<br />

exposure to attenuated whole parasites [5,6]. The most successful subunit<br />

c<strong>and</strong>idate vaccine to date, RTS,S, based on the circumsporozoite protein<br />

combined with hepatitis B surface antigen, is between 30-65% efficacious in<br />

delaying the first episode of clinical malaria in Africans [7]. Extended follow-up in<br />

Mozambican children demonstrated waning efficacy over a period of 20 months<br />

[8]. Immunity to Pf in humans can also be induced experimentally by exposure<br />

to the bites of more than 1000 irradiated sporozoite-infected mosquitoes. This<br />

results in <strong>protection</strong> of almost 100% volunteers, lasting up to 42 weeks in a few


196 Chapter 10<br />

Pre-erythrocytic stage<br />

Sporozoites<br />

Liver<br />

Blood stage<br />

Chloroquine<br />

mediated<br />

killing<br />

Merosome<br />

Merozoites<br />

individuals [5]. Recently we described a novel methodology for induction of<br />

immunity to Pf in humans, based on an immunisation procedure involving the<br />

repeated exposure to infectious mosquito bites under chloroquine prophylaxis<br />

[9]. Using this method, clinical disease was prevented by the drug that kills<br />

asexual blood-stage parasites, but allows complete liver-stage parasite<br />

development (Figure 1). Volunteers were protected from an experimental<br />

challenge with the homologous parasite strain two months later. Protected<br />

volunteers developed only low-level parasite-specific antibody activity, but<br />

strong parasite-specific effector memory T cell responses, characterized by the<br />

production of IFNγ <strong>and</strong> IL-2 after stimulation in vitro.<br />

The objective of the current follow-up study was to assess the persistence of<br />

<strong>protection</strong> <strong>against</strong> Pf <strong>infection</strong> after experimental immunisation. We rechallenged<br />

volunteers by the bites of sporozoite-infected mosquitoes after a<br />

period of 2.5 years.<br />

Skin<br />

Erythrocytes<br />

Lymph node<br />

Figure 1. Course of Plasmodium falciparum parasite development under<br />

chloroquine prophylaxis. Sporozoites are injected into the skin by an infected<br />

female Anopheles sp. mosquito. A proportion migrates through the bloodstream<br />

to the liver. Parasites develop <strong>and</strong> multiply in hepatocytes over a period of 6-7<br />

days. Subsequently, merosomes bud from infected hepatocytes into the sinusoid<br />

<strong>and</strong> the released merozoites invade red blood cells for further asexual<br />

multiplication. Chloroquine prophylaxis only targets <strong>and</strong> prevents replication of<br />

parasites in red blood cells, thereby allowing the induction of immune responses<br />

<strong>against</strong> preceding stages.


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 197<br />

Methods<br />

Number of fully engorged mosquitoes [# of<br />

mosquitoes per cage)<br />

Volunteer number Session 1 Session 2 Session 3<br />

1 – Control 5 [5)<br />

2 – Control 5 [5)<br />

3 – Control 5 [5)<br />

4 – Control 3 [5) 2 [2)<br />

5 – Control 5 [5)<br />

6 – Immunised 4 [5) 1 [1)<br />

7 – Immunised 4 [5) 1 [1)<br />

8 – Immunised 5 [5)<br />

9 – Immunised 4 [5) 1 [1)<br />

10 – Immunised 3 [5) 2 [2)<br />

11 – Immunised 4 [5) 0 [1) 1 [1)<br />

Table 1. Number of blood feeding mosquitoes per volunteer.<br />

Participants <strong>and</strong> study design<br />

This open-label clinical trial follows a previous trial where immunity to Pf malaria<br />

was induced in ten healthy, malaria-naïve volunteers [9]. In short, volunteers<br />

took chloroquine prophylaxis weekly for a period of 13 weeks during which they<br />

were exposed to the bites of 12-15 infected mosquitoes on three occasions at<br />

monthly intervals. Figure 1 gives an overview of the course of development of<br />

parasites during these immunising <strong>infection</strong>s. One month after stopping the<br />

prophylaxis, all ten volunteers were shown to be fully protected <strong>against</strong> a<br />

subsequent challenge <strong>infection</strong> via the bites of five Pf-infected mosquitoes, with<br />

complete absence of asexual parasitemia.<br />

The current trial was performed at the Radboud University Nijmegen Medical<br />

Centre, The Netherl<strong>and</strong>s, from November to December 2009, twenty-eight<br />

months after the initiation of the previous challenge <strong>infection</strong>. All ten previously<br />

immune volunteers were invited to participate, six were eligible for<br />

participation. In addition, five malaria-naive volunteers were recruited as<br />

infectivity controls. Volunteers aged 18-35 years old were screened for eligibility<br />

based on medical <strong>and</strong> family history, physical examination <strong>and</strong> general<br />

haematological <strong>and</strong> biochemical screening including HIV, hepatitis B <strong>and</strong><br />

hepatitis C serology, urine toxicology screening <strong>and</strong> a pregnancy test. The main<br />

exclusion criteria were residence in a malaria endemic area within the previous<br />

six months, positive Pf serology (all volunteers were tested according to


198 Chapter 10<br />

procedures described in [10]) or an estimated ten year risk of >5% of developing<br />

a cardiac event as estimated by the Systematic Coronary Evaluation (SCORE)<br />

system. All volunteers gave written informed consent prior to inclusion. The trial<br />

was performed in accordance with Good Clinical Practice <strong>and</strong> approved by the<br />

Central Committee for Research Involving Human Subjects of The Netherl<strong>and</strong>s<br />

(CCMO NL24193.091.09). Clinicaltrials.gov identifier: NCT00757887.<br />

Procedures<br />

Both the six immunised, previously immune individuals, <strong>and</strong> the five malarianaive<br />

control volunteers were exposed to the bites of five Anopheles stephensi<br />

mosquitoes infected with the NF54 strain of Pf for ten minutes. Infected<br />

mosquitoes were obtained by feeding on gametocytes of NF54 as described<br />

previously [11]. The salivary gl<strong>and</strong>s of all blood-engorged mosquitoes were<br />

dissected, confirming the presence of sporozoites in 97%, with a mean of 88,000<br />

sporozoites per mosquito. When necessary, feeding sessions were repeated with<br />

a smaller number of mosquitoes until precisely five infected mosquitoes had<br />

bitten. (Five volunteers required one blood feeding session, five volunteers two<br />

sessions <strong>and</strong> one volunteer three sessions, raw data provided in Table 1).<br />

Starting from day five post-challenge <strong>infection</strong>, volunteers were subjected to an<br />

intense follow-up regime with multiple daily visits to our out-patient clinical<br />

research department. All signs <strong>and</strong> symptoms (solicited <strong>and</strong> unsolicited) were<br />

recorded <strong>and</strong> graded by the attending physician as follows: mild (easily<br />

tolerated), moderate (interferes with normal activity), or severe (prevents<br />

normal activity), or in case of fever grade 1 (>37.5°C – 38.0°C), grade 2 (>38.0°C –<br />

39.0°C) or grade 3 (>39.0°C). Haematological <strong>and</strong> biochemical parameters were<br />

monitored daily. Because of a previously reported serious cardiac adverse event<br />

after a malaria challenge <strong>infection</strong> in a separate study [12], particular attention<br />

was paid to markers of coagulation or cardiac damage with daily follow-up of<br />

highly-sensitive troponin, platelets, d-dimer <strong>and</strong> lactate dehydrogenase during<br />

the period when blood stage parasitemia could be expected. Whenever<br />

abnormal, blood samples were checked for the presence of fragmentocytes <strong>and</strong><br />

von Willebr<strong>and</strong> cleaving protease activity, since previous reports have<br />

highlighted the occurrence of von Willebr<strong>and</strong> factor activation in experimental<br />

malaria [13].<br />

As soon as a blood-slide was found to contain parasites, volunteers were treated<br />

with a curative regimen of atovaquone/proguanil 1000/400mg once daily for<br />

three days. Volunteers who remained free of parasites by blood-slide until day


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 199<br />

21 after re-challenge presumptively received the same curative treatment.<br />

Complete cure was always confirmed by the occurrence of two consecutive<br />

parasite-negative blood-slides.<br />

Outcomes<br />

The primary outcome of the study was microscopic detection of parasites in a<br />

blood-slide, with sampling performed twice daily on days 5 <strong>and</strong> 6 post-challenge,<br />

thrice daily on days 7-11, twice daily on days 12-15 <strong>and</strong> once on days 16-21 postchallenge.<br />

Thick blood smears were made from 15µl of EDTA-anti-coagulated<br />

blood, spread over the st<strong>and</strong>ardised surface of one well of a 3-well glass slide<br />

(CEL-LINE Diagnostic Microscope Slides, 30-12A-black-CE24). After drying, wells<br />

were stained with Giemsa for 45 minutes. Slides were read at 1000x<br />

magnification by assessing 200 high-power fields, equal to approximately 0.5µl<br />

of blood. The smear was considered positive if two unambiguously identifiable<br />

parasites were found. The pre-patent period was defined as the period between<br />

challenge <strong>and</strong> first positive blood smear. Additionally, parasitemia was<br />

measured by real-time quantitative PCR (Q-PCR), performed retrospectively on<br />

all samples collected after challenge, as previously described [14].<br />

Secondary outcomes comprised immunological parameters. After preparation of<br />

thick blood smears, remaining plasma was collected <strong>and</strong> stored at -70°C.<br />

Antibody titres on the day prior to challenge were assessed by ELISA as<br />

previously described [9]. Plasma concentrations of IL-6 <strong>and</strong> IFNγ were measured<br />

every other day using the Bio-Plex system (Bio-Rad) [15].<br />

For cellular immunology, venous whole blood was collected into cell-preparation<br />

tubes (Becton <strong>and</strong> Dickinson, Basel) on the day before challenge. Peripheral<br />

blood mononuclear cells (PBMC) were isolated by density gradient<br />

centrifugation, frozen in fetal-calf serum containing 10% dimethylsulfoxide <strong>and</strong><br />

stored in liquid nitrogen. Cellular responses <strong>against</strong> cryopreserved NF54<br />

sporozoite <strong>and</strong> NF54 asexual-stage parasites were assessed by 24-hour in vitro<br />

stimulation as previously described for the asexual-stage [9] followed by<br />

intracellular cytokine staining.<br />

Preparation of sporozoite- <strong>and</strong> asexual-stage parasites for immunological assays<br />

NF54 strain P. falciparum sporozoites (PfSpz) were obtained from Anopheles<br />

stephensi mosquitoes that were reared according to st<strong>and</strong>ard procedures in our<br />

insectary. Infected mosquitoes were obtained by feeding on gametocytecontaining<br />

cultures of NF54 strain P. falciparum, as described previously [15]. On<br />

day 21–28 after <strong>infection</strong>, the salivary gl<strong>and</strong>s of the mosquitoes were collected


200 Chapter 10<br />

by h<strong>and</strong>-dissection. Salivary gl<strong>and</strong>s were collected in RPMI-1640 medium (Gibco)<br />

<strong>and</strong> homogenized in a custom glass grinder. Free sporozoites were counted in a<br />

Bürker-Türk counting chamber using phase-contrast microscopy. PfSpz were<br />

cryopreserved at 16x10 6 /ml in 15% glycerol/PBS in aliquots for use in individual<br />

stimulation assays. Sporozoites that had undergone one freeze-thaw cycle were<br />

morphologically intact, but no longer able to glide (assay described in [16], using<br />

a FITC-3SP2 conjugated antibody). Salivary gl<strong>and</strong>s from an equal number of<br />

uninfected mosquitoes were used as a background control.<br />

NF54 strain P. falciparum asexual blood-stage parasites (PfRBC), regularly<br />

screened for mycoplasma contamination, were grown in RPMI-1640 medium<br />

containing 10% human A+ serum at 5% haematocrit in a semi-automated<br />

suspension culture system, in the absence of antibiotics <strong>and</strong> in an atmosphere<br />

containing 3% CO2 <strong>and</strong> 4% O2. For in vitro stimulation experiments,<br />

asynchronous asexual-stage cultures of NF54 strain parasites were harvested at<br />

a parasitemia of approximately 5-10% <strong>and</strong> mature asexual stages purified by<br />

centrifugation on a 27% <strong>and</strong> 63% Percoll density gradient [17]. This purification<br />

step results in preparations of 80-90% parasitemia, consisting of more than 95%<br />

schizonts/mature trophozoites. PfRBC were washed twice in PBS <strong>and</strong><br />

cryopreserved at 150x10 6 /mL in 15% glycerol/PBS in aliquots for use in<br />

individual stimulation assays. Mock-cultured uninfected erythrocytes (uRBC)<br />

were obtained similarly <strong>and</strong> served as control.<br />

Stimulation assay <strong>and</strong> staining for flow cytometry<br />

After thawing, cells were stimulated with either cryopreserved NF54 PfRBC (as<br />

previously described [9]) at a final concentration of 5*10 6 /ml or cryopreserved<br />

PfSpz at a concentration of 4*10 5 /ml for 24 hours. Uninfected red blood cells<br />

(uRBC, 5*10 6 /ml) <strong>and</strong> uninfected mosquito salivary gl<strong>and</strong>s (MSG) preparations<br />

dissected from equal numbers of uninfected mosquitoes, respectively, were<br />

used as negative controls. For cell stimulations with sporozoites/MSG 0.8 µl/ml<br />

CD28/CD49d reagent (BD) was added to the culture as a co-stimulant.<br />

PMA/ionomycin stimulation (50ng/ml <strong>and</strong> 1µg/ml respectively, Sigma-Aldrich)<br />

was used as positive control <strong>and</strong> added four hours before harvest. Brefeldin A<br />

(final concentration 10μg/mL) was added to all wells four hours prior to harvest.<br />

For staining procedures, PBMC were transferred to a 96 V-wells micro-titre plate<br />

(500,000 cells/well), washed <strong>and</strong> incubated with LIVE/DEAD® cell stain kit Aqua<br />

(Invitrogen, Carlsbad, CA, USA) for 30 min at 4°C. Cells were washed in FACS<br />

buffer (PBS containing 0.5% Bovine Serum Albumin, Sigma-Aldrich Co.), <strong>and</strong>


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 201<br />

Figure 2. Trial profile. Timelines of previous immunisation study in grey.<br />

stained with IQTest CD45RO-phycoerythrin Texas Red (Beckman-Coulter), PerCP<br />

anti-human CD3 (BioLegend, San Diego, CA, USA) <strong>and</strong> PE-Cy7 anti-human CD62L<br />

(eBioscience) for 20 min at 4°C. After washing, cells were incubated for 15 min at<br />

4°C with fixation Medium A (Caltag, S. San Francisco, CA, USA), washed <strong>and</strong><br />

stained with FITC anti-human IFNγ (eBioscience) <strong>and</strong> APC anti-human IL-2<br />

(eBioscience) in permeabilization Medium B (Caltag) for 20 min at 4°C. 100,000<br />

events in the lymphocyte gate, based on forward- <strong>and</strong> side-scatter, were<br />

acquired on a CyAn ADP 9-color flow cytometer (Beckman-Coulter). Flow<br />

cytometry analysis was performed using FlowJo software version 9.1. Analysis<br />

was performed on CD3+ T cells <strong>and</strong> effector memory (EM) T cells. The IFNγ <strong>and</strong><br />

IL-2 gates were set based on negative control samples.<br />

Statistical analysis<br />

Data analysis was performed using SPSS software version 16.0. Differences in<br />

cellular immune responses between subjects in the vaccine group <strong>and</strong> control<br />

group were analyzed by the Mann–Whitney-U test. The correlation between<br />

peak temperature <strong>and</strong> peak d-dimer values was assessed by Pearson correlation<br />

analysis. A two-sided P value of less than 0.05 was considered to indicate<br />

statistical significance.


202 Chapter 10<br />

Figure 3. Mean number of adverse events per volunteer. Data from controls in black<br />

(n=5), volunteers that showed delayed patency in red (n=2) <strong>and</strong> protected volunteers in<br />

green (n=4). Only adverse events possibly or probably related to malaria are included.<br />

Results<br />

All ten volunteers previously immunised <strong>and</strong> protected <strong>against</strong> an experimental<br />

malaria <strong>infection</strong>, were contacted 32 months after the first immunisation [9]. Six<br />

of these volunteers passed screening for eligibility <strong>and</strong> were included in the<br />

current follow-up study. Three out of four excluded volunteers were unavailable<br />

<strong>and</strong> one had to be excluded because of a family history of myocardial infarction<br />

(second degree relative under 55 years of age). Five healthy volunteers were<br />

newly recruited as a control group. Figure 2 shows the trial profile. Eleven<br />

volunteers were challenged by the bites of five Pf-infected mosquitoes <strong>and</strong> all<br />

completed follow-up. The average age of participating volunteers was 23.6 years<br />

(range 21-28 years old), of whom four were male. Four controls <strong>and</strong> one<br />

immunised volunteer had previously (range 6 months to 9 years) travelled in<br />

malaria endemic areas during which time they all used malaria prophylaxis, none<br />

of the volunteers was a previous resident of a malaria endemic area. None of<br />

the volunteers reported having used antimalarials within the past 6 months <strong>and</strong><br />

none showed positive serology for Pf.<br />

When re-challenged according to the st<strong>and</strong>ard protocol by bites of five infected<br />

mosquitoes, four out of six of the immunised volunteers never developed bloodstage<br />

parasitemia by microscopy during daily follow-up. One of these volunteers<br />

requested presumptive treatment with anti-malarials on day 14 post-<strong>infection</strong><br />

for reasons unrelated to the trial. The remaining two volunteers showed a<br />

markedly delayed patent parasitemia on days 15 <strong>and</strong> 18, respectively.


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 203<br />

Any Adverse<br />

event frequency<br />

Protected (n=4) Delayed patency (n=2) Controls (n=5)<br />

Mean<br />

duration ±<br />

SD (days) frequency<br />

Mean<br />

duration ±<br />

SD (days) frequency<br />

Mean<br />

duration ±<br />

SD (days)<br />

Abdominal pain 1 1.0±0<br />

Arthralgia 2 1.8±0<br />

Chills 2 0.8±0.6<br />

Fatigue 3 5.0±5.3 2 5.6±4.1 5 5.3±3.1<br />

Fever 1 0.1±0 2 2.0±0.5 2 1.3±1.0<br />

Headache 3 0.7±0.3 2 1.6±1.5 3 1.7±2.0<br />

Itching 2 1.5±2.0 2 3.6±3.6 3 3.4±2.4<br />

Malaise 2 3.2±2.1<br />

Myalgia 1 1.0±0 1 3.0±0 3 1.9±1.9<br />

Nausea 2 0.1±0.09 1 3.0±0 2 0.5±0.5<br />

Vomiting 1 0.04±0<br />

Any 4 1.8±3.1 2 2.7±2.8 5 5.7±2.2<br />

Grade 3 adverse event<br />

Fatigue 2 9.0±1.8 1 7.9±0<br />

Fever 2 1.8±0.6<br />

Itching 1 0.1±0<br />

Malaise 2 3.2±2.1<br />

Any 1 0.1±0 2 9.0±1.8 2 8.4±2.7<br />

Table 2. Number of individuals reporting possibly or probably related solicited<br />

adverse events in immune volunteers, volunteers with delayed patency <strong>and</strong> control<br />

volunteers. Unrelated adverse events were myalgia after sports, headache related to<br />

exams, common cold, a possible vasovagal reaction without collapse <strong>and</strong> a fracture of<br />

the scaphoid bone.<br />

The four protected volunteers reported several mild to moderate adverse<br />

events, of which short episodes of headache was the most common symptom<br />

(1-3 episodes per volunteer). Interestingly, one of these volunteers reported<br />

fever up to 38.0°C measured sub-lingual 19 days after the challenge <strong>infection</strong>.<br />

The two volunteers with delayed patency reported adverse events similar to<br />

those in the control group. The number of adverse events reported per<br />

volunteer over time is shown in Figure 3.<br />

Four of five control volunteers developed parasitemia detected by thick smear<br />

within the expected time frame of 7-12 days post-<strong>infection</strong> <strong>and</strong> were treated per<br />

protocol [9]. One control volunteer presumptively received anti-malarial<br />

treatment on day 9.3 in the absence of a parasite-positive blood-smear but<br />

based on clinical malaria symptoms <strong>and</strong> elevated d-dimer levels [1180 ng/ml), a<br />

pre-defined safety criterion. All control volunteers, including the latter,<br />

developed asexual blood-stage parasitemia detectable by Q-PCR retrospective-


204 Chapter 10<br />

Figure 4. Number of parasites as measured by Q-PCR in controls (A) <strong>and</strong> immunised<br />

volunteers (B). Parasitemia of control group (n=5) is shown as geometric mean <strong>and</strong><br />

95% confidence interval, immunised volunteers are plotted individually (n=6).<br />

Parasitemia of volunteers with delayed patency is indicated by dashed lines.<br />

ly, <strong>and</strong> reported adverse events compatible with clinical malaria. Fatigue <strong>and</strong><br />

headache were most commonly reported. An overview of all solicited possibly or<br />

probably related adverse events is provided in Table 2.<br />

For safety reasons related to a previously reported cardiac event [12], we<br />

measured biochemical cardiovascular indicators assiduously throughout the<br />

trial. Shortly after parasite detection by microscopy <strong>and</strong> initiation of antimalarial<br />

treatment, d-dimer levels became elevated in all volunteers. The maximum<br />

increase in d-dimer varied between 570 to 14,600 ng/ml, with a median peak of<br />

1,600 ng/ml (n=7). Von Willebr<strong>and</strong> cleaving protease activity was decreased at


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 205<br />

Figure 5. In vitro production of IFNγ (A,B) or both IFNγ <strong>and</strong> IL-2 (C-F) by T-cells (A-D) or<br />

CD45RO+ <strong>and</strong> CD62L- effector memory (EM) T cells (E,F) on the day before (re-)challenge,<br />

upon stimulation with sporozoites (PfSpz, panel A,C,E) or asexual stage parasites (PfRBC,<br />

panel B,D,F). Numbers of cytokine producing cells are depicted as percentage of total T<br />

cells (A-D) or EM T cells (E-F). Symbols indicate individual values from immunised (n=6)<br />

<strong>and</strong> control (n=5) volunteers; horizontal lines represent group medians. Fully protected<br />

immunised volunteers <strong>and</strong> control volunteers are indicated as circles, immunised<br />

volunteers with delayed patency are indicated as triangles.<br />

this time in two of five volunteers with d-dimer above 1,000 ng/ml (58% <strong>and</strong><br />

38% compared with 80% <strong>and</strong> 67% respectively in a resting state).<br />

Fragmentocytes were never detected. Peak d-dimer values correlated with peak


206 Chapter 10<br />

temperature (Pearson R=0.83, p=0.02). Platelets decreased slightly below<br />

120x10 9 /l in one volunteer (nadir 108x10 9 /l). Lactate dehydrogenase levels were<br />

never elevated above 1000 U/l. Highly sensitive troponin T was never abnormal<br />

(max 0.011 µg/l). None of the volunteers showed any bleeding or thrombotic<br />

complications. None of the protected volunteers showed an elevation of ddimers<br />

above detection level (250 ng/ml).<br />

Real-time quantitative PCR (Q-PCR) was performed on all collected samples<br />

retrospectively. Control volunteers showed cyclical parasite growth, identical to<br />

control volunteers in previous malaria challenge <strong>infection</strong> trials [9,14]. Asexual<br />

blood-stage parasites were never detected by Q-PCR (detection limit 20<br />

parasites/ml) in any of the four protected volunteers during the entire 21 day<br />

follow-up period. The remaining two immunised volunteers with delayed<br />

patency by microscopy also showed delayed appearance of blood-stage<br />

parasitemia by Q-PCR (Figure 4).<br />

Specific anti-parasite antibody responses were detectable at 28 months postimmunisation<br />

in only one of six immunised volunteers, i.e. <strong>against</strong> the major<br />

circumsporozoite antigen (NANP6 repeats) <strong>and</strong> crude blood-stage antigens (data<br />

not shown). Antibodies with specificity for individual asexual blood stage<br />

antigens (AMA-1 <strong>and</strong> GLURP) were not detectable. Blood stage parasitemia was<br />

accompanied by increased plasma concentrations of systemic inflammatory<br />

markers, including IFNγ (median peak 69 pg/ml, IQR 51-135), IL-6 (median peak<br />

9.2 pg/ml, IQR 6.4-14) <strong>and</strong> C-reactive protein (median peak 52 mg/l, IQR 26.5-<br />

57.5) with no difference between controls <strong>and</strong> volunteers with delayed patency.<br />

None of the protected volunteers showed any increase in plasma concentrations<br />

of IFNγ, IL-6 or CRP (max 5 pg/ml, 7 pg/ml <strong>and</strong> 2.5 mg/l respectively).<br />

In vitro stimulation assays showed a sustained Pf-specific T cell IFNγ re-call<br />

response to both pre-erythrocytic (sporozoite) <strong>and</strong> blood-stage parasites that<br />

persisted over years in all six immunised individuals (Figure 5). These responses<br />

remained significantly higher than those of control volunteers (p


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 207<br />

Discussion<br />

Here we report for the first time the persistence of immunity in human<br />

volunteers to re-<strong>infection</strong> with Plasmodium falciparum more than two years<br />

after previous exposure. Using a recently developed artificial immunisation<br />

protocol [9] we observed long lasting <strong>protection</strong> in four of six re-challenged<br />

volunteers <strong>and</strong> markedly delayed patency in the two remaining volunteers. In<br />

addition, we describe the maintenance of Pf-specific T cell IFNγ responses.<br />

The notion that immunity to malaria is short-lived derives primarily from<br />

anecdotal reports of returning semi-immune immigrants. A critical review of the<br />

literature, however, reveals that such migrants appear to retain some<br />

<strong>protection</strong>, albeit only from severe disease <strong>and</strong> death [19]. The induction <strong>and</strong><br />

persistence of clinical <strong>and</strong> parasitological immunity is more controversial, due to<br />

the great diversity in malaria transmission intensity, age, genetic background<br />

<strong>and</strong> study endpoints, complicating interpretation [4]. Protective immunity to<br />

clinical malaria can be acquired relatively rapidly [4], <strong>and</strong> in settings of epidemic<br />

malaria, some measure of clinical <strong>protection</strong> may be sustained over several<br />

years [20]. However, meta-analyses of intermittent preventive treatment studies<br />

in naturally exposed infants <strong>and</strong> children show renewed susceptibility following<br />

discontinuation of the intervention [21] <strong>and</strong> a historical review of patients<br />

treated for neurosyphilis by (repeated) artificial malaria <strong>infection</strong> revealed no<br />

sterile protective immunity <strong>against</strong> subsequent re-challenge [22]. Moreover,<br />

<strong>protection</strong> induced in volunteers by irradiated sporozoite inoculation lasted 42<br />

weeks in only a few subjects [5] <strong>and</strong> the <strong>protection</strong> induced by the most<br />

successful sub-unit vaccine to date, RTS,S, seems to wane [6]. In summary,<br />

naturally acquired immunity is far from optimal, but protective immunity to<br />

severe disease can be maintained [4,19,23].<br />

The long-term <strong>protection</strong> <strong>against</strong> malaria in this study is thus surprising when<br />

compared with naturally acquired immunity. Several factors may account for<br />

this discrepancy. Firstly, the same Pf strain was used for both the immunisation<br />

protocol <strong>and</strong> the experimental challenges. Well-described target antigens for<br />

protective immunity exhibit high rates of genetic variation, circumventing crossprotective<br />

immunity in the field [24]. Secondly, our immunisation protocol<br />

prevents high blood-stage parasitemia, whilst repeated parasitemia in endemic<br />

areas is thought to suppress the development of immunity [25]. Thirdly, we<br />

immunised adult individuals, whereas natural exposure is first encountered by<br />

the immature immune system of infants4 or even in utero [26]. Finally, the


208 Chapter 10<br />

immune modulating effects of chloroquine might have enhanced the<br />

development of protective immune responses during the immunisation process<br />

[27].<br />

While Pf-specific antibody responses were negligible <strong>and</strong> declining (data not<br />

shown), we found a sustained Pf-specific T cell IFNγ re-call response that<br />

persisted over years <strong>and</strong> paralleled the sustained <strong>protection</strong> in all six immunised<br />

volunteers. The two volunteers with delayed patency did not show a distinctly<br />

different T cell response as compared to the fully protected individuals.<br />

However, a three to six day delay of exponentially growing parasites equals one<br />

to three logs (e.g. >95%) reduction in parasite burden. The variation in the level<br />

of <strong>protection</strong> between the volunteers might thus be relatively small. A large<br />

body of evidence supports the view that IFNγ responses are protective <strong>against</strong> Pf<br />

<strong>infection</strong>, although naturally acquired T-cell responses to individual antigens<br />

wane after several years of non-exposure [28]. The functional importance of<br />

IFNγ–producing cells, however, still needs to be established in larger study<br />

groups.<br />

We detected cellular responses to both sporozoites <strong>and</strong> blood-stage parasites<br />

with remarkably similar dynamics, although responses to blood-stage parasites<br />

were consistently higher than to sporozoites. Whether such responses are<br />

directed <strong>against</strong> both pre-erythrocytic <strong>and</strong> blood-stage antigens or represent<br />

cross-reactivity, will require further detailed immunological analysis. In either<br />

case, the lack of blood-stage parasites in the fully protected volunteers seems to<br />

indicate a primarily pre-erythrocytic immunological effector mechanism.<br />

<strong>Experimental</strong> malaria <strong>infection</strong>s provide a unique opportunity to study<br />

<strong>protection</strong> <strong>against</strong> Pf malaria in a controlled setting [29]. This trial reconfirms a<br />

solid correlation between the appearance of clinical symptoms <strong>and</strong> the presence<br />

of microscopically detectable blood stage parasites, including in volunteers with<br />

delayed patency. Clinical symptoms <strong>and</strong> the pattern of parasitemia by Q-PCR are<br />

very consistent between <strong>and</strong> within study groups illustrating the power of this<br />

experimental <strong>infection</strong> model (Chapter 6, [9]). Protected volunteers also<br />

reported some adverse events, without objective signs of inflammation. Possibly<br />

these events reflect natural fluctuations of subjective health perception.<br />

The occurrence of a cardiac event following experimental challenge after a<br />

phase I malaria vaccine trial in the past has necessitated an increased vigilance<br />

<strong>and</strong> surveillance of cardiac <strong>and</strong> coagulation markers [12]. We found increased<br />

concentrations of d-dimer that paralleled the increases in inflammatory markers,


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 209<br />

but without overt clinical thrombotic microangiopathy or bleeding<br />

complications. In addition we report incidental decreases of von Willebr<strong>and</strong><br />

cleaving protease, possibly as a result of increased consumption of the protease<br />

[30]. This is consistent with previous data concerning von Willebr<strong>and</strong> factor<br />

activation in experimental malaria [13]. The clinical relevance of these findings<br />

<strong>and</strong> their relation with the cardiac event is as yet unclear.<br />

Our relatively simple immunisation protocol represents a blueprint for induction<br />

of sustained anti-malarial immunity, providing a novel tool for exploring<br />

mechanisms of immunity <strong>and</strong> highlighting new research priorities. Precedence<br />

must be given to basic underst<strong>and</strong>ing of mechanisms of <strong>protection</strong> in this study<br />

<strong>and</strong> those factors which inhibit <strong>protection</strong> in naturally-exposed populations.<br />

Furthermore, strain specificity can be investigated with the use of heterologous<br />

challenge <strong>infection</strong>s <strong>and</strong> in field studies. Given the efficacy of our immunisation<br />

protocol, however, equal emphasis on a pragmatic parallel approach to develop<br />

an implementable whole-organism-based vaccine is justified.<br />

In conclusion, the irrefutable evidence that long-term immunity <strong>against</strong> malaria<br />

is possible holds a promise for an urgently needed malaria vaccine.<br />

Acknowledgements<br />

This trial was financed by a grant from the Dioraphte foundation. ACT is funded<br />

by the European Malaria Vaccine Development Association, MBBM was<br />

supported by an European Union FP6 Network of Excellence (BioMalPar)<br />

fellowship, KNM was funded by a Mozaiek grant from the Netherl<strong>and</strong>s<br />

Organisation for Scientific Research. We thank the trial volunteers <strong>and</strong> the staff<br />

from the Clinical Research Centre Nijmegen who made this study possible. We<br />

thank the following individuals for their assistance during the trial: Laura Pelser,<br />

Jol<strong>and</strong>a Klaassen, Astrid Pouwelsen, Jacqueline Kuhnen for the mosquito<br />

<strong>infection</strong> <strong>and</strong> dissection work, Wendy Arts, Nanny Huiberts, Chantal Siebes,<br />

Marlou Kooreman, Paul Daemen <strong>and</strong> Ella Driessen for reading many thick<br />

smears. We would also like to acknowledge the contributions of Alex<strong>and</strong>er<br />

Rennings for his help with monitoring volunteer safety <strong>and</strong> the cardiologists<br />

Gheorghe Pop <strong>and</strong> Marc Brouwer for their dedication to the cardiac monitoring<br />

of the trial volunteers. We acknowledge the work of Richard Huijbens for the


210 Chapter 10<br />

cytokine measurements by Luminex. We would like to thank Rob Woestenenk<br />

for use <strong>and</strong> support of the flowcytometer.<br />

Funding<br />

This trial was funded by the Dioraphte foundation.


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 211<br />

References<br />

1. Gallup JL, Sachs JD. The economic burden of malaria. Am J Trop Med Hyg 2001;<br />

64: 85-96.<br />

2. Morel CM, Lauer JA, Evans DB. Cost effectiveness analysis of strategies to<br />

combat malaria in developing countries. BMJ 2005; 331: 1299.<br />

3. Achtman AH, Bull PC, Stephens R, Langhorne J. Longevity of the immune<br />

response <strong>and</strong> memory to blood-stage malaria <strong>infection</strong>. Curr Top Microbiol<br />

Immunol 2005; 297: 71-102.<br />

4. Doolan DL, Dobano C, Baird JK. Acquired immunity to malaria. Clin Microbiol<br />

Rev 2009; 22: 13-36, Table.<br />

5. Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP, Doolan DL et al. Protection of<br />

humans <strong>against</strong> malaria by immunization with radiation-attenuated<br />

Plasmodium falciparum sporozoites. J Infect Dis 2002; 185: 1155-64.<br />

6. Crompton PD, Pierce SK, Miller LH. Advances <strong>and</strong> challenges in malaria vaccine<br />

development. J Clin Invest 2010; 120: 4168-78.<br />

7. Olotu A, Lusingu J, Leach A, Lievens M, Vekemans J, Msham S et al. Efficacy of<br />

RTS,S/AS01E malaria vaccine <strong>and</strong> exploratory analysis on anti-circumsporozoite<br />

antibody titres <strong>and</strong> <strong>protection</strong> in children aged 5-17 months in Kenya <strong>and</strong><br />

Tanzania: a r<strong>and</strong>omised controlled trial. Lancet Infect Dis 2011; 11: 102-9.<br />

8. Sacarlal J, Aide P, Aponte JJ, Renom M, Leach A, M<strong>and</strong>om<strong>and</strong>o I et al. Long-term<br />

safety <strong>and</strong> efficacy of the RTS,S/AS02A malaria vaccine in Mozambican children.<br />

J Infect Dis 2009; 200: 329-36.<br />

9. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, van Gemert GJ et al.<br />

Protection <strong>against</strong> a malaria challenge by sporozoite inoculation. N Engl J Med<br />

2009; 361: 468-77.<br />

10. Bousema JT, Roeffen W, van der KM, de Vlas SJ, van d, V, Bangs MJ et al. Rapid<br />

onset of transmission-reducing antibodies in javanese migrants exposed to<br />

malaria in Papua, Indonesia. Am J Trop Med Hyg 2006; 74: 425-31.<br />

11. Ponnudurai T, Lensen AH, van Gemert GJ, Bensink MP, Bolmer M, Meuwissen<br />

JH. Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes.<br />

Parasitology 1989; 98 Pt 2: 165-73.<br />

12. Nieman AE, de MQ, Roestenberg M, Wiersma J, Pop G, Stalenhoef A et al.<br />

Cardiac complication after experimental human malaria <strong>infection</strong>: a case report.<br />

Malar J 2009; 8: 277.<br />

13. de Mast Q, Groot E, Lenting PJ, de Groot PG, McCall M, Sauerwein RW et al.<br />

Thrombocytopenia <strong>and</strong> release of activated von Willebr<strong>and</strong> Factor during early<br />

Plasmodium falciparum malaria. J Infect Dis 2007; 196: 622-8.


212 Chapter 10<br />

14. Hermsen CC, Telgt DS, Linders EH, van de Locht LA, Eling WM, Mensink EJ et al.<br />

Detection of Plasmodium falciparum malaria parasites in vivo by real-time<br />

quantitative PCR. Mol Biochem Parasitol 2001; 118: 247-51.<br />

15. Ponnudurai T, Lensen AH, van Gemert GJ, Bensink MP, Bolmer M, Meuwissen<br />

JH. Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes.<br />

Parasitology 1989; 98 Pt 2: 165-73.<br />

16. Stewart MJ, V<strong>and</strong>erberg JP. Malaria sporozoites leave behind trails of<br />

circumsporozoite protein during gliding motility. J Protozool 1988; 35: 389-93.<br />

17. Rivadeneira EM, Wasserman M, Espinal CT. Separation <strong>and</strong> concentration of<br />

schizonts of Plasmodium falciparum by Percoll gradients. J Protozool 1983; 30:<br />

367-70.<br />

18. Wenink MH, Santegoets KC, Broen JC, van BL, bdollahi-Roodsaz S, Popa C et al.<br />

TLR2 promotes Th2/Th17 responses via TLR4 <strong>and</strong> TLR7/8 by abrogating the type<br />

I IFN amplification loop. J Immunol 2009; 183: 6960-70.<br />

19. Struik SS, Riley EM. Does malaria suffer from lack of memory? Immunol Rev<br />

2004; 201: 268-90.<br />

20. Deloron P, Chougnet C. Is immunity to malaria really short-lived? Parasitol<br />

Today 1992; 8: 375-8.<br />

21. Aponte JJ, Schellenberg D, Egan A, Breckenridge A, Carneiro I, Critchley J et al.<br />

Efficacy <strong>and</strong> safety of intermittent preventive treatment with sulfadoxinepyrimethamine<br />

for malaria in African infants: a pooled analysis of six<br />

r<strong>and</strong>omised, placebo-controlled trials. Lancet 2009; 374: 1533-42.<br />

22. Jeffery GM. Epidemiological significance of repeated <strong>infection</strong>s with<br />

homologous <strong>and</strong> heterologous strains <strong>and</strong> species of Plasmodium. Bull World<br />

Health Organ 1966; 35: 873-82.<br />

23. Langhorne J, Ndungu FM, Sponaas AM, Marsh K. Immunity to malaria: more<br />

questions than answers. Nat Immunol 2008; 9: 725-32.<br />

24. Kidgell C, Volkman SK, Daily J, Borevitz JO, Plouffe D, Zhou Y et al. A systematic<br />

map of genetic variation in Plasmodium falciparum. PLoS Pathog 2006; 2: e57.<br />

25. Bejon P, Mwacharo J, Kai O, Todryk S, Keating S, Lowe B et al. The induction <strong>and</strong><br />

persistence of T cell IFN-gamma responses after vaccination or natural exposure<br />

is suppressed by Plasmodium falciparum. J Immunol 2007; 179: 4193-201.<br />

26. Broen K, Brustoski K, Engelmann I, Luty AJ. Placental Plasmodium falciparum<br />

<strong>infection</strong>: causes <strong>and</strong> consequences of in utero sensitization to parasite<br />

antigens. Mol Biochem Parasitol 2007; 151: 1-8.<br />

27. Sauerwein RW, Bijker EM, Richie TL. Empowering malaria vaccination by drug<br />

administration. Curr Opin Immunol 2010; 22: 367-73.


Long-term <strong>protection</strong> <strong>against</strong> malaria after experimental sporozoite inoculation 213<br />

28. McCall MB, Sauerwein RW. Interferon-gamma--central mediator of protective<br />

immune responses <strong>against</strong> the pre-erythrocytic <strong>and</strong> blood stage of malaria. J<br />

Leukoc Biol 2010; 88: 1131-43.<br />

29. Sauerwein RW, Roestenberg M, Moorthy VS. <strong>Experimental</strong> human challenge<br />

<strong>infection</strong>s can accelerate clinical malaria vaccine development. Nat Rev<br />

Immunol 2011; 11: 57-64.<br />

30. Claus RA, Bockmeyer CL, Sossdorf M, Losche W. The balance between von-<br />

Willebr<strong>and</strong> factor <strong>and</strong> its cleaving protease ADAMTS13: biomarker in systemic<br />

inflammation <strong>and</strong> development of organ failure? Curr Mol Med 2010; 10: 236-<br />

48.


Chapter 11<br />

General Discussion


216 Chapter 11<br />

The development of a highly efficacious malaria vaccine faces many hurdles, key<br />

to which is our limited knowledge of what exactly constitutes <strong>protection</strong> <strong>against</strong><br />

Plasmodium falciparum malaria in humans. Malaria vaccine development relies<br />

on a trial-<strong>and</strong>-error approach, with many c<strong>and</strong>idates in (pre-)clinical<br />

development awaiting efficacy testing in the field. This thesis focuses on the<br />

development of research tools that facilitate immunological <strong>and</strong> vaccine<br />

research into human <strong>protection</strong> <strong>against</strong> Plasmodium falciparum malaria in early<br />

stages of clinical development. We used the following approaches:<br />

Firstly, we tested an asexual erythrocytic stage subunit vaccine c<strong>and</strong>idate (Apical<br />

Membrane Antigen 1) for its safety <strong>and</strong> immunogenicity in humans <strong>and</strong><br />

performed in depth analysis of the humoral immune response following<br />

immunization.<br />

Secondly, we assessed <strong>and</strong> optimized an important tool for malaria vaccine <strong>and</strong><br />

immunological research: controlled human malaria <strong>infection</strong>s. In these trials,<br />

healthy malaria-naïve volunteers are exposed to infectious mosquito bites in<br />

order to test the preliminary efficacy of vaccine c<strong>and</strong>idates or in order to<br />

perform immunological research.<br />

Thirdly, we developed a human model for <strong>protection</strong> <strong>against</strong> Plasmodium<br />

falciparum malaria to facilitate immunological research <strong>and</strong> facilitate the<br />

development of a whole-sporozoite vaccine for malaria.<br />

Apical Membrane Antigen 1 (AMA1)<br />

AMA1 is considered a promising vaccine c<strong>and</strong>idate, primarily due to a large body<br />

of data from animal studies indicating that AMA1 can induce immune responses<br />

capable of inhibiting the blood stage parasite multiplication in vitro <strong>and</strong> in vivo<br />

(reviewed in [1]). Specifically, <strong>protection</strong> can be conveyed by transferring anti-<br />

AMA1 antibodies [2-5] <strong>and</strong> epidemiological studies in malaria endemic areas<br />

show that the presence of AMA1 antibodies correlate with malaria <strong>protection</strong> [6,<br />

7]. Subsequently, AMA1 has been produced as recombinant protein both in<br />

Pichia pastoris <strong>and</strong> E. Coli. Animal studies showed satisfactory pharmacotoxicity,<br />

so the whole-length (amino acid 25 to 545) Pichia pastoris-expressed AMA1<br />

from the FVO strain has been taken forward into the first phase of clinical<br />

testing. We showed that PfAMA1[25-545] FVO can be safely administered to<br />

humans <strong>and</strong> is immunogenic (Chapter 2). We also showed that both<br />

reactogenicity <strong>and</strong> immunogenicity are dependent on the adjuvant, with the


General Discussion 217<br />

occurrence of unwanted side-effects such as an abscess at the injection site in<br />

the higher dose Montanide ISA 720 group (Chapter 2). Moreover, we confirmed<br />

that the antibodies induced by the AMA1 vaccine have the capacity to inhibit the<br />

invasion of parasites into erythrocytes in vitro (Chapter 2, 3). Similar results were<br />

obtained with other AMA1 vaccine formulations [8-15].<br />

Following these Phase I safety trials, several attempts were undertaken to test<br />

the efficacy of AMA1 formulations in the field (Phase IIb). Unfortunately, two<br />

separate field studies could not establish a significant effect of AMA1<br />

immunisation on primary parasitological endpoints [9, 16], although there was<br />

some evidence for homologous <strong>protection</strong>. There are two possible explanations<br />

for the apparent discrepancy between in vitro data, rodent <strong>and</strong> simian studies<br />

<strong>and</strong> in vivo human efficacy data: 1) allelic diversity or 2) insufficient immune<br />

responses.<br />

Ad 1). Allelic diversity may account for the failing efficacy of AMA1 in the field,<br />

as AMA1 is known to be highly polymorphic [17]. We showed that these allelic<br />

polymorphisms lead to reduced growth inhibition of antibodies measured<br />

<strong>against</strong> parasite strains expressing heterologous AMA1 in vitro (Chapter 3), but<br />

the translation of these in vitro effects to in vivo reduced efficacy was not found<br />

in controlled <strong>infection</strong> studies [18]. Detailed analysis of a field trial revealed<br />

specific efficacy <strong>against</strong> clinical episodes caused by parasites that expressed a<br />

form of AMA1 with the same residues as the vaccine strain (3D7) in the<br />

dominant cluster of polymorphisms in domain I [16]. However, the other field<br />

trial found no evidence of vaccine selection or strain-specific efficacy of a<br />

bivalent AMA1 vaccine [9], suggesting that the genetic diversity of AMA1 may<br />

not fully account for failure of the vaccine to provide <strong>protection</strong> [19]. Also blood<br />

stage <strong>infection</strong> of vaccines in a Phase I/IIa trial with homologous strain Pf did not<br />

show reduced parasite multiplication rates, indicating that failing efficacy could<br />

not be attributed to strain diversity [18]. In conclusion, the extent to which<br />

allelic diversity accounts for reduced efficacy of c<strong>and</strong>idate AMA1 vaccines is<br />

debated, but the importance of inducing strain transcending immunity has been<br />

well-recognised <strong>and</strong> efforts into the development of multivalent AMA1 vaccines<br />

are increasing [20, 21].<br />

Ad 2). A second possible explanation for failing efficacy of AMA1 vaccines is the<br />

quality of the immune response. Many AMA1 vaccines induce significant<br />

concentrations of antibodies but no <strong>protection</strong>, therefore antibody<br />

concentration alone does not suffice to protect. We explored the avidity of


218 Chapter 11<br />

antibodies induced by AMA1 in humans <strong>and</strong> rabbits <strong>and</strong> found that the avidity<br />

<strong>and</strong> concentration of AMA1-induced antibodies are linked by a negative<br />

feedback mechanism reaching saturation in most vaccinees (Chapter 4). Higher<br />

titre AMA1 antibodies may thus not necessarily implicate better binding<br />

efficiency <strong>and</strong> function (Chapter 4). The relevance of these findings for in vivo<br />

<strong>protection</strong> is unclear. A correlation between antibody avidity, as measured by<br />

thiocyanate ELISA, <strong>and</strong> <strong>protection</strong> was not found in a P. chabaudi rodent model,<br />

but the combination of titres to refolded <strong>and</strong> reduced/alkylated recombinant<br />

AMA1 proved an important predictor of <strong>protection</strong> [22]. These results illustrate<br />

the importance of underst<strong>and</strong>ing the nature of AMA1 immunogenic epitopes,<br />

their functionality <strong>and</strong> the antibody characteristics. Moreover, other Pf-specific<br />

antibodies may interfere with the biological activity of anti-AMA1 antibodies<br />

[23], underlining the fact that <strong>protection</strong> in the field is a composite of more than<br />

just anti-AMA1 antibodies. An immunological marker for <strong>protection</strong> is essential<br />

to assess immunogenicity of AMA1 vaccine c<strong>and</strong>idates in an early stage of<br />

clinical development.<br />

The in vitro growth inhibition assay (GIA) has been developed as a functional<br />

marker that include all antibody properties. The GIA is a biological assay<br />

investigating the capacity of AMA-1 induced antibodies to inhibit parasite<br />

growth [24]. Although promising at first, the inhibitory capacity measured in<br />

vitro did not correlate with <strong>protection</strong> in rhesus monkeys [25] or in human trials<br />

[18, 26]. The fact that we found substantial inhibition of AMA1-induced<br />

antibodies in vitro, is thus no guarantee of <strong>protection</strong> in vivo.<br />

Nonetheless, <strong>protection</strong> from Plasmodium falciparum malaria can be transferred<br />

from one human to the other by the passive transfer of antibodies from malariaimmune<br />

to non-immune subjects. Since 1917, such experiments were<br />

performed in humans, when it was discovered that patients improved clinically<br />

when inoculated with serum obtained from “chronic” cases. In the 60’s<br />

experiments were repeated <strong>and</strong> antibodies taken from children in West Africa<br />

proved sufficient for the treatment of children in East Africa [27], indicating that<br />

antibodies can even overcome strain diversity. An antibody dependent cellular<br />

inhibiting effect (ADCI) has been proposed as an effector mechanism to this<br />

<strong>protection</strong> [28], relying on the help of cytokines such as IFNγ, IL-4 [28] <strong>and</strong> TNFα<br />

[29] to upregulate phagocytic function of polymorphonuclear leukocytes or<br />

monocytes. However, T-cells alone have also shown to adoptively transfer<br />

<strong>protection</strong> <strong>against</strong> malaria in rodent models [30] <strong>and</strong> the central role of IFNγ in


General Discussion 219<br />

mediating this <strong>protection</strong> is increasingly appreciated [31]. Indeed, also in<br />

humans, repeated low grade blood stage <strong>infection</strong>s can induce <strong>protection</strong><br />

<strong>against</strong> Pf malaria in the absence of detectable antibodies [32]. In addition,<br />

multifunctional T-cell responses were identified following human AMA1<br />

vaccination [33] <strong>and</strong> T-cell memory responses to T-cell epitopes from the AMA1<br />

protein were detected in clinically immune subjects, although no clear-cut<br />

association with parasitemia was found [34, 35]. To conclude, AMA1-specific Tcells<br />

contribute to building an effective humoral response in mice [36-38], but<br />

antibodies are generally still thought to be the most critical effector for blood<br />

stage parasite inhibition [38, 39]. Apparently, not just the quantity of antibodies<br />

but possibly also the quality <strong>and</strong> the interplay between cellular <strong>and</strong> humoral<br />

responses play a role in building a protective immune response to AMA1.<br />

In order to underst<strong>and</strong> the mechanism by which AMA1 directed immune<br />

responses effectuate parasite growth inhibition, more basic underst<strong>and</strong>ing of<br />

the function of AMA1 is essential. AMA1 is expressed in merozoites <strong>and</strong> is<br />

thought to help reorienting the merozoite when aligning with the erythrocyte<br />

membrane <strong>and</strong> subsequently attaching with erythrocyte membrane proteins [1].<br />

More recent mechanistic in vitro studies, however, also highlight a possible role<br />

of AMA1 in parasite invasion into hepatocytes, showing expression of AMA1 in<br />

sporozoites <strong>and</strong> inhibition of AMA1 antibodies with invasion of hepatocytes [40].<br />

The possible clinical relevance of this function was confirmed by two controlled<br />

malaria <strong>infection</strong> (CHMI) studies with human volunteers [18, 26], in which no<br />

clinically relevant effect on prepatency or blood stage parasite inhibition could<br />

be found in AMA1 immunized volunteers challenged by mosquito bite [26] or<br />

blood stage parasites[18], but possible reduction of liver load parasites could be<br />

detected by Q-PCR analysis of parasitemia in the former trial [26].<br />

The versatility of AMA1 expression complicates the dissection of AMA1-induced<br />

inhibiting <strong>and</strong> enhancing immune responses. The induction of exactly the right<br />

quality <strong>and</strong> quantity immune responses, however, may be the key to AMA1<br />

success in humans. Adjuvants are a very heterogenic group of pharmacological<br />

or immunological agents that are co-administered with vaccines in order to<br />

potentiate <strong>and</strong>/or direct immune responses in humans. Particularly in malaria<br />

research adjuvants have proven to play a critical role: GlaxoSmithKline’s<br />

proprietary adjuvant AS01/2 was found crucial for enhancing responses to RTS,S<br />

to protective levels [41]. We found that adjuvants are also important<br />

determinants of cellular <strong>and</strong> humoral immune responses when combined with


220 Chapter 11<br />

the PfAMA1[25-545] FVO vaccine (Chapters 2, 3). Our work on AMA1 antibody<br />

avidity illustrates that the AS02A adjuvant directs the balance of antibody<br />

quantity <strong>and</strong> quality towards high titre antibody of lower avidity when compared<br />

to a conventional adjuvant such as Alhydrogel (Chapter 4).<br />

Recent advances in adjuvant research have led to the availability of many new<br />

adjuvants [42]. With the increased underst<strong>and</strong>ing of host/pathogen interactions<br />

new classes of adjuvants follow more rational design, such as Toll-like receptor<br />

agonists or cytokines. In addition, delivery platforms have been developed to<br />

ensure a specific presentation of the antigen to the immune system. For<br />

example, viral platforms are designed to direct the immune response from a<br />

classical humoral response following protein immunization to CD4+ or CD8+<br />

responses induced by, for example, adenovirus, fowlpox or modified vaccinia<br />

viruses [43]. Also bacterium-like particles or nanoparticles have shown to<br />

provide benefit as carriers to potential malaria vaccines [44, 45]. Heterologous<br />

prime-boost strategies utilizing two different adjuvants with one antigen is<br />

another recent development that has shown to increase efficacy [46]. In<br />

addition to adjuvants <strong>and</strong> delivery platforms, which are co-administrated with<br />

the vaccine <strong>and</strong> designed to boost or direct the immune response, research has<br />

focussed on the development of especially designed delivery systems, aimed at<br />

the specific delivery of an antigen at a certain anatomical site. Nasal delivery,<br />

needle free jet devices <strong>and</strong> intrabuccal delivery systems are examples of the<br />

most recent developments (for example [47, 48]). Their specific advantages are<br />

yet to be investigated.<br />

Also AMA1 has been subjected to the addition of new adjuvants in an attempt to<br />

increase immunogenicity [49]. Thorough adjuvant research will be required in<br />

order to delineate the properties <strong>and</strong> optimal combination of antigen, adjuvant,<br />

delivery platform <strong>and</strong> delivery system, a complex task requiring huge effort <strong>and</strong><br />

funds [42]. Direct comparisons of different adjuvants with one antigen, as we<br />

have described (Chapter 2), will prove to be instrumental to these<br />

developments. Unfortunately, the limited availability of newly developed<br />

adjuvants in the public domain has restricted the number of comparative trials<br />

so far.<br />

In conclusion, in vitro <strong>and</strong> animal studies have raised high hopes for AMA1<br />

vaccines, which could not be met in human efficacy trials. In addition, the phase<br />

III clinical development of RTS,S has set high st<strong>and</strong>ards for the development of<br />

any malaria vaccine, a challenge that also AMA1 has to face. The versatility of


General Discussion 221<br />

AMA1 expression <strong>and</strong> immune responses complicate the development of an<br />

AMA1-induced correlate of <strong>protection</strong>, but may also hold the biggest promise<br />

for AMA1 vaccines, being able to induce both pre-erythrocytic <strong>and</strong> erythrocytic<br />

immunity. Possibly, the development of new adjuvants, delivery systems, strain<br />

covering approaches or the combination of AMA1 with other antigens will<br />

provide a means by which animal results can be translated into human efficacy.<br />

The availability of an AMA1 induced correlate of <strong>protection</strong> will most certainly<br />

reduce costs <strong>and</strong> accelerate the development of any AMA1 vaccine. A proof-ofprinciple<br />

human trial may thus be necessary to supply data for immunological<br />

research <strong>and</strong> provide a new basis for rejuvenated preclinical research for AMA1.<br />

Meanwhile, the characterisation of adjuvants <strong>and</strong> other malaria antigens<br />

facilitate the rational design of the most effective AMA1-based (combination-)<br />

vaccine.<br />

Controlled human malaria <strong>infection</strong>s<br />

Adapted from Sauerwein et al [50].<br />

Malaria vaccine c<strong>and</strong>idate AMA1 illustrates the relevance of controlled malaria<br />

<strong>infection</strong> trials (CHMI) in acquiring novel insights on the mechanism by which a<br />

c<strong>and</strong>idate vaccine can induce <strong>protection</strong>. A major strength of controlled malaria<br />

<strong>infection</strong>s is the use of infectious mosquitoes, mimicking the natural route of<br />

<strong>infection</strong>. Moreover, these <strong>infection</strong>s are carried out in a controlled<br />

environment, allowing detailed evaluation of parasite growth <strong>and</strong> immunological<br />

determinants, which make them suitable to investigate the efficacy of<br />

vaccine c<strong>and</strong>idates <strong>and</strong> mechanisms of <strong>protection</strong>. The induction of immunity by<br />

exposure of malaria-naive volunteers to infectious mosquito bites while using<br />

chloroquine prophylaxis (Chapter 9) is one example. More basic research into<br />

immunological mechanisms following Pf <strong>infection</strong> in human is another [51]. The<br />

detailed analysis of parasitemia following <strong>infection</strong> may add to the<br />

underst<strong>and</strong>ing of immunological inhibiting effects in these trials.<br />

A real-time quantitative PCR (qPCR) assay based on 18S ribosomal RNA gene<br />

transcripts has been developed for tracking the kinetics of developing<br />

parasitemia before a positive diagnosis of <strong>infection</strong> can be made from a thick<br />

blood smear using microscopy [52]. This assay is becoming increasingly<br />

important for assessing very low parasite densities <strong>and</strong> incremental changes in<br />

density in small scale Phase IIa trials [53]. The detection of parasites below<br />

microscopy thresholds by qPCR allows for a detailed analysis of cyclical parasite


222 Chapter 11<br />

growth in the blood, albeit for a short time window of 2–3 days between liverstage<br />

<strong>infection</strong> <strong>and</strong> microscopic detection [52]. We have now formally shown<br />

that these molecular techniques enhance the power of controlled human<br />

<strong>infection</strong> trials to detect modest vaccine efficacy (which may not necessarily<br />

correspond with clinical <strong>protection</strong>) with only small numbers of volunteers<br />

(seven per group) (Chapter 6). Statistical models can be applied to further<br />

improve the discriminative power between control <strong>and</strong> test groups as well as to<br />

provide biological information about the parasite life cycle (including the<br />

duration of liver-stage maturation, number of infected hepatocytes, duration of<br />

blood-stage trophozoite maturation <strong>and</strong> multiplication rates [54-56]).<br />

Immediate treatment of volunteers at the earliest phase of microscopically<br />

detectable blood-stage <strong>infection</strong> ensures that the potential risks of<br />

complications associated with severe malaria are minimized to the greatest<br />

extent possible. Indeed, controlled human malaria <strong>infection</strong>s have shown to be<br />

safe in the 1,343 volunteers challenged so far [57-59]. Recently, safety concerns<br />

were raised because of a cardiac event in a young volunteer shortly after<br />

treatment for diagnosed malaria following a controlled <strong>infection</strong>, although a<br />

definite relationship between the cardiac event <strong>and</strong> the controlled malaria<br />

<strong>infection</strong> was not established [60]. The chronology of the event with the malaria<br />

<strong>infection</strong> has raised discussion on a possible pathophysiological link between<br />

cardiac events <strong>and</strong> malaria. An ischemic cardiac event has previously been<br />

described after CHMI, however, this volunteer was never parasitemic [59]. There<br />

have been no other reports of ischemic cardiac events in the context of malaria,<br />

but events of myocarditis, although rare, have been found in several malaria<br />

patients [61-65]. Myocarditis has also been recognised as an immunological<br />

complication following all types of vaccinations, although particularly smallpox<br />

vaccines are renown [66-68]. Notwithst<strong>and</strong>ing the aetiology, the identification of<br />

volunteers that may possibly develop cardiac complications before start of the<br />

CHMI <strong>and</strong> during the <strong>infection</strong> is important to safeguard volunteers. Therefore,<br />

it has been agreed that volunteers with an increased risk of cardiac disease<br />

should be excluded from such trials. Currently, the SCORE risk assessment [69] is<br />

used to identify those high risk volunteers, although risk factors for malaria<br />

induced ischemia may not resemble those for atherosclerotic processes. In<br />

addition, regular measurements of highly sensitive troponin are performed in<br />

order to detect cardiac damage in an early stage. However, the increased<br />

sensitivity of t-troponin detection parallels decreased clinical relevance with<br />

detection of minimal cardiac damage. For example, increased highly-sensitive


General Discussion 223<br />

troponins can also be found in marathon runners [70], providing evidence for<br />

minimal cardiac damage that may not be clinically relevant. D-dimers <strong>and</strong> LDH<br />

have also been proposed as markers for ischemia, based on the hypothesis that<br />

endothelial cell activation <strong>and</strong> increased turnover of coagulation would lead to<br />

thrombotic events. Again, although highly sensitive, these markers lack<br />

specificity for clinically relevant cardiac damage. Highly elevated d-dimers,<br />

particularly after initiation of antimalarial-treatment, have been found in several<br />

CHMI volunteers since the initiation of monitoring without re-occurrence of<br />

cardiac damage. The correlation of d-dimers with peak temperature suggests<br />

that d-dimers are a byst<strong>and</strong>er in inflammation rather than a predictor of cardiac<br />

damage (Chapter 10). In conclusion, the lack of knowledge on the aetiology of<br />

the cardiac event makes the identification of a “risk correlate” for cardiac<br />

damage, <strong>and</strong> the subsequent identification of volunteers at risk difficult, if not<br />

impossible. The continuous monitoring of volunteers for the occurrence of<br />

cardiac damage, by means of regular measurement of highly-sensitive troponins<br />

thus seems reasonable in order to detect any cardiac event in an early stage.<br />

Moreover, the occurrence of the cardiac event has, once again, stressed the<br />

importance of close <strong>and</strong> committed monitoring by the study physicians in order<br />

to detect any events in an early stage <strong>and</strong> prevent escalation. However, the<br />

apparently infrequent occurrence of serious adverse events in the large group of<br />

volunteers exposed to controlled <strong>infection</strong>s so far <strong>and</strong> the wealth of data on<br />

clinical cases lacking evidence for cardiac damage, may be somewhat reassuring.<br />

In addition to the clinical manifestations, participation in a controlled malaria<br />

<strong>infection</strong> trial has a major impact on the daily life of volunteers. This is<br />

particularly due to the intense follow-up with blood sampling several times daily.<br />

Volunteers’ perception of their participation in such a trial depends mainly on<br />

whether they have realistic expectations of trial procedures <strong>and</strong> the severity of<br />

symptoms, indicating the importance of providing accurate <strong>and</strong> sufficient<br />

information to volunteers before the onset of the trial.<br />

As is true for any type of clinical research, risks must be minimized <strong>and</strong> scientific<br />

benefits maximized. We believe that the benefits of Phase IIa trials outweigh the<br />

potential risks in well-designed studies <strong>and</strong> will be essential to the development<br />

of an effective malaria vaccine, provided that all safeguards are in place for the<br />

safety of volunteers [71].<br />

Differences between natural <strong>and</strong> controlled experimental <strong>infection</strong>s signify the<br />

importance of validating the results of Phase IIa challenge trials with data from


224 Chapter 11<br />

Phase IIb field trials in malaria-endemic areas. Only four c<strong>and</strong>idate vaccines have<br />

been assessed by both types of trial, allowing a comparison of the protective<br />

outcomes. The best studied c<strong>and</strong>idate vaccine, RTS,S, which is currently in Phase<br />

III trials, has repeatedly demonstrated a protective efficacy of ~30–50% in Phase<br />

IIa trials with sterile <strong>protection</strong> as the study end point [41, 72, 73]. Interestingly,<br />

a similar ~30–50% efficacy of RTS,S was found in Phase IIb trials in the field using<br />

time to first clinical malaria episode as the primary study end point [74, 75]. A<br />

similar association between the results of Phase IIa <strong>and</strong> Phase IIb trials was<br />

found when testing long-term <strong>protection</strong> in adults [74, 75]. A second preerythrocytic<br />

stage c<strong>and</strong>idate vaccine, ME-TRAP (a multi-epitope string fused to<br />

thrombospondin-related adhesion protein), delivered by a DNA prime <strong>and</strong><br />

attenuated poxvirus boost, induced complete <strong>protection</strong> in only a few<br />

volunteers (three out of 74) in Phase IIa trials, <strong>and</strong> no <strong>protection</strong> was found in<br />

adult Phase IIb field studies in the Gambia [76, 77]. Artificial blood-stage<br />

challenge has been used in a Phase II trial after immunization with Combination<br />

B, a combination of merozoite surface protein 1 (MSP1), MSP2 <strong>and</strong> ring-infected<br />

erythrocyte surface antigen (RESA) in 17 volunteers, which resulted in no<br />

decrease in parasite growth rates [78]; this is in line with results from a Phase IIb<br />

trial of Combination B conducted in Papua New Guinea [79]. Finally, the vaccine<br />

c<strong>and</strong>idate AMA1 (3D7 strain) did not show <strong>protection</strong> in a controlled <strong>infection</strong><br />

trial [26] <strong>and</strong> parasitological outcome in an efficacy trial was also non-significant<br />

[16]. These limited data indicate that results obtained in controlled malaria<br />

<strong>infection</strong> trials are generally in line with results in the field, but more<br />

comparisons are required before definite conclusions can be drawn.<br />

The extrapolation of results from controlled <strong>infection</strong>s to the field may also be<br />

limited by several potential differences between controlled human malaria<br />

<strong>infection</strong>s <strong>and</strong> naturally acquired <strong>infection</strong>s. One important difference is the<br />

delivery of parasites. In an experimental situation the parasite load is delivered<br />

almost instantly by five infected mosquitoes. Such a high parasite burden has<br />

been considered unnatural <strong>and</strong> might be an overly stringent test for the<br />

protective capacity of the vaccine-induced immune response [80]. However,<br />

although the frequency of infectious mosquito bites is generally less than this in<br />

malaria-endemic areas, intense transmission can occur. A person may be<br />

subjected to 35–96 mosquito bites per night, <strong>and</strong> in certain areas approximately<br />

10% of mosquitoes are infected with Pf [81].<br />

Second, controlled malaria <strong>infection</strong>s are carried out using one parasite strain<br />

only, whereas it is well known that Pf field strains are genetically diverse within


General Discussion 225<br />

<strong>and</strong> between regions [82]. Genetic diversity of the parasite strains is a major<br />

challenge for vaccines that target strain-specific antigens, of which the vaccine<br />

c<strong>and</strong>idate AMA1 is one example, <strong>and</strong> puts limitations on the direct translation of<br />

results from experimental <strong>infection</strong>s into the field situation. The availability of a<br />

small portfolio of genetically well-characterized Pf strains for controlled malaria<br />

<strong>infection</strong>s would be a major asset. We have taken the first step towards a field<br />

strain <strong>infection</strong> model by showing that NF135, a field strain from Cambodia is<br />

infectious to humans (Chapter 7). Future studies will need to establish whether<br />

the infectivity of NF135 is comparable to NF54 <strong>and</strong> the dose that should be<br />

administered to achieve 100% <strong>infection</strong> rates. Once several field strains are<br />

available, the controlled <strong>infection</strong> of different groups of volunteers with<br />

heterologous strain parasites would be highly instrumental to investigate the<br />

promiscuity of responses induced particularly with diversity covering vaccines or<br />

whole-parasite vaccine strategies.<br />

A final potential limitation of controlled malaria <strong>infection</strong> models relates to the<br />

uncontrolled number of sporozoites inoculated by biting mosquitoes. This<br />

number is generally thought to vary between 100-300 sporozoites per bite up to<br />

a maximum of several thous<strong>and</strong> sporozoites [83-86]. Use of a well-defined<br />

number of inoculated sporozoites will strengthen the power of the model<br />

(Chapter 6). In principle, the most accurate way of dosing sporozoites is to inject<br />

them directly by needle <strong>and</strong> syringe, as the number of sporozoites counted in<br />

mosquito salivary gl<strong>and</strong>s or the number of mosquito bites is a poor predictor of<br />

the number of sporozoites injected [84]<br />

Whole-sporozoite inoculation<br />

The concept of inoculation of whole sporozoites into humans is not novel. The<br />

first reports can be found in 1926, when five patients were injected with P. vivax<br />

in order to test antimalarial drugs [87]. More frequent records are found<br />

between 1937 <strong>and</strong> 1963 when malariatherapy was given to neurosyphilis<br />

patients, mostly by bites of infected mosquitoes but also by sporozoite injection<br />

[88, 89]. Infection of patients with P. falciparum or P. vivax sporozoites was<br />

primarily by intravenous route of freshly extracted sporozoites, since the<br />

viability was known to heavily depend on storage time, condition <strong>and</strong> h<strong>and</strong>ling<br />

[88]. Intradermal injection of sporozoites to investigate its relationship with<br />

prepatent period or clinical outcome were variably successful, mostly because of<br />

the unstable <strong>infection</strong> rates [90, 91]. Techniques to freeze <strong>and</strong> thaw sporozoites


226 Chapter 11<br />

were also tried at that time, sometimes accompanied by an intramuscular<br />

injection of penicillin to prevent bacterial co-<strong>infection</strong> [92-94]. The practice of<br />

malariatherapy stopped with the advent of antibiotics. Techniques to induce<br />

malaria were subsequently used in the vaccine research field, when the first<br />

available malaria vaccine c<strong>and</strong>idates were tested [95, 96]. However, by that<br />

time, Pf gametocytes could be produced in culture <strong>and</strong> Anopheles mosquitoes<br />

were infected by feeding on culture material [97, 98]. Because these methods<br />

were safer <strong>and</strong> less costly, mosquitoes bites have been used for controlled<br />

<strong>infection</strong>s ever since.<br />

The translation of mosquito-delivered inoculations into whole-sporozoite needle<br />

administration is a technical challenge because of difficulties to isolate, purify<br />

<strong>and</strong> cryopreserve sporozoites according to current regulatory st<strong>and</strong>ards. Recent<br />

progress has been made by Sanaria Inc., which has developed technology for the<br />

purification <strong>and</strong> cryopreservation of aseptic sporozoites for use in humans<br />

according to the current safety st<strong>and</strong>ards [99]. Initially developed as an<br />

attenuated sporozoite vaccine, these sporozoites were irradiated <strong>and</strong> tested for<br />

safety, immunogenicity <strong>and</strong> efficacy in a clinical vaccine trial. Unfortunately,<br />

although safe, the trial showed limited protectivity <strong>and</strong> immunogenicity [100].<br />

We subsequently tested the unattenuated cryopreserved sporozoites for<br />

infectiousness by intradermal injection, proving their potency in five of six<br />

volunteers from each of three dose groups (Chapter 8). Future studies will focus<br />

on improving the administration of these sporozoites in order to achieve 100%<br />

<strong>infection</strong> rates. Results of these studies will not only attribute to the<br />

development of a st<strong>and</strong>ardized controlled human malaria <strong>infection</strong> model, but<br />

also advance the development of a whole-sporozoite vaccine. However, one<br />

must bear in mind that needle <strong>and</strong> syringe administration of a bolus of<br />

sporozoites is clearly different from mosquito bite delivery. Mosquitoes deliver a<br />

proportion of sporozoites intracapillary <strong>and</strong> a proportion intradermally [101].<br />

Sporozoites are embedded in mosquito saliva when inoculated, components of<br />

which may possibly improve infectivity [102]. The volume of injection by<br />

mosquito is also considerably smaller than will ever be reached by needle <strong>and</strong><br />

syringe. These factors may be important to consider particularly if sporozoites<br />

are injected for the testing sporozoite vaccines that aim to induce antibodies to<br />

immobilize sporozoites.<br />

Murine studies indicate that the intramuscular administration of sporozoites by<br />

needle may be more efficient than the intradermal delivery, possible due to<br />

better circulation of the muscular tissue (Ploemen pers comm.). The


General Discussion 227<br />

immunological consequences of dermal versus intramuscular administration are<br />

currently being investigated (Nganou-Makamdop pers comm.). Whether or not<br />

the administration of local vasodilators or the use of smaller injection volumes<br />

improves the immunogenicity or infectivity of sporozoites will also be<br />

investigated. In any case, it will likely take several years before the needle<br />

administration of sporozoites has been optimized for human use. However<br />

elaborate, these studies are not only important to the development of a<br />

st<strong>and</strong>ardized controlled malaria <strong>infection</strong> model, but will also prove to be crucial<br />

to the translation of mosquito-delivered sporozoites into a vaccine strategy.<br />

Whole sporozoite vaccines<br />

The concept of whole-parasite immunisation originates from studies in which<br />

humans were immunized by irradiated infected mosquito bite <strong>and</strong> proved to be<br />

protected <strong>against</strong> subsequent Pf challenge (summarized in [103]). Irradiation of<br />

infectious mosquitoes disrupts the gene expression of sporozoites, which remain<br />

capable of hepatocyte invasion but are no longer capable of complete liver-stage<br />

maturation or progression to the pathogenic blood stage [104]. Exposure of<br />

humans to the antigen repertoire of the irradiated sporozoites proved to be<br />

sufficient to induce <strong>protection</strong> <strong>against</strong> subsequent challenge with viable<br />

sporozoites, which was shown to last 14 months in several instances [103]. The<br />

radiation dose was crucial: too much radiation reduced potency while too low<br />

radiation doses caused break-through <strong>infection</strong>s [105].<br />

Although these results were published already in the 1960’s they were not<br />

considered a vaccine strategy, because the production of a whole-sporozoite<br />

vaccine that would comply with regulatory st<strong>and</strong>ards seemed technically<br />

impossible. Moreover, the potency of these attenuated sporozoites was limited:<br />

high doses [> 1000 mosquito bites) were needed to achieve full <strong>protection</strong> [103].<br />

This requirement for large immunisation doses might reflect the lack of normal<br />

intrahepatocytic differentiation of irradiated sporozoites into thous<strong>and</strong>s of<br />

hepatic merozoites, an amplification process that is expected to result in an<br />

exp<strong>and</strong>ed antigen repertoire <strong>and</strong> thus drive immune responses qualitatively <strong>and</strong><br />

quantitatively [106].<br />

The <strong>protection</strong> of human volunteers from Pf challenge by mosquito-delivered<br />

whole sporozoite inoculation under chloroquine prophylaxis (CPS, Chapter 9)<br />

shed new light on the concept of whole-parasite immunisation, because this<br />

strategy proved to be much more efficient at inducing <strong>protection</strong>. In this case,


228 Chapter 11<br />

only ~45 infected mosquito bites were needed to induce full <strong>protection</strong>.<br />

Moreover, the follow-up study (Chapter 10) after 2.5 years proved that<br />

<strong>protection</strong> was long-lasting in at least a proportion of volunteers. Moreover, the<br />

CPS trial has led to the identification of pluripotent (IFNγ+IL-2+) effector memory<br />

responses as a potential immunological correlate of <strong>protection</strong> (Chapter 9).<br />

Serial in depth immunological analyses revealed that various lymphocyte subsets<br />

contribute to the IFNγ response, including αβT cells, γδT <strong>and</strong> NK cells. Both γδT<br />

cells <strong>and</strong> αβT cells were found to independently contribute to immunological<br />

memory [107]. Interestingly, individual antigens responsible for the induction of<br />

the IFNγ response (in vitro) could not be identified. Apparently, the induction of<br />

IFNγ by Pf is complex, not only involving several cell types with a possible central<br />

role for multifunctional T-cells, but also relying on a combination of multiple<br />

antigens. Moreover, interindividual variation in the quantity <strong>and</strong> quality of IFNγ<br />

responses reveals significant heterogeneity in Pf-induced immune responses.<br />

The true correlate of <strong>protection</strong> may thus not be universal, but also display<br />

diversity with several different immunological profiles correlating with<br />

<strong>protection</strong>. Whether or not multifunctional T-cells form part of this correlate,<br />

remains to be investigated. However, it seems likely that the true correlate of<br />

<strong>protection</strong> will be a composite of multiple responses that form an individual<br />

immunological fingerprint.<br />

Although the CPS model is not a formal vaccine strategy, the CPS studies raised<br />

the question of whether radiation alone produces the most efficient attenuated<br />

parasites. One possible explanation for the greater efficiency of the CPSapproach<br />

might be that the co-administration of chloroquine exhibits immune<br />

modulating effects [108]. Alternatively, the exposure of the immune system to a<br />

greater array of both pre-erythrocytic <strong>and</strong> intraerythrocytic antigens, while<br />

restricting the development of symptomatic <strong>and</strong> potentially immunosuppressive<br />

blood stage parasitemia may be critical to enhance the potency of wholesporozoite<br />

immunization. Indeed, there is some evidence to support the notion<br />

that late-liver stage arresting parasites might be more efficient in inducing<br />

<strong>protection</strong> than early-liver stage arresting parasites [109]. In addition, recent<br />

Plasmodium transcriptome <strong>and</strong> proteome analyses showed an increase in<br />

shared antigens between late-liver stage parasites <strong>and</strong> blood-stage parasites,<br />

supporting the use of late-liver stage arresting parasites as a tool to induce<br />

cross-stage <strong>protection</strong> [110]. To dissect the immunomodulatory capacity of<br />

chloroquine from its parasitocidal effect, it would be of interest to test the<br />

potency of alternative drugs, such as azithromycin or mefloquine when coadministered<br />

with live sporozoites in humans. Indeed, azithromycin has shown


General Discussion 229<br />

to induce <strong>protection</strong> when co-administered with live sporozoites in a mouse<br />

model [111]. Interestingly, both drugs also exhibit immunoregulatory properties<br />

[112-114]. Alternatively, the passive transfer of antibodies directed <strong>against</strong><br />

blood-stage antigens could also prevent clinical disease <strong>and</strong> modulate the<br />

immune response [115].<br />

As an alternative to radiation, several other methods of attenuation, mainly in<br />

rodent models, have been developed. Treatment of sporozoites with the DNA<br />

alkylating agent centanamycin, a compound that particularly attenuates malaria<br />

parasites because it exploits the AT-richness of the parasite genome, impairs the<br />

<strong>infection</strong> of hepatocytes by sporozoites <strong>and</strong> arrests liver stage parasite<br />

development. Mice inoculated with centanamycin-treated sporozoites failed to<br />

produce blood stage <strong>infection</strong>s <strong>and</strong> were protected <strong>against</strong> subsequent<br />

challenge with wild-type sporozoites [116]. Moreover, cross-species <strong>protection</strong><br />

could be induced <strong>and</strong> parasite-specific antibodies <strong>and</strong> IFN-gamma-producing<br />

CD8(+) T cells were induced that were not significantly different from radiationattenuated<br />

sporozoites [116].<br />

In fact, CPS immunisation also implies chemical attenuation, albeit in vivo. CPS as<br />

currently presented is not a widely implementable vaccine strategy, but one<br />

could envision the co-administration of a drug with a needle administered live<br />

vaccine product for a selected group of subjects. However, safety requirements<br />

for such in vivo attenuated product will likely be very stringent, e.g. full<br />

attenuation must be independent from compliance <strong>and</strong> pharmacokinetics of the<br />

drug. Moreover, in vivo attenuation precludes the ability to carry out viability<br />

checks before the vaccine is administered, necessitating extensive postvaccination<br />

follow-up.<br />

In addition to the r<strong>and</strong>om genetic attenuation induced by radiation or chemicals,<br />

targeted attenuation of sporozoites can be achieved by inactivation of specific<br />

genes by molecular techniques. The main advantage of Genetically Attenuated<br />

Parasites [GAP) over radiation <strong>and</strong> chemically attenuated parasites is the fact<br />

that a homogeneous parasite population with a defined phenotype is obtained.<br />

The recent availability of complete Plasmodium genome sequences permits the<br />

development of live-attenuated parasites by more precise <strong>and</strong> defined genetic<br />

manipulations [117]. Based on their expression profile <strong>and</strong> phenotype,<br />

approximately seven genes have been selected for attenuation <strong>and</strong> knock-out<br />

strains created primarily in rodent malaria parasites P. berghei <strong>and</strong> P. yoelii (p52,<br />

p36, uis3, uis4, fabb/f, fabz, sap1 [118-126]). Immunisation of mice with GAP


230 Chapter 11<br />

results in protective immune responses that are similar to those induced by<br />

irradiated sporozoites [127, 128]. To ensure safety of the GAP vaccine, the liver<br />

arrest of a genetically attenuated parasite vaccine needs to be complete <strong>and</strong><br />

irreversible [129]. Occasional breakthrough <strong>infection</strong>s in GAP lacking genes uis4<br />

<strong>and</strong> p52, emphasize that multiple genes will have to be removed in order to<br />

achieve complete arrest [119, 120]. Recently studies, however, show that also<br />

parasites lacking expression of both P52 <strong>and</strong> P36 are capable of developing<br />

through the liver stage into blood stage <strong>infection</strong>s in vitro <strong>and</strong> in mouse models<br />

[130]. This raises concerns about the number of genes that need to be<br />

attenuated to ensure safety of a whole-sporozoite vaccine. The potency of a<br />

GAP, on the contrary, depends on development of the attenuated parasites into<br />

the late liver stage, engendering higher levels of T-cell responses <strong>and</strong> cross-stage<br />

<strong>and</strong> cross–species <strong>protection</strong> [124]. Whether or not safety <strong>and</strong> potency<br />

requirements can both be adequately met in one multiply-attenuated whole<br />

parasite product will have to be investigated. In addition, the GAP vaccines also<br />

require precautions with respect to the introduction of attenuated parasites into<br />

the environment, most likely necessitating the removal of foreign DNA<br />

sequences from the parasite clones [131].<br />

Regardless of the method of attenuation, a live attenuated sporozoite vaccine<br />

will have to meet stringent safety regulations, as accidental parasite<br />

multiplication may induce a potentially fatal disease particularly if the vaccine is<br />

to be used in immunologically vulnerable populations: HIV-infected subjects or<br />

malnourished infants. An assay that could be reliably <strong>and</strong> repeatedly used to<br />

check the potency of sporozoites for vaccination would be an important asset.<br />

Such a potency assay could also potentially address the issue of translating the<br />

dose of mosquito-delivered inoculation into a needle-delivered vaccines. The<br />

only currently available potency assay is the in vitro hepatocyte invasion assay.<br />

This assay relies on the in vitro invasion of Pf sporozoites in the hepatoma cell<br />

line HepG2, the human transformed hepatocyte cell line HC04 or primary human<br />

hepatocytes, all of which are unfortunately limited by very low <strong>infection</strong><br />

efficiencies (


General Discussion 231<br />

Conclusions <strong>and</strong> perspectives<br />

Controlled human malaria <strong>infection</strong>s (CHMI) provide a model to predict malaria<br />

vaccine efficacy in a well-controlled clinical setting. The CHMI model using Pfinfected<br />

mosquito bites is well established in several international sites <strong>and</strong> is<br />

increasingly used as a crucial check point for the clinical development of preerythrocytic<br />

stage malaria vaccines. The only c<strong>and</strong>idate malaria vaccine showing<br />

protective efficacy in Phase IIb field trials so far is RTS,S. This c<strong>and</strong>idate vaccine<br />

would almost certainly never have been developed without optimization after a<br />

series of Phase IIa trials. Moreover, efficacy data from Phase IIa trials can help to<br />

support the decision-making process by ethical boards <strong>and</strong> communities in<br />

malaria-endemic countries to justify further testing c<strong>and</strong>idate vaccines in Phase<br />

IIb trials in susceptible populations.<br />

Several caveats in st<strong>and</strong>ardisation lead to variability in the primary endpoints of<br />

CHMI trials between institutions, which still need to be addressed. Nevertheless,<br />

small CHMI trials are sufficiently powered to evaluate >50% effective bloodstage<br />

vaccines (Chapter 6). CHMI can be strengthened by the administration of<br />

sporozoites by needle <strong>and</strong> the availability of several heterologous field strains.<br />

Particularly the needle administration of sporozoites can boost the setup of new<br />

clinical trial settings hosted by institutions in malaria-endemic countries, ideally<br />

mimicking local transmission <strong>and</strong> incorporating investigations on vaccine effects<br />

in semi-immune individuals [134]. However, the availability of live sporozoites<br />

for a larger number of trial centres also warrants increased efforts for<br />

st<strong>and</strong>ardisation <strong>and</strong> quality control of the infrastructure. After all, the stringent<br />

selection of volunteers <strong>and</strong> intense clinical follow-up schedule are important to<br />

ensure safety of volunteers.<br />

Clinical development of AMA1 vaccine development has also benefited from the<br />

CHMI model, confirming satisfactory immunogenicity but lack of significant<br />

effects on primary endpoints. Our incomplete underst<strong>and</strong>ing of immunological<br />

correlates to AMA1-induced <strong>protection</strong> in humans seems the main hurdle on the<br />

road to an effective AMA1 vaccine. CHMI can contribute to unravelling these<br />

immunological correlates, provided trials are well designed. For example,<br />

assuming that immunoglobulins are the main effectors to AMA1 induced<br />

immunity, these specific antibodies should be capable of passively transferring<br />

<strong>protection</strong> from one subject to the other, allowing for the identification of<br />

specific antibodies responsible for <strong>protection</strong>. Such antibodies could be the longsought<br />

for correlate of <strong>protection</strong>. Unfortunately, transfer of antigen specific


232 Chapter 11<br />

immunoglobulins has never been tried in malaria research. Such experiments<br />

are difficult to perform nowadays because of increased safety requirements<br />

associated with the transfer of blood products. Once proof of principle for AMA1<br />

is established, diversity covering [20, 21] or combination vaccines with the blood<br />

stage antigen MSP1 [135, 136], Circumsporozoite protein [137, 138] or a mix of<br />

more than two antigens [139], have better chances for success <strong>and</strong> the search<br />

for effective adjuvants, delivery platforms <strong>and</strong> delivery systems can be guided by<br />

a comparison of antibody responses.<br />

The inoculation of whole sporozoites is a very potent <strong>and</strong> efficient way of<br />

inducing <strong>protection</strong> <strong>against</strong> Pf malaria. However, the translation of this approach<br />

into a whole sporozoite vaccine faces several safety issues. In order to ensure<br />

safety the development of an in vitro sporozoite potency test will be of vital<br />

importance. Recent results with multiply-attenuated parasite lines leading to<br />

blood-<strong>infection</strong>, are a signal to scientists to take a prudent approach before<br />

administering live vaccine products to large populations.<br />

In conclusion, immunological research elucidating mechanisms of <strong>protection</strong> <strong>and</strong><br />

enhancing efficacy through antigen-adjuvant combinations may be warranted<br />

for the subunit AMA1 vaccine c<strong>and</strong>idate, whereas the promising efficacy results<br />

of the CPS immunisation strategy might stimulate a more pragmatic approach to<br />

develop an implementable whole-organism based vaccine. This thesis shows<br />

that clinical <strong>infection</strong> trials in small numbers of healthy human volunteers,<br />

although subject to rigorous ethical procedures, can provide valuable insight for<br />

the development of any type of malaria vaccine that would not have been<br />

acquired otherwise. Such tools may prove to be essential to meet the ambitious<br />

goals of the Malaria Vaccine Technology Roadmap by 2015–2025.


General Discussion 233<br />

References<br />

1. Remarque EJ, Faber BW, Kocken CH, Thomas AW. Apical membrane antigen 1: a<br />

malaria vaccine c<strong>and</strong>idate in review. Trends Parasitol 2008; 24:74-84.<br />

2. Dutta S, Sullivan JS, Grady KK et al. High antibody titer <strong>against</strong> apical membrane<br />

antigen-1 is required to protect <strong>against</strong> malaria in the Aotus model. PLoS One<br />

2009; 4:e8138.<br />

3. Crewther PE, Matthew ML, Flegg RH, Anders RF. Protective immune responses<br />

to apical membrane antigen 1 of Plasmodium chabaudi involve recognition of<br />

strain-specific epitopes. Infect Immun 1996; 64:3310-3317.<br />

4. Anders RF, Crewther PE, Edwards S et al. Immunisation with recombinant AMA-<br />

1 protects mice <strong>against</strong> <strong>infection</strong> with Plasmodium chabaudi. Vaccine 1998;<br />

16:240-247.<br />

5. Narum DL, Ogun SA, Thomas AW, Holder AA. Immunization with parasitederived<br />

apical membrane antigen 1 or passive immunization with a specific<br />

monoclonal antibody protects BALB/c mice <strong>against</strong> lethal Plasmodium yoelii<br />

yoelii YM blood-stage <strong>infection</strong>. Infect Immun 2000; 68:2899-2906.<br />

6. Dodoo D, Aikins A, Kusi KA et al. Cohort study of the association of antibody<br />

levels to AMA1, MSP119, MSP3 <strong>and</strong> GLURP with <strong>protection</strong> from clinical malaria<br />

in Ghanaian children. Malar J 2008; 7:142.<br />

7. Nebie I, Diarra A, Ouedraogo A et al. Humoral responses to Plasmodium<br />

falciparum blood-stage antigens <strong>and</strong> association with incidence of clinical<br />

malaria in children living in an area of seasonal malaria transmission in Burkina<br />

Faso, West Africa. Infect Immun 2008; 76:759-766.<br />

8. Pierce MA, Ellis RD, Martin LB et al. Phase 1 safety <strong>and</strong> immunogenicity trial of<br />

the Plasmodium falciparum blood-stage malaria vaccine AMA1-C1/ISA 720 in<br />

Australian adults. Vaccine 2010; 28:2236-2242.<br />

9. Sagara I, Dicko A, Ellis RD et al. A r<strong>and</strong>omized controlled phase 2 trial of the<br />

blood stage AMA1-C1/Alhydrogel malaria vaccine in children in Mali. Vaccine<br />

2009; 27:3090-3098.<br />

10. Saul A, Lawrence G, Allworth A et al. A human phase 1 vaccine clinical trial of<br />

the Plasmodium falciparum malaria vaccine c<strong>and</strong>idate apical membrane antigen<br />

1 in Montanide ISA720 adjuvant. Vaccine 2005; 23:3076-3083.<br />

11. Thera MA, Doumbo OK, Coulibaly D et al. Safety <strong>and</strong> immunogenicity of an<br />

AMA1 malaria vaccine in Malian children: results of a phase 1 r<strong>and</strong>omized<br />

controlled trial. PLoS One 2010; 5:e9041.<br />

12. Dicko A, Sagara I, Ellis RD et al. Phase 1 study of a combination AMA1 blood<br />

stage malaria vaccine in Malian children. PLoS One 2008; 3:e1563.<br />

13. Polhemus ME, Magill AJ, Cummings JF et al. Phase I dose escalation safety <strong>and</strong><br />

immunogenicity trial of Plasmodium falciparum apical membrane protein<br />

[AMA-1) FMP2.1, adjuvanted with AS02A, in malaria-naive adults at the Walter<br />

Reed Army Institute of Research. Vaccine 2007; 25:4203-4212.<br />

14. Thera MA, Doumbo OK, Coulibaly D et al. Safety <strong>and</strong> immunogenicity of an<br />

AMA-1 malaria vaccine in Malian adults: results of a phase 1 r<strong>and</strong>omized<br />

controlled trial. PLoS One 2008; 3:e1465.


234 Chapter 11<br />

15. Lyke KE, Daou M, Diarra I et al. Cell-mediated immunity elicited by the blood<br />

stage malaria vaccine apical membrane antigen 1 in Malian adults: results of a<br />

Phase I r<strong>and</strong>omized trial. Vaccine 2009; 27:2171-2176.<br />

16. Thera MA, Doumbo OK, Coulibaly D et al. A field trial to assess a blood-stage<br />

malaria vaccine. N Engl J Med 2011; 365:1004-1013.<br />

17. Thomas AW, Waters AP, Carr D. Analysis of variation in PF83, an erythrocytic<br />

merozoite vaccine c<strong>and</strong>idate antigen of Plasmodium falciparum. Mol Biochem<br />

Parasitol 1990; 42:285-287.<br />

18. Duncan CJ, Sheehy SH, Ewer KJ et al. Impact on malaria parasite multiplication<br />

rates in infected volunteers of the protein-in-adjuvant vaccine AMA1-<br />

C1/Alhydrogel+CPG 7909. PLoS One 2011; 6:e22271.<br />

19. Ouattara A, Mu J, Takala-Harrison S et al. Lack of allele-specific efficacy of a<br />

bivalent AMA1 malaria vaccine. Malar J 2010; 9:175.<br />

20. Remarque EJ, Faber BW, Kocken CH, Thomas AW. A diversity-covering approach<br />

to immunization with Plasmodium falciparum apical membrane antigen 1<br />

induces broader allelic recognition <strong>and</strong> growth inhibition responses in rabbits.<br />

Infect Immun 2008; 76:2660-2670.<br />

21. Kusi KA, Faber BW, Thomas AW, Remarque EJ. Humoral immune response to<br />

mixed PfAMA1 alleles; multivalent PfAMA1 vaccines induce broad specificity.<br />

PLoS One 2009; 4:e8110.<br />

22. Lynch MM, Cernetich-Ott A, Weidanz WP, Burns JM, Jr. Prediction of merozoite<br />

surface protein 1 <strong>and</strong> apical membrane antigen 1 vaccine efficacies <strong>against</strong><br />

Plasmodium chabaudi malaria based on prechallenge antibody responses. Clin<br />

Vaccine Immunol 2009; 16:293-302.<br />

23. Miura K, Perera S, Brockley S et al. Non-apical membrane antigen 1 [AMA1)<br />

IgGs from Malian children interfere with functional activity of AMA1 IgGs as<br />

judged by growth inhibition assay. PLoS One 2011; 6:e20947.<br />

24. Kennedy MC, Wang J, Zhang Y et al. In vitro studies with recombinant<br />

Plasmodium falciparum apical membrane antigen 1 [AMA1): production <strong>and</strong><br />

activity of an AMA1 vaccine <strong>and</strong> generation of a multiallelic response. Infect<br />

Immun 2002; 70:6948-6960.<br />

25. Hamid MM, Remarque EJ, El Hassan IM et al. Malaria <strong>infection</strong> by sporozoite<br />

challenge induces high functional antibody titres <strong>against</strong> blood stage antigens<br />

after a DNA prime, poxvirus boost vaccination strategy in Rhesus macaques.<br />

Malar J 2011; 10:29.<br />

26. Spring MD, Cummings JF, Ockenhouse CF et al. Phase 1/2a study of the malaria<br />

vaccine c<strong>and</strong>idate apical membrane antigen-1 [AMA-1) administered in<br />

adjuvant system AS01B or AS02A. PLoS One 2009; 4:e5254.<br />

27. McGregor IA. The passive transfer of human malarial immunity. Am J Trop Med<br />

Hyg 1964; 13:SUPPL-9.<br />

28. Bouharoun-Tayoun H, Oeuvray C, Lunel F, Druilhe P. Mechanisms underlying<br />

the monocyte-mediated antibody-dependent killing of Plasmodium falciparum<br />

asexual blood stages. J Exp Med 1995; 182:409-418.<br />

29. Kumaratilake LM, Rathjen DA, Mack P, Widmer F, Prasertsiriroj V, Ferrante A. A<br />

synthetic tumor necrosis factor-alpha agonist peptide enhances human


General Discussion 235<br />

polymorphonuclear leukocyte-mediated killing of Plasmodium falciparum in<br />

vitro <strong>and</strong> suppresses Plasmodium chabaudi <strong>infection</strong> in mice. J Clin Invest 1995;<br />

95:2315-2323.<br />

30. Wipasa J, Hirunpetcharat C, Mahakunkijcharoen Y, Xu H, Elliott S, Good MF.<br />

Identification of T cell epitopes on the 33-kDa fragment of Plasmodium yoelii<br />

merozoite surface protein 1 <strong>and</strong> their antibody-independent protective role in<br />

immunity to blood stage malaria. J Immunol 2002; 169:944-951.<br />

31. McCall MB, Sauerwein RW. Interferon-gamma--central mediator of protective<br />

immune responses <strong>against</strong> the pre-erythrocytic <strong>and</strong> blood stage of malaria. J<br />

Leukoc Biol 2010; 88:1131-1143.<br />

32. Pombo DJ, Lawrence G, Hirunpetcharat C et al. Immunity to malaria after<br />

administration of ultra-low doses of red cells infected with Plasmodium<br />

falciparum. Lancet 2002; 360:610-617.<br />

33. Huaman MC, Mullen GE, Long CA, Mahanty S. Plasmodium falciparum apical<br />

membrane antigen 1 vaccine elicits multifunctional CD4 cytokine-producing <strong>and</strong><br />

memory T cells. Vaccine 2009; 27:5239-5246.<br />

34. Lal AA, Hughes MA, Oliveira DA et al. Identification of T-cell determinants in<br />

natural immune responses to the Plasmodium falciparum apical membrane<br />

antigen [AMA-1) in an adult population exposed to malaria. Infect Immun 1996;<br />

64:1054-1059.<br />

35. Udhayakumar V, Kariuki S, Kolczack M et al. Longitudinal study of natural<br />

immune responses to the Plasmodium falciparum apical membrane antigen<br />

[AMA-1) in a holoendemic region of malaria in western Kenya: Asembo Bay<br />

Cohort Project VIII. Am J Trop Med Hyg 2001; 65:100-107.<br />

36. Amante FH, Crewther PE, Anders RF, Good MF. A cryptic T cell epitope on the<br />

apical membrane antigen 1 of Plasmodium chabaudi adami can prime for an<br />

anamnestic antibody response: implications for malaria vaccine design. J<br />

Immunol 1997; 159:5535-5544.<br />

37. Burns JM, Jr., Flaherty PR, Nanavati P, Weidanz WP. Protection <strong>against</strong><br />

Plasmodium chabaudi malaria induced by immunization with apical membrane<br />

antigen 1 <strong>and</strong> merozoite surface protein 1 in the absence of gamma interferon<br />

or interleukin-4. Infect Immun 2004; 72:5605-5612.<br />

38. Xu H, Hodder AN, Yan H, Crewther PE, Anders RF, Good MF. CD4+ T cells acting<br />

independently of antibody contribute to protective immunity to Plasmodium<br />

chabaudi <strong>infection</strong> after apical membrane antigen 1 immunization. J Immunol<br />

2000; 165:389-396.<br />

39. Dutta S, Haynes JD, Moch JK, Barbosa A, Lanar DE. Invasion-inhibitory<br />

antibodies inhibit proteolytic processing of apical membrane antigen 1 of<br />

Plasmodium falciparum merozoites. Proc Natl Acad Sci U S A 2003; 100:12295-<br />

12300.<br />

40. Silvie O, Franetich JF, Charrin S et al. A role for apical membrane antigen 1<br />

during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol<br />

Chem 2004; 279:9490-9496.<br />

41. Casares S, Brumeanu TD, Richie TL. The RTS,S malaria vaccine. Vaccine 2010;<br />

28:4880-4894.


236 Chapter 11<br />

42. Korsholm KS. One does not fit all: new adjuvants are needed <strong>and</strong> vaccine<br />

formulation is critical. Expert Rev Vaccines 2011; 10:45-48.<br />

43. Douradinha B, Doolan DL. Harnessing immune responses <strong>against</strong> Plasmodium<br />

for rational vaccine design. Trends Parasitol 2011; 27:274-283.<br />

44. Nganou-Makamdop K, van Roosmalen ML, Audouy SA et al. Bacterium-like<br />

particles as multi-epitope delivery platform for Plasmodium berghei<br />

circumsporozoite protein induce complete <strong>protection</strong> <strong>against</strong> malaria in mice.<br />

Malar J 2012; 11:50.<br />

45. Moon JJ, Suh H, Polhemus ME, Ockenhouse CF, Yadava A, Irvine DJ. Antigendisplaying<br />

lipid-enveloped PLGA nanoparticles as delivery agents for a<br />

Plasmodium vivax malaria vaccine. PLoS One 2012; 7:e31472.<br />

46. Hill AV, Reyes-S<strong>and</strong>oval A, O'Hara G et al. Prime-boost vectored malaria<br />

vaccines: progress <strong>and</strong> prospects. Hum Vaccin 2010; 6:78-83.<br />

47. Raviprakash K, Porter KR. Needle-free injection of DNA vaccines: a brief<br />

overview <strong>and</strong> methodology. Methods Mol Med 2006; 127:83-89.<br />

48. Senel S, Rathbone MJ, Cansiz M, Pather I. Recent developments in buccal <strong>and</strong><br />

sublingual delivery systems. Expert Opin Drug Deliv 2012.<br />

49. Mahdi Abdel HM, Remarque EJ, van Duivenvoorde LM et al. Vaccination with<br />

Plasmodium knowlesi AMA1 formulated in the novel adjuvant co-vaccine HT<br />

protects <strong>against</strong> blood-stage challenge in rhesus macaques. PLoS One 2011;<br />

6:e20547.<br />

50. Sauerwein RW, Roestenberg M, Moorthy VS. <strong>Experimental</strong> human challenge<br />

<strong>infection</strong>s can accelerate clinical malaria vaccine development. Nat Rev<br />

Immunol 2011; 11:57-64.<br />

51. McCall MB, Netea MG, Hermsen CC et al. Plasmodium falciparum <strong>infection</strong><br />

causes proinflammatory priming of human TLR responses. J Immunol 2007;<br />

179:162-171.<br />

52. Hermsen CC, Telgt DS, Linders EH et al. Detection of Plasmodium falciparum<br />

malaria parasites in vivo by real-time quantitative PCR. Mol Biochem Parasitol<br />

2001; 118:247-251.<br />

53. Felger I, Genton B, Smith T, Tanner M, Beck HP. Molecular monitoring in<br />

malaria vaccine trials. Trends Parasitol 2003; 19:60-63.<br />

54. Hermsen CC, de Vlas SJ, van Gemert GJ, Telgt DS, Verhage DF, Sauerwein RW.<br />

Testing vaccines in human experimental malaria: statistical analysis of<br />

parasitemia measured by a quantitative real-time polymerase chain reaction.<br />

Am J Trop Med Hyg 2004; 71:196-201.<br />

55. Dietz K, Raddatz G, Molineaux L. Mathematical model of the first wave of<br />

Plasmodium falciparum asexual parasitemia in non-immune <strong>and</strong> vaccinated<br />

individuals. Am J Trop Med Hyg 2006; 75:46-55.<br />

56. Bejon P, Andrews L, Andersen RF et al. Calculation of liver-to-blood inocula,<br />

parasite growth rates, <strong>and</strong> preerythrocytic vaccine efficacy, from serial<br />

quantitative polymerase chain reaction studies of volunteers challenged with<br />

malaria sporozoites. J Infect Dis 2005; 191:619-626.


General Discussion 237<br />

57. Church LW, Le TP, Bryan JP et al. Clinical manifestations of Plasmodium<br />

falciparum malaria experimentally induced by mosquito challenge. J Infect Dis<br />

1997; 175:915-920.<br />

58. Epstein JE, Rao S, Williams F et al. Safety <strong>and</strong> clinical outcome of experimental<br />

challenge of human volunteers with Plasmodium falciparum-infected<br />

mosquitoes: an update. J Infect Dis 2007; 196:145-154.<br />

59. Verhage DF, Telgt DS, Bousema JT et al. Clinical outcome of experimental<br />

human malaria induced by Plasmodium falciparum-infected mosquitoes. Neth J<br />

Med 2005; 63:52-58.<br />

60. Nieman AE, de MQ, Roestenberg M et al. Cardiac complication after<br />

experimental human malaria <strong>infection</strong>: a case report. Malar J 2009; 8:277.<br />

61. Costenaro P, Benedetti P, Facchin C, Mengoli C, Pellizzer G. Fatal Myocarditis in<br />

Course of Plasmodium falciparum Infection: Case Report <strong>and</strong> Review of Cardiac<br />

Complications in Malaria. Case Report Med 2011; 2011:202083.<br />

62. Horstmann RD, Ehrich JH, Beck J, Dietrich M. [Fatal complications of tropical<br />

malaria in non-immune patients. A retrospective clinico-pathologic analysis of<br />

25 cases]. Dtsch Med Wochenschr 1985; 110:1651-1656.<br />

63. Mohsen AH, Green ST, West JN, McKendrick MW. Myocarditis associated with<br />

Plasmodium falciparum malaria: a case report <strong>and</strong> a review of the literature. J<br />

Travel Med 2001; 8:219-220.<br />

64. Goljan J, Nahorski WL, Wroczynska A, Felczak-Korzybska I, Pietkiewicz H. Severe<br />

malaria--analysis of prognostic symptoms <strong>and</strong> signs in 169 patients treated in<br />

Gdynia in 1991-2005. Int Marit Health 2006; 57:149-162.<br />

65. Wichmann O, Loscher T, Jelinek T. Fatal malaria in a German couple returning<br />

from Burkina Faso. Infection 2003; 31:260-262.<br />

66. Eckart RE, Shry EA, Jones SO, Atwood JE, Grabenstein JD. Comparison of clinical<br />

presentation of acute myocarditis following smallpox vaccination to acute<br />

coronary syndromes in patients


238 Chapter 11<br />

72. Kester KE, McKinney DA, Tornieporth N et al. Efficacy of recombinant<br />

circumsporozoite protein vaccine regimens <strong>against</strong> experimental Plasmodium<br />

falciparum malaria. J Infect Dis 2001; 183:640-647.<br />

73. Kester KE, Cummings JF, Ockenhouse CF et al. Phase 2a trial of 0, 1, <strong>and</strong> 3<br />

month <strong>and</strong> 0, 7, <strong>and</strong> 28 day immunization schedules of malaria vaccine<br />

RTS,S/AS02 in malaria-naive adults at the Walter Reed Army Institute of<br />

Research. Vaccine 2008; 26:2191-2202.<br />

74. Bojang KA, Milligan PJ, Pinder M et al. Efficacy of RTS,S/AS02 malaria vaccine<br />

<strong>against</strong> Plasmodium falciparum <strong>infection</strong> in semi-immune adult men in The<br />

Gambia: a r<strong>and</strong>omised trial. Lancet 2001; 358:1927-1934.<br />

75. Kester KE, Cummings JF, Ofori-Anyinam O et al. R<strong>and</strong>omized, double-blind,<br />

phase 2a trial of falciparum malaria vaccines RTS,S/AS01B <strong>and</strong> RTS,S/AS02A in<br />

malaria-naive adults: safety, efficacy, <strong>and</strong> immunologic associates of <strong>protection</strong>.<br />

J Infect Dis 2009; 200:337-346.<br />

76. Moorthy VS, Imoukhuede EB, Milligan P et al. A r<strong>and</strong>omised, double-blind,<br />

controlled vaccine efficacy trial of DNA/MVA ME-TRAP <strong>against</strong> malaria <strong>infection</strong><br />

in Gambian adults. PLoS Med 2004; 1:e33.<br />

77. Dunachie SJ, Walther M, Epstein JE et al. A DNA prime-modified vaccinia virus<br />

ankara boost vaccine encoding thrombospondin-related adhesion protein but<br />

not circumsporozoite protein partially protects healthy malaria-naive adults<br />

<strong>against</strong> Plasmodium falciparum sporozoite challenge. Infect Immun 2006;<br />

74:5933-5942.<br />

78. Lawrence G, Cheng QQ, Reed C et al. Effect of vaccination with 3 recombinant<br />

asexual-stage malaria antigens on initial growth rates of Plasmodium falciparum<br />

in non-immune volunteers. Vaccine 2000; 18:1925-1931.<br />

79. Genton B, al-Yaman F, Betuela I et al. Safety <strong>and</strong> immunogenicity of a threecomponent<br />

blood-stage malaria vaccine [MSP1, MSP2, RESA) <strong>against</strong><br />

Plasmodium falciparum in Papua New Guinean children. Vaccine 2003; 22:30-<br />

41.<br />

80. Genton B, D'Acremont V, Lurati-Ruiz F et al. R<strong>and</strong>omized double-blind<br />

controlled Phase I/IIa trial to assess the efficacy of malaria vaccine PfCS102 to<br />

protect <strong>against</strong> challenge with P. falciparum. Vaccine 2010; 28:6573-6580.<br />

81. Trape JF, Zoulani A. Malaria <strong>and</strong> urbanization in central Africa: the example of<br />

Brazzaville. Part II: Results of entomological surveys <strong>and</strong> epidemiological<br />

analysis. Trans R Soc Trop Med Hyg 1987; 81 Suppl 2:10-18.<br />

82. Takala SL, Coulibaly D, Thera MA et al. Extreme polymorphism in a vaccine<br />

antigen <strong>and</strong> risk of clinical malaria: implications for vaccine development. Sci<br />

Transl Med 2009; 1:2ra5.<br />

83. Rosenberg R, Wirtz RA, Schneider I, Burge R. An estimation of the number of<br />

malaria sporozoites ejected by a feeding mosquito. Trans R Soc Trop Med Hyg<br />

1990; 84:209-212.<br />

84. Ponnudurai T, Lensen AH, van Gemert GJ, Bolmer MG, Meuwissen JH. Feeding<br />

behaviour <strong>and</strong> sporozoite ejection by infected Anopheles stephensi. Trans R Soc<br />

Trop Med Hyg 1991; 85:175-180.


General Discussion 239<br />

85. Beier JC, Davis JR, Vaughan JA, Noden BH, Beier MS. Quantitation of<br />

Plasmodium falciparum sporozoites transmitted in vitro by experimentally<br />

infected Anopheles gambiae <strong>and</strong> Anopheles stephensi. Am J Trop Med Hyg<br />

1991; 44:564-570.<br />

86. Frischknecht F, Baldacci P, Martin B et al. Imaging movement of malaria<br />

parasites during transmission by Anopheles mosquitoes. Cell Microbiol 2004;<br />

6:687-694.<br />

87. James SP. Report on the first results of laboratory work on malaria in Engl<strong>and</strong>.<br />

Publications of the League of Nations, Health Organization 1926;1-30.<br />

88. Glynn JR, Bradley DJ. Inoculum size, incubation period <strong>and</strong> severity of malaria.<br />

Analysis of data from malaria therapy records. Parasitology 1995; 110 [ Pt 1):7-<br />

19.<br />

89. Collins WE, Jeffery GM. A retrospective examination of secondary sporozoite-<br />

<strong>and</strong> trophozoite-induced <strong>infection</strong>s with Plasmodium falciparum: development<br />

of parasitologic <strong>and</strong> clinical immunity following secondary <strong>infection</strong>. Am J Trop<br />

Med Hyg 1999; 61:20-35.<br />

90. Shute PG, Lupascu G, Branzei P et al. A strain of Plasmodium vivax characterized<br />

by prolonged incubation: the effect of numbers of sporozoites on the length of<br />

the prepatent period. Trans R Soc Trop Med Hyg 1976; 70:474-481.<br />

91. Ungureanu E, Killick-Kendrick R, Garnham PC, Branzei P, Romanescu C, Shute<br />

PG. Prepatent periods of a tropical strain of Plasmodium vivax after inoculations<br />

of tenfold dilutions of sporozoites. Trans R Soc Trop Med Hyg 1976; 70:482-483.<br />

92. Jeffery GM, Rendtorff RC. Preservation of viable human malaria sporozoites by<br />

low-temperature freezing. Exp Parasitol 1955; 4:445-454.<br />

93. Mayne B. The injection of mosquito sporozoites in malaria therapy. Pub Health<br />

Rep 1933;909-913.<br />

94. Mayne B. The technique of induced malaria as used in the South Carolina State<br />

Hospital. Venereal Disease Information 1941; 22:271-276.<br />

95. Ballou WR, Hoffman SL, Sherwood JA et al. Safety <strong>and</strong> efficacy of a recombinant<br />

DNA Plasmodium falciparum sporozoite vaccine. Lancet 1987; 1:1277-1281.<br />

96. Herrington DA, Clyde DF, Losonsky G et al. Safety <strong>and</strong> immunogenicity in man of<br />

a synthetic peptide malaria vaccine <strong>against</strong> Plasmodium falciparum sporozoites.<br />

Nature 1987; 328:257-259.<br />

97. Ifediba T, V<strong>and</strong>erberg JP. Complete in vitro maturation of Plasmodium<br />

falciparum gametocytes. Nature 1981; 294:364-366.<br />

98. Campbell CC, Collins WE, Nguyen-Dinh P, Barber A, Broderson JR. Plasmodium<br />

falciparum gametocytes from culture in vitro develop to sporozoites that are<br />

infectious to primates. Science 1982; 217:1048-1050.<br />

99. Hoffman SL, Billingsley PF, James E et al. Development of a metabolically active,<br />

non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria.<br />

Hum Vaccin 2010; 6:97-106.<br />

100. Epstein JE, Tewari K, Lyke KE et al. Live attenuated malaria vaccine designed to<br />

protect through hepatic CD8 T cell immunity. Science 2011; 334:475-480.


240 Chapter 11<br />

101. Jin Y, Kebaier C, V<strong>and</strong>erberg J. Direct microscopic quantification of dynamics of<br />

Plasmodium berghei sporozoite transmission from mosquitoes to mice. Infect<br />

Immun 2007; 75:5532-5539.<br />

102. Rodriguez MH, Hern<strong>and</strong>ez-Hern<strong>and</strong>ez FL. Insect-malaria parasites interactions:<br />

the salivary gl<strong>and</strong>. Insect Biochem Mol Biol 2004; 34:615-624.<br />

103. Hoffman SL, Goh LM, Luke TC et al. Protection of humans <strong>against</strong> malaria by<br />

immunization with radiation-attenuated Plasmodium falciparum sporozoites. J<br />

Infect Dis 2002; 185:1155-1164.<br />

104. Mellouk S, Lunel F, Sedegah M, Beaudoin RL, Druilhe P. Protection <strong>against</strong><br />

malaria induced by irradiated sporozoites. Lancet 1990; 335:721.<br />

105. V<strong>and</strong>erberg JP, Nussenzweig RS, Most H, Orton CG. Protective immunity<br />

produced by the injection of x-irradiated sporozoites of Plasmodium berghei. II.<br />

Effects of radiation on sporozoites. J Parasitol 1968; 54:1175-1180.<br />

106. Borrmann S, Matuschewski K. Targeting Plasmodium liver stages: better late<br />

than never. Trends Mol Med 2011.<br />

107. Teirlinck AC, McCall MB, Roestenberg M et al. Longevity <strong>and</strong> composition of<br />

cellular immune responses following experimental Plasmodium falciparum<br />

malaria <strong>infection</strong> in humans. PLoS Pathog 2011; 7:e1002389.<br />

108. Sauerwein RW, Bijker EM, Richie TL. Empowering malaria vaccination by drug<br />

administration. Curr Opin Immunol 2010; 22:367-373.<br />

109. Butler NS, Vaughan AM, Harty JT, Kappe SH. Whole parasite vaccination<br />

approaches for prevention of malaria <strong>infection</strong>. Trends Immunol 2012.<br />

110. Tarun AS, Peng X, Dumpit RF et al. A combined transcriptome <strong>and</strong> proteome<br />

survey of malaria parasite liver stages. Proc Natl Acad Sci U S A 2008; 105:305-<br />

310.<br />

111. Friesen J, Silvie O, Putrianti ED, Hafalla JC, Matuschewski K, Borrmann S. Natural<br />

immunization <strong>against</strong> malaria: causal prophylaxis with antibiotics. Sci Transl<br />

Med 2010; 2:40ra49.<br />

112. Lin SJ, Yan DC, Lee WI, Kuo ML, Hsiao HS, Lee PY. Effect of azithromycin on<br />

natural killer cell function. Int Immunopharmacol 2012; 13:8-14.<br />

113. Iwamoto S, Kumamoto T, Azuma E et al. The effect of azithromycin on the<br />

maturation <strong>and</strong> function of murine bone marrow-derived dendritic cells. Clin<br />

Exp Immunol 2011; 166:385-392.<br />

114. Bygbjerg IC, The<strong>and</strong>er TG, Andersen BJ, Flachs H, Jepsen S, Larsen PB. In vitro<br />

effect of chloroquine, mefloquine <strong>and</strong> quinine on human lymphocyte<br />

proliferative responses to malaria antigens <strong>and</strong> other antigens/mitogens. Trop<br />

Med Parasitol 1986; 37:245-247.<br />

115. Ballow M. The IgG molecule as a biological immune response modifier:<br />

mechanisms of action of intravenous immune serum globulin in autoimmune<br />

<strong>and</strong> inflammatory disorders. J Allergy Clin Immunol 2011; 127:315-323.<br />

116. Purcell LA, Yanow SK, Lee M, Spithill TW, Rodriguez A. Chemical attenuation of<br />

Plasmodium berghei sporozoites induces sterile immunity in mice. Infect<br />

Immun 2008; 76:1193-1199.<br />

117. Gardner MJ, Hall N, Fung E et al. Genome sequence of the human malaria<br />

parasite Plasmodium falciparum. Nature 2002; 419:498-511.


General Discussion 241<br />

118. Mueller AK, Labaied M, Kappe SH, Matuschewski K. Genetically modified<br />

Plasmodium parasites as a protective experimental malaria vaccine. Nature<br />

2005; 433:164-167.<br />

119. Mueller AK, Camargo N, Kaiser K et al. Plasmodium liver stage developmental<br />

arrest by depletion of a protein at the parasite-host interface. Proc Natl Acad<br />

Sci U S A 2005; 102:3022-3027.<br />

120. van Dijk MR, Douradinha B, Franke-Fayard B et al. Genetically attenuated, P36pdeficient<br />

malarial sporozoites induce protective immunity <strong>and</strong> apoptosis of<br />

infected liver cells. Proc Natl Acad Sci U S A 2005; 102:12194-12199.<br />

121. Silvie O, Goetz K, Matuschewski K. A sporozoite asparagine-rich protein controls<br />

initiation of Plasmodium liver stage development. PLoS Pathog 2008;<br />

4:e1000086.<br />

122. Labaied M, Harupa A, Dumpit RF, Coppens I, Mikolajczak SA, Kappe SH.<br />

Plasmodium yoelii sporozoites with simultaneous deletion of P52 <strong>and</strong> P36 are<br />

completely attenuated <strong>and</strong> confer sterile immunity <strong>against</strong> <strong>infection</strong>. Infect<br />

Immun 2007; 75:3758-3768.<br />

123. Vaughan AM, O'Neill MT, Tarun AS et al. Type II fatty acid synthesis is essential<br />

only for malaria parasite late liver stage development. Cell Microbiol 2009;<br />

11:506-520.<br />

124. Butler NS, Schmidt NW, Vaughan AM, Aly AS, Kappe SH, Harty JT. Superior<br />

antimalarial immunity after vaccination with late liver stage-arresting<br />

genetically attenuated parasites. Cell Host Microbe 2011; 9:451-462.<br />

125. Aly AS, Lindner SE, MacKellar DC, Peng X, Kappe SH. SAP1 is a critical posttranscriptional<br />

regulator of infectivity in malaria parasite sporozoite stages. Mol<br />

Microbiol 2011; 79:929-939.<br />

126. Aly AS, Mikolajczak SA, Rivera HS et al. Targeted deletion of SAP1 abolishes the<br />

expression of infectivity factors necessary for successful malaria parasite liver<br />

<strong>infection</strong>. Mol Microbiol 2008; 69:152-163.<br />

127. Jobe O, Lumsden J, Mueller AK et al. Genetically attenuated Plasmodium<br />

berghei liver stages induce sterile protracted <strong>protection</strong> that is mediated by<br />

major histocompatibility complex Class I-dependent interferon-gammaproducing<br />

CD8+ T cells. J Infect Dis 2007; 196:599-607.<br />

128. Mueller AK, Deckert M, Heiss K, Goetz K, Matuschewski K, Schluter D.<br />

Genetically attenuated Plasmodium berghei liver stages persist <strong>and</strong> elicit sterile<br />

<strong>protection</strong> primarily via CD8 T cells. Am J Pathol 2007; 171:107-115.<br />

129. Vaughan AM, Wang R, Kappe SH. Genetically engineered, attenuated whole-cell<br />

vaccine approaches for malaria. Hum Vaccin 2010; 6:107-113.<br />

130. Annoura T, Ploemen IH, van Schaijk BC et al. Assessing the adequacy of<br />

attenuation of genetically modified malaria parasite vaccine c<strong>and</strong>idates.<br />

Vaccine 2012; 30:2662-70<br />

131. van Schaijk BC, Vos MW, Janse CJ, Sauerwein RW, Khan SM. Removal of<br />

heterologous sequences from Plasmodium falciparum mutants using FLPerecombinase.<br />

PLoS One 2010; 5:e15121.


242 Chapter 11<br />

132. House BL, Hollingdale MR, Sacci JB, Jr., Richie TL. Functional immunoassays<br />

using an in-vitro malaria liver-stage <strong>infection</strong> model: where do we go from<br />

here? Trends Parasitol 2009; 25:525-533.<br />

133. Morosan S, Hez-Deroubaix S, Lunel F et al. Liver-stage development of<br />

Plasmodium falciparum, in a humanized mouse model. J Infect Dis 2006;<br />

193:996-1004.<br />

134. Chilengi R. Clinical development of malaria vaccines: should earlier trials be<br />

done in malaria endemic countries? Hum Vaccin 2009; 5:627-636.<br />

135. Malkin E, Hu J, Li Z et al. A phase 1 trial of PfCP2.9: an AMA1/MSP1 chimeric<br />

recombinant protein vaccine for Plasmodium falciparum malaria. Vaccine 2008;<br />

26:6864-6873.<br />

136. Ellis RD, Sagara I, Durbin A et al. Comparing the underst<strong>and</strong>ing of subjects<br />

receiving a c<strong>and</strong>idate malaria vaccine in the United States <strong>and</strong> Mali. Am J Trop<br />

Med Hyg 2010; 83:868-872.<br />

137. Okitsu SL, Silvie O, Westerfeld N et al. A virosomal malaria peptide vaccine<br />

elicits a long-lasting sporozoite-inhibitory antibody response in a phase 1a<br />

clinical trial. PLoS One 2007; 2:e1278.<br />

138. Genton B, Pluschke G, Degen L et al. A r<strong>and</strong>omized placebo-controlled phase Ia<br />

malaria vaccine trial of two virosome-formulated synthetic peptides in healthy<br />

adult volunteers. PLoS One 2007; 2:e1018.<br />

139. Wang R, Richie TL, Baraceros MF et al. Boosting of DNA vaccine-elicited gamma<br />

interferon responses in humans by exposure to malaria parasites. Infect Immun<br />

2005; 73:2863-2872.


Chapter 12<br />

Summary<br />

Samenvatting<br />

List of publications<br />

Dankwoord<br />

Curriculum vitae


Summary, Samenvatting, List of publications, Dankwoord, C.V. 245<br />

Summary<br />

Plasmodium falciparum (Pf) malaria is one of the most frequent infectious<br />

diseases <strong>and</strong> is responsible for severe malaria morbidity <strong>and</strong> mortality mainly<br />

among young children in Sub-Saharan Africa. Humans are infected through the<br />

bite of an infected Anopheles mosquito. An effective malaria vaccine is a key tool<br />

of critical research that is needed to support malaria control <strong>and</strong> eradication.<br />

Decades of research have led to the identification of approximately 36 antigens<br />

that have been put forward as vaccine c<strong>and</strong>idates. These antigens are grouped<br />

according to their expression in the Pf lifecycle. Pre-erythrocytic antigens are<br />

expressed in the sporozoite forms of the parasite that invade the human liver<br />

causing subclinical <strong>infection</strong>. Blood stage antigens are expressed during the<br />

parasite development within the host red blood cells, where they are<br />

responsible for the clinical malaria disease, with fever, headache <strong>and</strong> myalgia.<br />

Transmission blocking antigens are derived from the parasite’s sexual stages,<br />

which will complete the lifecycle by reproduction within the mosquito host after<br />

biting the infectious human.<br />

This thesis focuses on the development of research tools that facilitate<br />

immunological <strong>and</strong> vaccine research into human <strong>protection</strong> <strong>against</strong> Plasmodium<br />

falciparum malaria in early stages of clinical development.<br />

In section 1, we describe the safety <strong>and</strong> immunogenicity of the Apical<br />

Membrane Antigen 1 (AMA1), which has been put forward as a promising blood<br />

stage vaccine c<strong>and</strong>idate (Chapter 2). We combined AMA1 with different<br />

adjuvants, Alhydrogel, Montanide <strong>and</strong> AS02 <strong>and</strong> find distinct reactogenicity<br />

profiles, with all vaccine formulations being immunogenic. The magnitude of the<br />

humoral immune response can be enhanced by a more potent adjuvant, but<br />

breadth <strong>and</strong> subclass distribution appear much less influenced by the adjuvant<br />

(Chapter 3). We performed in depth analysis of the concentration <strong>and</strong> avidity of<br />

AMA1 antibodies induced <strong>and</strong> found an inverse relation between these<br />

parameters (Chapter 4). Since Phase II field efficacy trials with different AMA1<br />

formulations have been unsuccessful in inducing <strong>protection</strong> so far, the<br />

development of new adjuvants, delivery systems, strain covering approaches or<br />

the combination of AMA1 with other antigens may provide a means by which<br />

antibody responses can be boosted <strong>and</strong> animal results can be translated into<br />

human efficacy.


246 Chapter 12<br />

Controlled human malaria <strong>infection</strong>s (CHMI) are trials in which healthy<br />

volunteers are exposed to malaria-infected mosquito bites, followed closely <strong>and</strong><br />

treated as soon as blood-stage parasites are detected by microscopy. These<br />

studies are used to assess preliminary efficacy of primarily pre-erythrocytic<br />

vaccine c<strong>and</strong>idates in a limited number of institutions worldwide. In section 2,<br />

we compared the safety <strong>and</strong> parasitological outcome of CHMI in different<br />

institutions <strong>and</strong> conclude that CHMI can be safely conducted, but will lead to<br />

grade 3 adverse events in a proportion of volunteers (Chapter 5). The primary<br />

parasitological outcome of such experiments is highly reproducible within<br />

institutions but may vary between trial centres. In addition, we evaluated the<br />

power of CHMI to detect vaccine efficacy of c<strong>and</strong>idate malaria vaccines (Chapter<br />

6) <strong>and</strong> found that CHMI can confidently assess a reduction in parasitemia for<br />

both pre-erythrocytic <strong>and</strong> blood stage vaccines, possibly broadening its<br />

application in malaria vaccine research.<br />

We subsequently improved the protocol of CHMI trials by increasing the<br />

portfolio of Pf parasites for CHMI with the successful establishment of a new Pf<br />

strain from Cambodia, NF135.C10 (Chapter 7). The availability of this strain<br />

allows for future assessment of cross-strain immunity induced by c<strong>and</strong>idate<br />

vaccines through heterologous CHMI trials. Furthermore, we tested<br />

unattenuated cryopreserved sporozoites for their infectivity by intradermal<br />

injection, proving potency in five of six volunteers from each of three dose<br />

groups (Chapter 8). The needle administration of sporozoites has advantages<br />

over mosquito delivery, in terms of dosing <strong>and</strong> exp<strong>and</strong>ing the global capacity to<br />

perform CHMI trials. However, future studies will need to focus on improving<br />

the administration of these sporozoites in order to achieve 100% <strong>infection</strong> rates.<br />

Moreover, with an increasing number of CHMI centres being installed, priority<br />

should be given to initiatives to st<strong>and</strong>ardize challenge procedures in order to<br />

ensure comparability of results worldwide.<br />

In section 3 we show that viable intact sporozoites are capable of inducing<br />

<strong>protection</strong> to Pf malaria in humans very efficiently, if high levels of blood stage<br />

parasitemia <strong>and</strong> clinical disease are prevented by co-administration of the drug<br />

chloroquine (CPS, Chapter 9). Chloroquine kills blood stage parasites but does<br />

not affect the hepatic development of malaria parasites. A protocol in which we<br />

exposed healthy, malaria-naïve volunteers to the bites of infected mosquitoes<br />

under chloroquine prophylaxis on three occasions with monthly intervals,<br />

rendered 10 of 10 volunteers protected <strong>against</strong> subsequent controlled <strong>infection</strong>.<br />

We found that <strong>protection</strong> was associated with an increased response of


Summary, Samenvatting, List of publications, Dankwoord, C.V. 247<br />

pluripotent effector memory T-cells. We showed that <strong>protection</strong> lasts for more<br />

than two years in four of six re-challenged volunteers <strong>and</strong> found a markedly<br />

delayed patency in the two remaining volunteers (Chapter 10). Whereas the<br />

methodology described here does not itself represent a widely implementable<br />

vaccine strategy, the induction of <strong>protection</strong> <strong>against</strong> an homologous malaria<br />

challenge suggests that the concept of a whole parasite malaria vaccine<br />

warrants further consideration. However, the development of a whole<br />

sporozoite vaccine requires the development of an in vitro sporozoite potency<br />

test, in order to ensure the complete attenuation of the vaccine. Recent results<br />

with multiply genetically-attenuated parasite lines leading to blood-<strong>infection</strong>,<br />

are a signal to scientists to take a prudent approach before administering live<br />

vaccine products to large populations.<br />

In conclusion, a more careful approach incorporating immunological research<br />

elucidating mechanisms of <strong>protection</strong> <strong>and</strong> enhancing efficacy through antigenadjuvant<br />

combinations may be warranted for the subunit AMA1 vaccine<br />

c<strong>and</strong>idate, whereas the promising efficacy results of the CPS immunisation<br />

strategy might stimulate a more pragmatic approach to develop an<br />

implementable whole-organism based vaccine. Controlled human <strong>infection</strong> trials<br />

performed in small numbers of subjects will provide a valuable tool accelerating<br />

the development of malaria vaccines.


Summary, Samenvatting, List of publications, Dankwoord, C.V. 249<br />

Samenvatting<br />

Plasmodium falciparum (Pf) malaria is één van de meest voorkomende<br />

infectieziekten en verantwoordelijk voor veel ziekte en sterfte, in het bijzonder<br />

onder jonge kinderen in het Afrika ten zuiden van de Sahara. De mens wordt<br />

geïnfecteerd met malaria parasieten door de beet van een Anopheles mug. Een<br />

effectief malaria vaccin is essentieel onderzoek om de verspreiding van malaria<br />

te voorkomen en de ziekte uit te roeien. Tientallen jaren van onderzoek hebben<br />

geleid tot de identificatie van ongeveer 36 Pf antigenen, mogelijke vaccin<br />

k<strong>and</strong>idaten. Deze antigenen worden gegroepeerd naar het tijdstip van expressie<br />

in de parasitaire levenscyclus. Pre-erythrocytaire antigenen worden tot<br />

expressie gebracht in de zogenaamde sporozoiet. Deze sporozoiten worden door<br />

muggen overgedragen, dringen de menselijke lever binnen en veroorzaken een<br />

subklinische infectie. Bloed stadium antigenen komen tot expressie gedurende<br />

de ontwikkeling van de parasiet in de menselijke rode bloedcel, waar ze de<br />

klinische ziekte met koorts, hoofdpijn en spierpijn veroorzaken. Transmissie<br />

blokkerende antigenen worden verkregen uit de seksuele stadia van de parasiet,<br />

die de levenscyclus compleet maken door zich in de volgezogen muggenmaag<br />

voort te planten.<br />

Dit proefschrift richt zich op de ontwikkeling van onderzoeksinstrumenten<br />

waarmee immunologisch humaan vaccin onderzoek naar bescherming tegen<br />

Plasmodium falciparum malaria in een vroeg stadium verricht kan worden.<br />

In sectie 1 beschrijven wij de veiligheid en immunogeniciteit van Apical<br />

Membrane Antigen 1, een veelbelovende bloed stadium vaccin k<strong>and</strong>idaat<br />

(Hoofdstuk 2). Wij hebben AMA1 gecombineerd met verschillende adjuvantia<br />

(hulpstoffen), Alhydrogel, Montanide en AS02, en vonden een verschillend<br />

bijwerkingprofiel, terwijl alle formuleringen immunogeen waren. De omvang van<br />

de humorale immuun respons werd versterkt door toevoeging van een sterker<br />

adjuvans, maar de breedte en subklasse verdeling van de antilichamen leken<br />

veel minder beïnvloed te worden door de adjuvantia (Hoofdstuk 3). Wij<br />

onderzochten de concentratie en aviditeit van antilichamen geinduceerd tegen<br />

AMA1 en vonden een omgekeerd evenredige relatie tussen deze twee<br />

parameters (Hoofdstuk 4). Omdat Fase II veld studies met verschillende AMA1<br />

formuleringen tot nu toe geen bescherming lieten zien, zal het onderzoek zich


250 Chapter 12<br />

nu wellicht moeten richten op de ontwikkeling van nieuwe adjuvantia,<br />

toedieningsmethoden en manieren om parasitaire stamverschillen te<br />

overbruggen of een combinatie van AMA1 met <strong>and</strong>ere antigenen. Dit alles in<br />

een poging om de antilichaamresponsen te versterken en veelbelovende<br />

dierexperimentele data te vertalen naar effectiviteit in mensen.<br />

Gecontroleerde humane malaria infecties (CHMI) zijn studies waarin gezonde<br />

vrijwilligers blootgesteld worden aan met malaria geïnfecteerde muggenbeten.<br />

Deze vrijwilligers worden op de voet gevolgd en beh<strong>and</strong>eld met anti-malaria<br />

middelen zodra bloedstadium parasieten onder de microscoop zichtbaar zijn.<br />

Deze studies worden in een beperkt aantal centra wereldwijd toegepast om<br />

vroegtijdig effectiviteit van voornamelijk pre-erythrocytaire vaccin k<strong>and</strong>idaten<br />

aan te tonen. In sectie 2 vergelijken wij de veiligheid en parasitologische<br />

uitkomsten van CHMI in verschillende centra en concluderen wij dat CHMI veilig<br />

verricht kunnen worden, maar in een deel van de vrijwilligers wel tot graad 3<br />

symptomen leidt (Hoofdstuk 5). De primaire parasitologische uitkomst van zulke<br />

experimenten is zeer reproduceerbaar binnen een centrum, maar kan variëren<br />

tussen onderzoekscentra. Bovendien evalueerden wij de power van CHMI om<br />

vaccin effectiviteit te kunnen meten (Hoofdstuk 6) en vonden dat CHMI een<br />

reductie in parasieten-dichtheid voor zowel pre-erythrocytaire als erythrocytaire<br />

vaccin k<strong>and</strong>idaten kan aantonen, waarmee de toepassing van CHMI in het<br />

malaria onderzoek mogelijk uitgebreid kan worden.<br />

Vervolgens verbeterden wij het CHMI protocol door het portfolio aan Pf parasiet<br />

stammen aan te vullen met een nieuwe Cambodjaanse stam NF135.C10<br />

(Hoofdstuk 7). De beschikbaarheid van deze stam maakt het mogelijk om in de<br />

toekomst immuniteit tegen verschillende stammen te evalueren. Daarnaast<br />

hebben wij levende, onverzwakte, ingevroren sporozoieten getest op hun<br />

infectiviteit door middel van intradermale injectie, en hebben bewezen dat hier<br />

mee vijf van de zes vrijwilligers uit iedere dosis groep geïnfecteerd konden<br />

worden (Hoofdstuk 8). De toediening van sporozoieten met naald en spuit heeft<br />

voordelen ten opzichte van de toediening met muggen, omdat parasieten zo<br />

nauwkeurig gedoseerd kunnen worden en de wereldwijde capaciteit voor CHMI<br />

studies nu niet meer afhankelijk is van de plaatselijke productie van<br />

geïnfecteerde muggen. Echter toekomstig onderzoek zal nog nodig zijn om met<br />

methode 100% infectiviteit te bewerkstelligen. Bovendien zal met een<br />

toenemend aantal centra dat CHMI uitvoert prioriteit gegeven moeten worden<br />

aan het st<strong>and</strong>aardiseren van studieprocedures zodat onderlinge resultaten<br />

vergelijkbaar zijn.


Summary, Samenvatting, List of publications, Dankwoord, C.V. 251<br />

In sectie 3 laten wij zien dat levende, intacte sporozoiten heel efficiënt<br />

bescherming tegen Pf malaria in mensen kunnen induceren, indien grote<br />

aantallen bloed stadium parasieten en derhalve klinische ziekte voorkomen<br />

wordt door toediening van het geneesmiddel chloroquine (CPS, Hoofdstuk 9).<br />

Chloroquine doodt bloed stadium parasieten, maar tast de leverstadium<br />

ontwikkeling niet aan. Door middel van een protocol waarbij wij vrijwilligers<br />

ma<strong>and</strong>elijks blootstelden aan de beten van infectieuze muggen terwijl zij<br />

chloroquine gebruikten, konden wij laten zien dat 10 van 10 vrijwilligers<br />

vervolgens beschermd waren tegen gecontroleerde infecties. Wij vonden een<br />

verb<strong>and</strong> tussen bescherming en een toegenomen respons van pluripotente<br />

effector memory T-cellen. Wij lieten zien dat de bescherming in vier uit zes<br />

vrijwilligers meer dan twee jaar aanhield en vonden een vertraagde ontwikkeling<br />

van parasieten in de overige twee vrijwilligers (Hoofdstuk 10). De beschreven<br />

methode is geen implementeerbaar vaccin als zodanig, maar de efficiënte<br />

inductie van bescherming suggereert wel dat vaccin strategieën gebaseerd op de<br />

gehele parasiet als antigeen heroverwogen dienen te worden. Echter, de<br />

ontwikkeling van een zogenaamd “hele parasiet” vaccin vereist de ontwikkeling<br />

van een in vitro test voor de levendigheid van sporozoieten, zodat de<br />

verzwakking van dit soort vaccins in vitro getest kan worden. Recente resultaten<br />

met meervoudig genetisch verzwakte parasieten die vervolgens bloed stadium<br />

infectie veroorzaakten in diermodellen, zijn een signaal voor onderzoekers dat<br />

men voorzichtig moet zijn met het toedienen van vaccins gebaseerd op levende<br />

parasieten aan grote populaties.<br />

Concluderend lijkt een wat terughoudende aanpak voor k<strong>and</strong>idaat vaccin AMA1<br />

op zijn plaats, met de nadruk op immunologisch onderzoek naar mechanismen<br />

van bescherming en het versterken van immuniteit door antigeen-adjuvant<br />

combinaties, terwijl de veelbelovende bescherming van het CPS model wellicht<br />

een meer pragmatische aanpak rechtvaardigt om een implementeerbaar “hele<br />

parasiet” vaccin te ontwikkelen. Gecontroleerde humane infectie studies in<br />

kleine aantallen vrijwilligers vormen een belangrijk instrument om de<br />

ontwikkeling van malaria vaccins te versnellen.


Summary, Samenvatting, List of publications, Dankwoord, C.V. 253<br />

List of publications<br />

Roestenberg M, Bijker EM, Sim BK, Billingsley PF, James ER, Bastiaens GJH,<br />

Teirlinck AC, Scholzen A, Teelen K, Arens T, van der Ven AJAM, Gunasekera A,<br />

Chakravarty S, Velmurugan S, Hermsen CC, Sauerwein RW, <strong>and</strong> Hoffman SL.<br />

Controlled human malaria <strong>infection</strong>s by intradermal injection of cryopreserved<br />

Plasmodium falciparum sporozoites. Am J Trop Med Hyg. 2012<br />

Teirlinck AC, Roestenberg M, van de Vegte-Bolmer M, Scholzen A, Heinrichs<br />

MJL, Siebelink-Stoter R, Graumans W, van Gemert GJ, Teelen K, Vos MW,<br />

Nganou-Makamdop K, Borrmann S, Rozier YPA, Erkens MAA, Luty AJF, Hermsen<br />

CC, Sim BK, van Lieshout L, Hoffman SL, Visser LG, Sauerwein RW. NF135.C10: a<br />

new Plasmodium falciparum clone for controlled human malaria <strong>infection</strong>s. J<br />

Infect Dis. 2012<br />

Nganou-Makamdop K, Ploemen I, Behet M, van Gemert GJ, Hermsen C,<br />

Roestenberg M, Sauerwein RW. Reduced Plasmodium berghei sporozoite liver<br />

load associates with low protective efficacy after intradermal immunization.<br />

Parasite Immunol. 2012 Aug 6.<br />

Remarque EJ, Roestenberg M, Younis S, Walraven V, van der Werff N, Faber BW,<br />

Leroy O, Sauerwein R, Kocken CH, Thomas AW. Humoral Immune Responses to a<br />

Single Allele PfAMA1 Vaccine in Healthy Malaria-Naïve Adults. PLoS One.<br />

2012;7(6):e38898.<br />

Roestenberg M, O'Hara GA, Duncan CJ, Epstein JE, Edwards NJ, Scholzen A, van<br />

der Ven AJ, Hermsen CC, Hill AV, Sauerwein RW. Comparison of clinical <strong>and</strong><br />

parasitological data from controlled human malaria <strong>infection</strong> trials. PLoS One.<br />

2012;7(6):e38434.<br />

Laurens MB, Duncan CJ, Epstein JE, Hill AV, Komisar JL, Lyke KE, Ockenhouse CF,<br />

Richie TL, Roestenberg M, Sauerwein RW, Spring MD, Talley AK, Moorthy VS;<br />

The Consensus Group on Design of Clinical Trials of Controlled Human Malaria<br />

Infection. A consultation on the optimization of controlled human malaria<br />

<strong>infection</strong> by mosquito bite for evaluation of c<strong>and</strong>idate malaria vaccines. Vaccine.<br />

2012 Aug 3;30(36):5302-5304.<br />

Roestenberg M, de Vlas SJ, Nieman AE, Sauerwein RW, Hermsen CC. Efficacy of<br />

preerythrocytic <strong>and</strong> blood-stage malaria vaccines can be assessed in small<br />

sporozoite challenge trials in human volunteers. J Infect Dis. 2012<br />

Aug;206(3):319-23.<br />

Teirlinck AC, McCall MB, Roestenberg M, Scholzen A, Woestenenk R, de Mast Q,<br />

van der Ven AJ, Hermsen CC, Luty AJ, Sauerwein RW. Longevity <strong>and</strong> composition<br />

of cellular immune responses following experimental Plasmodium falciparum<br />

malaria <strong>infection</strong> in humans. PLoS Pathog. 2011 Dec;7(12):e1002389.


254 Chapter 12<br />

Roestenberg M, Teirlinck AC, McCall MB, Teelen K, Makamdop KN, Wiersma J,<br />

Arens T, Beckers P, van Gemert G, van de Vegte-Bolmer M, van der Ven AJ, Luty<br />

AJ, Hermsen CC, Sauerwein RW. Long-term <strong>protection</strong> <strong>against</strong> malaria after<br />

experimental sporozoite inoculation: an open-label follow-up study. Lancet.<br />

2011 May 21;377(9779):1770-6.<br />

Sauerwein RW, Roestenberg M, Moorthy VS. <strong>Experimental</strong> human challenge<br />

<strong>infection</strong>s can accelerate clinical malaria vaccine development. Nat Rev<br />

Immunol. 2011 Jan;11(1):57-64.<br />

McCall MB, Roestenberg M, Ploemen I, Teirlinck A, Hopman J, de Mast Q, Dolo<br />

A, Doumbo OK, Luty A, van der Ven AJ, Hermsen CC, Sauerwein RW. Memorylike<br />

IFN-γ response by NK cells following malaria <strong>infection</strong> reveals the crucial role<br />

of T cells in NK cell activation by P. falciparum. Eur J Immunol. 2010<br />

Dec;40(12):3472-7.<br />

de Mast Q, de Groot PG, van Heerde WL, Roestenberg M, van Velzen JF,<br />

Verbruggen B, Roest M, McCall M, Nieman AE, Westendorp J, Syafruddin D,<br />

Fijnheer R, van Dongen-Lases EC, Sauerwein RW, van der Ven AJ.<br />

Thrombocytopenia in early malaria is associated with GP1b shedding in absence<br />

of systemic platelet activation <strong>and</strong> consumptive coagulopathy. Br J Haematol.<br />

2010 Dec;151(5):495-503.<br />

Nieman AE, de Mast Q, Roestenberg M, Wiersma J, Pop G, Stalenhoef A, Druilhe<br />

P, Sauerwein R, van der Ven A. Cardiac complication after experimental human<br />

malaria <strong>infection</strong>: a case report. Malar J. 2009 Dec 3;8:277.<br />

Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, van Gemert GJ, van de<br />

Vegte-Bolmer M, van Schaijk B, Teelen K, Arens T, Spaarman L, de Mast Q,<br />

Roeffen W, Snounou G, Rénia L, van der Ven A, Hermsen CC, Sauerwein R.<br />

Protection <strong>against</strong> a malaria challenge by sporozoite inoculation. N Engl J Med.<br />

2009 Jul 30;361(5):468-77.<br />

de Mast Q, van Dongen-Lases EC, Swinkels DW, Nieman AE, Roestenberg M,<br />

Druilhe P, Arens TA, Luty AJ, Hermsen CC, Sauerwein RW, van der Ven AJ. Mild<br />

increases in serum hepcidin <strong>and</strong> interleukin-6 concentrations impair iron<br />

incorporation in haemoglobin during an experimental human malaria <strong>infection</strong>.<br />

Br J Haematol. 2009 Jun;145(5):657-64.<br />

Roestenberg M, Remarque E, de Jonge E, Hermsen R, Blythman H, Leroy O,<br />

Imoukhuede E, Jepsen S, Ofori-Anyinam O, Faber B, Kocken CH, Arnold M,<br />

Walraven V, Teelen K, Roeffen W, de Mast Q, Ballou WR, Cohen J, Dubois MC,<br />

Ascarateil S, van der Ven A, Thomas A, Sauerwein R. Safety <strong>and</strong> immunogenicity<br />

of a recombinant Plasmodium falciparum AMA1 malaria vaccine adjuvanted<br />

with Alhydrogel, Montanide ISA 720 or AS02. PLoS One. 2008;3(12):e3960.<br />

Roestenberg M, McCall M, Mollnes TE, van Deuren M, Sprong T, Klasen I,<br />

Hermsen CC, Sauerwein RW, van der Ven A. Complement activation in


Summary, Samenvatting, List of publications, Dankwoord, C.V. 255<br />

experimental human malaria <strong>infection</strong>. Trans R Soc Trop Med Hyg. 2007<br />

Jul;101(7):643-9.<br />

Hermsen CC, Verhage DF, Telgt DS, Teelen K, Bousema JT, Roestenberg M, Bolad<br />

A, Berzins K, Corradin G, Leroy O, Theisen M, Sauerwein RW. Glutamate-rich<br />

protein (GLURP) induces antibodies that inhibit in vitro growth of Plasmodium<br />

falciparum in a phase 1 malaria vaccine trial. Vaccine. 2007 Apr 12;25(15):2930-<br />

40.<br />

Damoiseaux J, van der Ven A, Hermsen R, Telgt D, Roestenberg M, Tervaert JW,<br />

Sauerwein R. <strong>Experimental</strong> <strong>infection</strong> with Plasmodium falciparum does not<br />

result in the induction of anticardiolipin antibodies in healthy volunteers. Ann<br />

Rheum Dis. 2005 Dec;64(12):1804-5.


Summary, Samenvatting, List of publications, Dankwoord, C.V. 257<br />

Dankwoord<br />

Zoals dit proefschrift meer bloed, zweet en tranen heeft gekost dan het op het<br />

oog doet vermoeden, zo hebben ook veel meer mensen een bijdrage aan de<br />

diverse hoofdstukken geleverd dan alleen de auteurslijst aangeeft.<br />

Hoewel dit laatste hoofdstuk bedoeld is om dit recht te zetten, is dit bij voorbaat<br />

al een onmogelijke opgave. Acht jaar collegialiteit, vriendschap, lief en leed is<br />

niet te vatten in enkele pagina’s van woorden. Desalniettemin hierbij een<br />

poging.<br />

Allereerst ben ik natuurlijk enorme dank verschuldigd aan alle vrijwilligers. Het<br />

vertrouwen dat jullie mij gaven en jullie geloof in het belang van de studie was<br />

vaak overweldigend. Om maar niet te spreken over jullie talent om zelfs van de<br />

vroege ochtendbezoeken een feestje te maken en jullie doorzettingsvermogen<br />

ondanks het soms rigoureuze prikschema. Jullie zijn de echte helden van dit<br />

verhaal.<br />

Robert, het grote avontuur begon bij jou. André stelde mij aan je voor, je legde<br />

mij uit wat malaria was, en vanaf toen werden we samen meegenomen op een<br />

tumultueuze reis die promotie schijnt te heten. Ik ontwikkelde een talent om jou<br />

bij voorkeur ’s nachts of op vakantie te bellen, met immer urgente en<br />

onvoorziene ellende die á la minute opgelost dienden te worden. Op de meest<br />

cruciale momenten waren we afhankelijk van het telefoonnetwerk in Trinidad,<br />

Tanzania, Mali, of een willekeurige luchthaven ter wereld. Toch heb ik je niet<br />

één maal horen zuchten en heb ik me altijd, zelfs in die kleine uurtjes, door jou<br />

gesteund gevoeld. Het werk is nog lang niet af, en dit boekje slechts een<br />

spreekwoordelijke luchthaven, ons faciliterend in onze verdere reizen. Ik hoop<br />

dat we nog vaak samen zullen optrekken.<br />

Rob, na vele nachten of weken bikkelen kon ik altijd even bij jou binnenvallen<br />

voor goede raad. Jij, altijd geïnteresseerd in de mens achter de onderzoeker,<br />

stuurde me vaak naar huis, sprak me vaderlijk toe of nodigde zowel mij als<br />

Jeroen uit voor heerlijk eten, wat je samen met Elly bereidde. Ik waardeer je<br />

zeer, en hoop dat je ook trots bent op dit proefschrift. Het is immers een<br />

reflectie van niet alleen mijn, maar ook jouw “levenswerk”!<br />

Jorien, jij bent de personificatie van “malaria-moeder”. Je zorgde niet alleen<br />

voor alle vrijwilligers met een liefde en toewijding die grenzeloos leek, maar<br />

hebt ook mij onder je vleugels genomen. Ik mocht altijd bij jou in de keuken,<br />

achtertuin of op het CRCN even uitblazen onder het genot van een bak thee,<br />

terwijl jij me nieuwe moed gaf, alleen al door je grenzeloze geloof in het belang<br />

van het onderzoek. Zowel Jeroen als ik zijn je erg dankbaar en vinden het vooral


258 Chapter 12<br />

bijzonder dat je ook Simon in je hart gesloten hebt. We hopen dat je nog vaak bij<br />

ons in Wassenaar langs komt.<br />

Anne, wij deelden “gouden tulpen” in Leiden, leefden weken op enkele uurtjes<br />

slaap per nacht, en wisten de aspergesoep en Pavlov’s broodjes pas echt te<br />

waarderen. Nederl<strong>and</strong> vierde Koninginnedag terwijl wij prikten, pipetteerde,<br />

centrifugeerden en eindeloze hoeveelheden droogijs verslonden en onze<br />

mannen met militaire precisie vele honderden epjes beplakten, intussen jullie<br />

huwelijk bekokstovend. Je bent een geweldige collega, vriendin en onderzoeker,<br />

je weet dat ik je openheid, integriteit en eerlijkheid waardeer. Kom je nog vaak<br />

bij ons “logeren”?<br />

Matthew, na negen ma<strong>and</strong>en zwoegen, produceerden wij samen ons “gouden”<br />

kind, dat wij vervolgens, gelukkig succesvol, in een m<strong>and</strong>je op de rivier hebben<br />

losgelaten. Elke alinea kostte ons vele zondagen, op zoek naar perfectie.<br />

Gelukkig werd ons werken, koppigheid en eigenzinnigheid gewaardeerd en<br />

uiteindelijk beloond. Ik waardeer je als collega, vriend en briljant onderzoeker.<br />

Laten we hopen op nog vele “projectjes” samen!<br />

Karina, je bent haast een vanzelfsprekendheid op het lab, maar wij weten maar<br />

al te goed hoe onmisbaar je bent. Wat mij betreft ben jij de ultieme multitasker:<br />

je weet een druk gezin en gezellig sociaal leven te combineren met een baan die<br />

nauwelijks te plannen valt. Bij elke studie was je tot in de kleine uurtjes present.<br />

Bovendien ben jij de enige die foutloos drie batches extracties tegelijk kan<br />

runnen, gebruikmakend van diverse centrifuges die over verschillende labs<br />

verspreid zijn, zonder dat je humeur daar onder lijdt. Ik geloof niet dat ik dat ooit<br />

zal evenaren.<br />

Else, je kwam als geroepen toen het praktische werk van <strong>TI</strong>P2 op de drempel lag.<br />

Je ontpopte je tot een gezellige collega en stortte je vol enthousiasme in het<br />

malaria-avontuur waarbij de druk in zeer korte tijd opgevoerd werd en je<br />

soepeltjes verscheidene studies op verschillende lokaties tegelijk begeleidt.<br />

Gelukkig blijf je een ontnuchterend gevoel voor humor houden. Ik hoop nog<br />

altijd dat je je toekomstplannen wijzigt en we weer samen zullen werken ;)<br />

An-Emmie, vriendin en zeer gewaardeerde collega. Ik heb bewondering voor je<br />

flexibiliteit en durf om een <strong>and</strong>ere richting te kiezen in je leven en ben je<br />

dankbaar voor vele uren aan steun in de flexplek. Fijn dat je het naar je zin hebt<br />

bij de microbiologie en vooral super dat je eigenlijk nog altijd een “intramurale”<br />

collega bent, al is het nu op iets meer "afst<strong>and</strong>".<br />

André, ik kwam als piepjonge dokter bij je binnen en je vroeg me wat ik leuk<br />

vond: “Malaria, HIV of TB?”. Je ontdekte malaria-Meta en stelde me aan Robert<br />

voor, waarbij je me verzekerde dat ook een fase I studie “uitdagend” kan zijn.


Summary, Samenvatting, List of publications, Dankwoord, C.V. 259<br />

Dat bleek het zeker. Dank voor je bezielende klinische begeleiding, je<br />

betrokkenheid bij het welzijn van de vrijwilligers en je enthousiasme. Quirijn, als<br />

“opvolger” van André stond je aan de zijlijn om in te springen waar nodig. Je<br />

flexibiliteit is zeer gewaardeerd.<br />

Geert-Jan, Marga, Wouter en Rianne, zonder jullie was geen letter van dit<br />

proefschrift mogelijk geweest. Zonder overdrijven leveren jullie bij elke klinisch<br />

studie weer een topprestatie, die voor velen wellicht “gewoon” voelt, maar<br />

waarvan ik mij terdege bewust ben. Jullie zijn toppers van letterlijk<br />

wereldformaat!<br />

Ben, samen deelden wij vele jaren op het lab. Ik heb groot respect voor je<br />

doorzettingsvermogen en kundigheid. Maar de grootste uitdaging “is yet to<br />

come”. Count me in! Annemieke, ik kan me niet herinneren dat er ooit een<br />

moment was dat jij nog niet in Nijmegen werkte. Je bent een grote steun en<br />

toeverlaat voor mij, regelt altijd op het juiste moment de juiste zaken, niet in de<br />

laatste plaats een babybadje en luieremmer ;). Wat moet ik zonder je? Krystelle,<br />

je bent een fijne vriendin en collega, waarom moest je nou zo ver weg gaan<br />

wonen? Ivo, wat hebben we gelachen! Maar vergeet niet, als we “klaar” zijn met<br />

malaria, dan gaan we samen HIV “doen”, toch? Dunja, wat fijn om iem<strong>and</strong> te<br />

ontmoeten die blijkt te “houden” van het schuiven met papieren!<br />

Maarten, Martijn, Will, Joost, Adrian en alle <strong>and</strong>ere lab-collegae. Ook al lijkt het<br />

werk eindeloos en soms herhaaldelijk te mislukken, vergeet niet dat elk<br />

experiment vooruitgang betekent!<br />

Leo, zelden smaakte een Starbucks’ “chai” zo goed als wanneer door jou op<br />

zondagochtend op sportschoenen gebracht. Je toewijding bleek grenzeloos en je<br />

voorzag niet alleen het onderzoek in Leiden van bezielende leiding, maar<br />

suppleerde ook al mijn broodnodige voedingsstoffen met heerlijk, “fatsoenlijk<br />

Vlaams” eten. Je bent een ongelooflijk goede arts, onderzoeker en vriendelijk<br />

mens. Ik ben er trots op dat je mijn opleider bent.<br />

Steve, Kim Lee, Pete, Eric, Anusha, Tooba <strong>and</strong> the Sanaria team. I absolutely<br />

loved working with you <strong>and</strong> am proud of what we have achieved so far. I cherish<br />

the memory of us sharing turkey with Thanksgiving or breaking scooter records<br />

in an attempt to reduce transport time. I have been very lucky to enjoy many<br />

aspects of your hospitality both in Maryl<strong>and</strong> <strong>and</strong> in Holl<strong>and</strong> where I felt part of<br />

the Sanaria-family. I hope we will get a chance to work again in the future to<br />

pursue our common dream <strong>and</strong> to show you some Dutch hospitality in Leiden.<br />

T_O, jij nam met veel aanstekelijk enthousiasme het “dikke druppel team<br />

Nijmegen” onder je hoede, zorgde voor de roosters en nam bovendien vaak het<br />

leeuwendeel van de “diensten” voor je rekening. Vele nachten en weekenden


260 Chapter 12<br />

hebben wij samen de juiste kleur M&M’s opgegeten, terwijl ik wachtte op jouw<br />

oordeel. Je bent een fijne en gezellige collega!<br />

Pieter, Nanny, Chantal, Wendy, Paul, Ella en Marlou, jullie keken vele, vele<br />

dikke druppels en toch waren jullie bij elke studie weer opnieuw enthousiast en<br />

allemaal aanwezig. Jullie betrokkenheid en deskundigheid waren onmisbaar bij<br />

al dit werk. Ik hoop dat jullie met evenveel trots en plezier terugkijken als ik.<br />

Lisette, Jaco, Eric, Marjan, Meriam, Els, Jol<strong>and</strong>a, Heleen, Fons en Jeanette jullie<br />

werden plots overvallen door een rollercoaster die het klinisch malaria<br />

onderzoek heet, maar stortte je hier samen met mij vol enthousiasme in. Ik heb<br />

jullie stuk voor stuk leren kennen als zeer kundige, zorgvuldige en gezellige<br />

collega’s, en ik ben vereerd dat ik nu vaker met jullie kom te werken.<br />

Chris, Shahid <strong>and</strong> the rest of the Leiden-malaria-team, the collaboration<br />

between Leiden <strong>and</strong> Nijmegen has become quite intense in the last few years<br />

<strong>and</strong> has proven to be fruitful. I admire your expertise <strong>and</strong> was astonished to find<br />

you at the lab at the most incredible hours of the night. I hope we will continue<br />

to be in touch over the next years <strong>and</strong> hopefully also share some “daylighthours”<br />

together.<br />

Ed, Bart, Alan, Clemens en <strong>and</strong>ere BPRCers, ik heb de afgelopen jaren<br />

meermaals genoten van jullie gastvrijheid in Rijswijk, voel je welkom om eens in<br />

Leiden een biertje te komen drinken op het terras!<br />

Collega’s uit het CWZ, in het bijzonder Laura, Margreet, Pieter, Anja, Inge, jullie<br />

zijn allemaal hardwerkende, ondergewaardeerde schatten! Een steun en<br />

toeverlaat in “barre” tijden, ploeterend om de dienst/SEH/afdeling of poli te<br />

overleven. Wat zou het leuk zijn als we weer samen zouden kunnen werken!<br />

CWZ supervisoren, zonder jullie allen bij naam te noemen, draag ik jullie erfenis<br />

elke dag een klein beetje met me mee. Dank voor jullie kennis en enthousiasme<br />

om zelfs van een zeer eigenwijze arts-assistent een voorbeeldig arts te maken.<br />

Jacqueline de Graaf, jij bent een opleider pur sang. Altijd een luisterend oor<br />

paraat en altijd. Dank voor je steun en je geloof in mij, ik ben je veel dank<br />

verschuldigd.<br />

Yol<strong>and</strong>a, helaas verliet je de research om terug te gaan naar je stek bij de<br />

hematologie. Je was echter mijn grote steun en toeverlaat, een echte regelaar<br />

die nergens voor terug deinsde, aanpakte en zelfs om 6 uur ‘s ochtends een bus<br />

met “lachende” chauffeur wist te versieren om iedereen naar Nijmegen te<br />

vervoeren. Ik hoop dat ik je ooit nog eens kan verleiden om weer met me samen<br />

te werken…<br />

Kitty (en Jos), jullie flexibiliteit en hulp bij de <strong>TI</strong>P1 studie was onmisbaar. We zien<br />

elkaar op de vaccinatiepoli!


Summary, Samenvatting, List of publications, Dankwoord, C.V. 261<br />

Harm, het kostte soms wat moeite voor je om te begrijpen hoe en wat ik wilde,<br />

maar het uiteindelijke resultaat mag er zijn! Ik ben trots op wat je gebouwd<br />

hebt, en hoop ook in de toekomst op je IT ondersteuning te kunnen vertrouwen.<br />

Sake, vele telefoontjes hebben toch zijn vruchten afgeworpen! We zullen vast in<br />

de toekomst nog wel vaker bellen, als ik weer een leuk “modelmatig” projectje<br />

bedenk.<br />

Audrey, Nicole en Sylvie van de Klinische Farmacie, ondanks de soms<br />

onmogelijke papieren rompslomp, hebben jullie mij altijd op zeer professionele<br />

wijze in de studies ondersteund. Hulde!<br />

Medewerkers van het Clinical Research Center Nijmegen, ondanks mijn soms<br />

onmogelijke korte-termijn planning en ingewikkelde prik-schema’s, lukte het<br />

jullie toch altijd weer om ons te faciliteren. Bovendien wisten wij ons samen<br />

verder te professionaliseren, opdat wij kwalitatief hoogwaardig onderzoek neer<br />

konden zetten. Ik kijk terug op een vruchtbare samenwerking!<br />

Safety monitors en independent physicians, zonder jullie bij naam te noemen,<br />

ben ik jullie absoluut veel dank verschuldigd. Dank voor het vertrouwen en jullie<br />

kundige support.<br />

Pa en ma Roestenberg, Timo en Liliane, Pa en ma Mulder, Thijs en Karin, dank<br />

voor jullie eindeloze steun en begrip. Vooral natuurlijk grote dank voor pa<br />

Roestenberg, die een formidabele prestatie leverde in de layout van dit boekje.<br />

Dankzij jouw inspanningen is het niet alleen af, maar ook leesbaar.<br />

Jeroen, je bent de eerste en de laatste, mijn rots in de br<strong>and</strong>ing en liefde van<br />

mijn leven. Jij stond aan de wieg van dit proefschrift, volgde het op de voet en<br />

steunde mij onvoorwaardelijk met een onwrikbaar je vertrouwen in mij en in het<br />

onderzoek. In de afgelopen jaren werden jij en ik “wij” en stond je opnieuw aan<br />

de wieg. Samen verleggen wij stenen.


Summary, Samenvatting, List of publications, Dankwoord, C.V. 263<br />

Curriculum Vitae<br />

Meta Roestenberg was born on the 24 th of January 1981 in De Bilt, The<br />

Netherl<strong>and</strong>s. She completed bilingual highschool at the Alberdingk Thijm College<br />

in Hilversum with the international baccalaureate diploma. She studied<br />

Medicine at the University of Maastricht, where she obtained her medical<br />

degree cum laude in 2004. During her medical training Meta had the opportunity<br />

to follow internships in Africa <strong>and</strong> South-East Asia, where she became fascinated<br />

by infectious diseases. She returned to India for additional training several<br />

months after her graduation <strong>and</strong> then decided to dedicate her professional<br />

carreer to poverty related infectious diseases. Upon returning to The<br />

Netherl<strong>and</strong>s, she started malaria vaccine research at the department of Medical<br />

Microbiology of the Radboud University Medical Center. In 2006 she began with<br />

her training in internal medicine, first at the Radboud University Medical Center,<br />

later at the Canisius Wilhelmina Hospital, both in Nijmegen. In the meantime,<br />

she continued to work on her PhD, alternating between clinical work <strong>and</strong><br />

research. In 2012 she started her subspecialisation for infectious diseases at the<br />

Leiden University Medical Center. In the future, Meta hopes to continue clinical<br />

research on vaccines to fight infectious diseases that cause significant morbidity<br />

<strong>and</strong> mortality among the poorest people in the world.<br />

Meta is married Jeroen Mulder <strong>and</strong> they have one son, Simon, born in 2011.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!