ArticlePDF Available

Cytotoxic Markers Associate With Protection Against Malaria in Human Volunteers Immunized With Plasmodium falciparum Sporozoites

Authors:

Abstract and Figures

Background. Immunization of healthy volunteers by bites from Plasmodium falciparum–infected mosquitoes during chloroquine chemoprophylaxis (hereafter, chemoprophylaxis and sporozoites [CPS] immunization) induces sterile protection against malaria. CPS-induced protection is mediated by immunity against pre-erythrocytic stages, presumably at least partially by cytotoxic cellular responses. We therefore aimed to investigate the association of CPS-induced cytotoxic T-cell markers with protection. Methods. In a double-blind randomized controlled trial, we performed dose titration of CPS immunization followed by homologous challenge infection in 29 subjects. Immune responses were assessed by in vitro restimulation of peripheral blood mononuclear cells and flow cytometry. Results. Dose-dependent complete protection was obtained in 4 of 5 volunteers after immunization with bites from 45 P. falciparum–infected mosquitoes, in 8 of 9 volunteers with bites from 30, and in 5 of 10 volunteers with bites from 15 (odds ratio [OR], 5.0; 95% confidence interval [CI], 1.5–17). Completely protected subjects had significantly higher proportions of CD4 T cells expressing the degranulation marker CD107a (OR, 8.4; 95% CI, 1.5–123; P = .011) and CD8 cells producing granzyme B (OR, 11; 95% CI, 1.9–212; P = .004) after P. falciparum restimulation. Conclusions. These data underline the efficiency of CPS immunization to induce sterile protection and support a possible role for cytotoxic CD4 and CD8 T-cell responses in pre-erythrocytic immunity. Clinical Trials Registration. NCT01218893.
Cytotoxic immune responses upon in vitro Plasmodium falciparum–infected red blood cell stimulation 1 day before challenge infection (C − 1). Each symbol represents a single protected (black symbols) or chemoprophylaxis and sporozoites–immunized unprotected (gray symbols) individual from group 1 (exposed to bites from 15 infected mosquitoes on three occasions; dots), group 2 (exposed to bites from 10 infected and 5 uninfected mosquitoes on three occasions; triangles), or group 3 (exposed to bites from 5 infected and 10 uninfected mosquitoes on three occasions; squares). Horizontal bars and whiskers represent means and standard errors of the mean (SEM). Panels show CD107a+ CD4 T cells (A), CD8 T cells (B), and γδ T cells (C); granzyme B expression by CD4 T cells (D), CD8 T cells (E), and γδ T cells (F); and interferon γ (IFN-γ) expression by CD4 T cells (G), CD8 T cells (H), and γδ T cells (I). Values are corrected for uninfected red blood cell (uRBC) background and for baseline response before immunization. Mean background responses (±SEM) to uRBC stimulation for CD4, CD8, and γδ T cells were 0.19 ± 0.01, 0.41 ± 0.02, and 0.61 ± 0.05, respectively, for CD107a; 1.65 ± 0.50, 15.34 ± 1.46, and 64.56 ± 1.74, respectively, for granzyme B; and 0.09 ± 0.00, 0.07 ± 0.00, and 0.14 ± 0.01, respectively, for IFN-γ (calculated for all volunteers at both baseline and C − 1). High uRBC granzyme B responses in CD8 and γδ T cells indicate that a significant percentage of these cells contain granzyme B even in a resting situation. uRBC responses did not change significantly from baseline for any of the readouts. The differences between responses of protected and unprotected volunteers that are not indicated with a P value are nonsignificant. The differences between protected and unprotected volunteers were evaluated using logistic regression.
… 
Content may be subject to copyright.
MAJOR ARTICLE
Cytotoxic Markers Associate With Protection
Against Malaria in Human Volunteers
Immunized With Plasmodium falciparum
Sporozoites
Else M. Bijker,1,a Anne C. Teirlinck,1,a,b Remko Schats,3Geert-Jan van Gemert,1Marga van de Vegte-Bolmer,1
Lisette van Lieshout,4,5 Joanna IntHout,2Cornelus C. Hermsen,1Anja Scholzen,1Leo G. Visser,3and Robert W. Sauerwein1
1
Department of Medical Microbiology, and
2
Department for Health Evidence, Section Biostatistics, Radboud university medical center, Nijmegen,
3
Department of Infectious Diseases,
4
Department of Medical Microbiology, and
5
Department of Parasitology, Leiden University Medical Center,
The Netherlands
Background.Immunization of healthy volunteers by bites from Plasmodium falciparuminfected mosquitoes
during chloroquine chemoprophylaxis (hereafter, chemoprophylaxis and sporozoites [CPS] immunization) induces
sterile protection against malaria. CPS-induced protection is mediated by immunity against pre-erythrocytic stages,
presumably at least partially by cytotoxic cellular responses. We therefore aimed to investigate the association of
CPS-induced cytotoxic T-cell markers with protection.
Methods.In a double-blind randomized controlled trial, we performed dose titration of CPS immunization fol-
lowed by homologous challenge infection in 29 subjects. Immune responses were assessed by in vitro restimulation of
peripheral blood mononuclear cells and ow cytometry.
Results.Dose-dependent complete protection was obtained in 4 of 5 volunteers after immunization with bites
from 45 P. falciparuminfected mosquitoes, in 8 of 9 volunteers with bites from 30, and in 5 of 10 volunteers with
bites from 15 (odds ratio [OR], 5.0; 95% condence interval [CI], 1.517). Completely protected subjects had sig-
nicantly higher proportions of CD4 T cells expressing the degranulation marker CD107a (OR, 8.4; 95% CI, 1.5123;
P= .011) and CD8 cells producing granzyme B (OR, 11; 95% CI, 1.9212; P= .004) after P. falciparum restimulation.
Conclusions.These data underline the efciency of CPS immunization to induce sterile protection and support
a possible role for cytotoxic CD4 and CD8 T-cell responses in pre-erythrocytic immunity.
Clinical Trials Registration.NCT01218893.
Keywords.malaria; Plasmodium; immunization; protection; immunity; chloroquine; T cells; degranulation;
granzyme B; cytotoxicity.
Malaria remains a major public health problem, with an
estimated incidence of 207 million clinical cases and ap-
proximately 627 000 deaths every year [1]. Plasmodium
falciparum is the most severe and lethal of 5 species
that can cause malaria in humans. Availability of an
effective vaccine will be critical to ght this disease, but
currently there is no licensed vaccine available, despite
decades of research. Most efforts have focused on the de-
velopment of subunit vaccines, unfortunately showing
Received 13 February 2014; accepted 7 April 2014; electronically published 27
May 2014.
Presented in part: Scientic Spring Meeting of the Royal Netherlands Society for Mi-
crobiology and Netherlands Society for Medical Microbiology, Papendal, the Netherlands,
April 2013; Malaria Vaccines for the World Conference, Lausanne, Switzerland, April
2013; Measuring Antigen Specic Immune Responses, Dubrovnik, Croatia, May 2013;
Vaccine Symposium NWO, Utrecht, the Netherlands, September 2013.
a
E. M. B. and A. C. T. contributed equally to this work.
b
Present afliation: Center for Infectious Disease Control Netherlands, National
Institute for Public Health and the Environment, Bilthoven, the Netherlands.
Correspondence: Robert W. Sauerwein, MD, PhD, Radboud university medic al
center, Medical Microbiology 268, PO Box 9101, 6500 HB Nijmegen, the Nether-
lands (robert.sauerwein@radboudumc.nl).
The Journal of Infectious Diseases
®
2014;210:160515
© The Author 2014. Published by Oxford University Press on behalf of the Infectious
Diseases Societyof America. Thisis an Open Access article distributed under the terms
of the Creative Commons Attribution-NonCommercial-NoDerivs licence (http://
creativecommons.org/licenses/by-nc-nd/ .0/), which permits non-commercial
reproduction and distribution of the work, in any medium, provided the original work
is not altered or tra nsfor med in an y way, and that t he work is properl y cited. For
commercial re-use, please contact journals.permissions@oup.com.
DOI: 10.1093/infdis/jiu293
Markers of Protection Against Malaria JID 2014:210 (15 November) 1605
4
only limited protective efcacy [2,3]. Immunization strategies
based on whole parasites, however, have repeatedly induced
high levels of protection in experimental settings [47]. Previous-
ly, we showed that immunization of healthy, malaria-naive sub-
jects with live sporozoites delivered by 3645 mosquito bites
during chloroquine chemoprophylaxis (hereafter, chemoprophy-
laxis and sporozoites [CPS] immunization) induces robust, long-
lasting sterile protection against P. falciparum malaria [8,9]. CPS
immunization is about 20 times more efcient than the only al-
ternative approach for complete sterile protection against malaria
in humans, immunization with radiation-attenuated P. falcipa-
rum sporozoites (RAS), which requires bites from >1000 infected
and irradiated mosquitoes [4] or intravenous administration of
675 000 sporozoites [10].
CPS-induced protective immunity targets the earliest stages of
the parasite life cycle (ie, sporozoites and/or liver stages), rather
than the subsequently developing asexual blood stages [11]. The
immune pathways responsible for this pre-erythrocytic protec-
tion, however, remain unknown. In murine malaria models, cy-
totoxic killing of Plasmodium-infected hepatocytes appears to
play a role in protection, but the exact contribution and mecha-
nism of cytotoxicity remain elusive [12,13]. Also, in humans, a
role for both cytotoxic CD4 T cells and CD8 T cells has been sug-
gested, but evidence is scarce and largely circumstantial [14]. We
conducted a double-blind, randomized, controlled CPS immuni-
zation dose titration and challenge study. Subjects, while receiv-
ing chloroquine prophylaxis, were immunized by bites from 45,
30, or 15 infected mosquitoes, followed by a challenge infection,
resulting in dose-dependent protection. Next, we explored mark-
ers of cytotoxic T-cell responses induced by CPS immunization
and identied 2 cytotoxic markers associated with protection.
MATERIALS AND METHODS
Human Ethics Statement
All subjects provided written informed consent before screen-
ing. The study was approved by the Central Committee for
Research Involving Human Subjects of the Netherlands
(NL33904.091.10) and complied with the Declaration of Hel-
sinki and good clinical practice, including monitoring of data.
Clinical Trial Design and Procedures
A single-center, double-blind study was conducted at the Lei-
den University Medical Center from April 2011 until April
2012. Healthy subjects aged 1835 years with no history of ma-
laria were screened as described previously [11]. Thirty subjects
were randomly divided into 4 groups, using a computer-
generated random-number table. Subjects, investigators, and
primary outcome assessors were blinded to the allocation. All
subjects received 3 CPS immunizations at monthly intervals, as
described previously [8,11], but the number of NF54
P. falciparuminfected versus uninfected mosquitoes varied
per group: 5 subjects were each exposed to bites from 15 infect-
ed mosquitoes 3 times (group 1), 10 subjects were each exposed
to bites from 10 infected and 5 uninfected mosquitoes on three
occasions (group 2), 10 subjects were each exposed to bites from
5 infected and 10 uninfected mosquitoes on three occasions
(group 3), and 5 control subjects were exposed to bites from
15 uninfected mosquitoes on three occasions (group 4). Nine-
teen weeks after the last immunization (15 weeks after the last
chloroquine dose), all subjects were challenged by exposure to
bites from 5 mosquitoes infected with the homologous NF54 P.
falciparum strain, according to previous protocols [8,15]. The
primary outcome was prepatent period, dened as the time be-
tween challenge and rst P. falciparumpositive thick blood
smear. Thick blood smears were prepared and read as described
previously [11]. For more details about the immunization and
challenge procedures andfollow-up,seetheSupplementary
Materials.
Immunological Methods
Peripheral blood mononuclear cells (PBMCs) were collected at
the following time points: before initiation of chloroquine pro-
phylaxis (baseline [B]), 27 days after immunization 1 (I1; 1 day
before the second immunization), 27 days after immunization 2
(I2; 1 day before the third immunization), 27 days after immu-
nization 3 (I3), the day before the challenge infection (C 1),
and 20 weeks after the challenge infection (C + 140). For the as-
sessment of P. falciparumspecic immune responses, PBMCs
were restimulated in vitro with P. falciparuminfected red blood
cells (RBCs) as described before [16]. Expression of the degra-
nulation marker CD107a, the cytotoxic molecule granzyme B,
and the cytokine interferon γ(IFN-γ) by CD4, CD8, and γδ
T cells was assessed by ow cytometry. For a detailed descrip-
tion of these methods, see the Supplementary Materials.
Statistical Analysis
The dose-dependent induction of protection was tested by logistic
regression, using SPSS 20. Comparison of CD107a expression and
granzyme B and IFN-γproduction by T-cell subsets between im-
munized unprotected and protected volunteers after CPS immu-
nization was done per selectedcellular response by means of Firth
penalized logistic regression [17,18], resulting in Pvalues, odds
ratios (ORs) related to a change of 1 interquartile range (IQR),
and 95% prole likelihood condence intervals (CIs) for the
OR. Analyses were performed using R, version 3.0.1 [19], with
the packages logistf, version 1.21 [20]; rms, version 4.13[21];
and penalized, version 0.942 [22,23]. The ability of (a combina-
tion of) markers to discriminate between protected and unpro-
tected volunteers was assessed with the area under the receiver
operator curve (ROC), based on leave-one-out cross-validation,
using R and pROC, version 1.7.1 [24]. For further details about
these methods, see the Supplementary Materials.
1606 JID 2014:210 (15 November) Bijker et al
RESULTS
CPS Immunization
Thirty volunteers were included (median age, 21 years; range,
1931 years), out of 63 subjects screened for eligibility (Supple-
mentary Figure 1). Volunteers were randomly assigned to one of
the 4 groups described in Materials and Methods. After each con-
secutive immunization, the number of subjects with parasitemia,
as retrospectively detected by quantitative polymerase chain
reaction (qPCR) analysis, steadily decreased in groups 1 and
2. In group 3, however, 5 volunteers still showed parasitemia
after the second and third immunization (Figure 1). Remarkably,
in 4 immunized subjects (3 in group 2 and 1 in group 3), para-
sitemia was never detectable by qPCR. One subject from group 2
withdrew consent after the rst immunization for reasons unre-
lated to the trial and was excluded from the analysis.
Challenge Infection
Nineteen weeks after the last immunization, volunteers were
challenged by standard exposure to bites from 5 homologous
strain NF54-infected mosquitoes [5]. Protection by CPS immu-
nization was dose-dependently induced in 4 of 5 subjects in
group 1, 8 of 9 subjects in group 2, and 5 of 10 subjects in
group 3, while all control subjects became thick smear positive
(OR, 5.0; 95% CI, 1.517; P= .01). The median prepatent period
was 2.5 days longer in CPS-immunized unprotected subjects,
compared with controls, both by thick smear and qPCR. Al-
though these differences were not statistically signicant
(P= .22 for thick smear ndings and P= .31 for qPCR ndings),
this delay suggests the presence of partial protection at least in
some of the unprotected CPS-immunized subjects (Figure 2and
Table 1). In retrospect, all 6 volunteers with detectable parasite-
mia by qPCR after the third immunization were not completely
protected from challenge infection, while 17 of 18 subjects with
a negative qPCR result after the third immunization were fully
protected.
Platelet levels decreased below the reference value (150 × 10
9
platelets/L) in 8 of 12 thick-smear-positive subjects (ie, both
controls and CPS-unprotected) at any point after challenge
(median for all thick-smear-positive subjects, 134 × 10
9
plate-
lets/L;range,79×10
9
213 × 10
9
platelets/L). D-dimer levels
were elevated in all thick-smear-positive subjects after challenge
(median peak concentration, 2431 ng/mL; range, 10145000
ng/mL). Parameters normalized in all subjects after treatment
without complications. All thick-smear-positive subjects expe-
rienced solicited adverse events (AEs) during challenge
Figure 1. Parasitemia after the rst, second and third chemoprophylaxis and sporozoites immunization. Parasitemia was determined once daily by quan-
titative polymerase chain reaction (qPCR) analysis from day 6 until day 10 after each immunization. Each line represents an individual subject. Panels show
data for volunteers from group 1 (A; exposure to bites from 15 infected mosquitoes on three occasions), group 2 (B; exposure to bites from 10 infected and 5
uninfected mosquitoes on three occasions), group 3 (C; exposure to bites from 5 infected and 10 uninfected mosquitoes on three occasions), and group 4 (D;
exposure to bites from 15 uninfected mosquitoes on three occasions; control). Values shown as 10 on the log scale were negative (ie, half the lower
detection limit of the qPCR, 20 parasites/mL). Proportions along the x-axes denote subjects with a positive qPCR result/total number of subjects immunized.
Markers of Protection Against Malaria JID 2014:210 (15 November) 1607
infection consistent with uncomplicated malaria (median num-
ber/subject, 9.5 AEs [range, 414 AEs); median duration of each
AE,1.1days[range,0.012.3 days]). As expected, protected
subjects presented with fewer AEs: 15 of 17 subjects experienced
solicited AEs possibly or probably related to the challenge
(median number/subject, 2 AEs [range, 015 AEs]; median
duration, 0.7 days [range, 0.0015.9 days]). One subject from
group 2 was preliminarily treated with atovaquone/proguanil
on day 10.5 after challenge because of unrelated exertional rhab-
domyolysis after extensive sports activity (weightlifting) fol-
lowed by sauna visits. No other severe AEs occurred. One
volunteer from group 1 was treated for reasons unrelated to
the trial at day 19. Both of these volunteers remained parasite
negative by qPCR analysis after the third immunization and
at any time point after challenge and were considered protected
in further analysis.
Figure 2. Parasitemia after challenge infection. Parasitemia was assessed retrospectively by real-time quantitative PCR (qPCR) from day 5 after challenge
onward, until day 21, at 2 time points per day for thick-smear-positive volunteers and at 1 time point per day for protected volunteers. Each line represents
an individual subject. Gray dotted lines show chemoprophylaxis and sporozoitesimmunized volunteers from group 1 (exposed to bites from 15 infected
mosquitoes on three occasions; n = 5), gray dashed lines subjects from group 2 (exposed to bites from 10 infected and 5 uninfected mosquitoes on three
occasions; n = 9), gray solid lines represent subjects from group 3 (exposed to bites from 5 infected and 10 uninfected mosquitoes on three occasions;
n = 10), and black lines represent malaria-naive control subjects (exposed to bites from 15 uninfected mosquitoes on three occasions; n = 5). Values shown
as 10 on the log scale were negative. The 2 thick-smear-positive subjects from groups 1 and 2 became qPCR positive on days 9.5 and 8.5 respectively; both
became thick smear positive on day 12.0.
Table 1. Protection Against Challenge Infection After Chemoprophylaxis and Sporozoites Immunization
Group
Protection
Day of Positivity After Challenge, Median
(Range)
c
Proportion
a
Percentage (95% CI
b
) Thick Smear qPCR
Group 1 4/5 80 (36.098.0) 12.0 9.5
Group 2 8/9 89 (54.3 to >99.9) 12.0 8.5
Group 3 5/10 50 (23.776.3) 11.0 (9.015.0) 9.0 (6.513.0)
Group 4 0/5 0 (0.048.9) 9.5 (9.013.5) 6.5 (6.510.5)
Subjects were exposed to bites from 15 infected mosquitoes on th ree occasions ( group 1), to bites from 10 infected and 5 uninfected mosquit oes on three
occasions (group 2), to bites from 5 infected and 10 uninfected mosquitoes on three occasions (group 3), or to bites from 15 uninfected mosquitoes on three
occasions (group 4; control).
a
Data are no. of protected subjects/total no. of subjects.
b
95% confidence intervals (CIs) were calculated by the modified Wald method.
c
Data are for thick-smear-positive subjects.
1608 JID 2014:210 (15 November) Bijker et al
Table 2. Cytotoxic T-Cell Markers, Relative to Immunization Time, for 24 Volunteers Who Received Chemoprophylaxis and Sporozoites (CPS) Immunizations and 5 Controls
Study Group, Marker
a
CD4 T Cells CD8 T Cells γδ T Cells
BI1
P
Value I2
P
Value I3
P
Value C1
P
Value B I1
P
Value I2
P
Value I3 PC1
P
Value B I1
P
Value I2
P
Value I3
P
Value C1
P
Value
CPS
CD107a
Expression, %, mean 0.26 0.53 <.001 0.64 <.001 0.59 <.001 0.53 <.001 0.08 0.09 0.15 0.14 0.19 26.6 36.5 <.001 41.2 <.001 40.3 <.001 33.8 <.01
iMFI 15.7 38.5 <.001 46.0 <.001 41.0 <.001 37.0 <.001 7.6 14.5 19.2 <.05 17.5 <.05 20.9 <.01 4472 6925 <.001 7925 <.001 7594 <.001 6262 <.01
Granzyme B
Production, %, mean 0.25 0.49 0.41 0.38 0.43 0.50 0.89 0.92 1.03 0.94 6.06 7.35 8.27 8.96 8.79
iMFI 1.40 9.80 11.3 7.50 3.24 5.11 71.3 <.05 40.5 26.7 19.9 2.54 15.3 <.001 15.0 <.001 12.4 <.001 8.18
IFN-γ
Production, %, mean 0.08 0.43 <.001 0.54 <.001 0.42 <.001 0.47 <.001 0.04 0.11 0.13 <.05 0.08 0.13 <.01 6.01 13.6 <.001 16.3 <.001 14.9 <.001 12.0 <.01
iMFI 5.16 32.2 <.001 37.7 <.001 25.7 <.05 37.2 <.001 2.96 9.8 9.3 6.4 12.5 <.01 407 1020 <.01 1173 <.001 1029 <.01 982 <.05
Control
CD107a
Expression, %, mean 0.25 0.27 0.16 0.20 0.42 0.02 0.06 0.02 0.00 0.05 26.3 22.6 22.6 21.1 29.2
iMFI 15.6 17.2 10.6 13.4 27.3 3.38 7.04 3.28 1.56 7.92 4438 4439 4191 4512 5608
Granzyme B
Production, %, mean 0.31 0.18 0.20 0.10 0.60 0.95 0.43 0.96 0.72 1.16 5.68 2.41 1.00 1.73 8.56
iMFI 1.06 4.04 2.98 0.36 11.1 23.2 3.02 14.6 25.2 25.3 45.1 77.4 258 326 302
IFN-γ
Production, %, mean 0.09 0.02 0.00 0.02 0.12 0.02 0.02 0.01 0.01 0.07 5.58 3.51 3.40 3.13 7.06
iMFI 4.62 0.86 0.36 0.70 6.98 1.44 0.38 0.22 0.26 5.44 356 161 146 143 486
Plasmodium falciparuminfected red blood cell (RBC)specific responses were corrected for the value for the uninfected RBC background. Pvalues were calculated using 1-way analysis of variance with the Dunnett post
hoc test, with the B value serving as a control.
Abbreviations: B, baseline; C 1, 1 day before challenge; I1, 27 days after immunization 1; I2, 27 days after immunization 2; I3, 27 days after immunization 3.
a
The geometric mean fluorescence intensity (iMFI) is calculated as the percentage of positive cells multiplied by the MFI of this positive population.
Markers of Protection Against Malaria JID 2014:210 (15 November) 1609
Analysis of Cytotoxic T-Cell Markers After In Vitro
P. falciparum Stimulation
Next, we tested a panel of representative cytotoxic T-cell mark-
ers, including surface expression of degranulation marker
CD107a and granzyme B and IFN-γproduction, in CD4,
CD8, and γδ T cells after in vitro restimulation with P. falcipa-
ruminfected RBCs in all immunized subjects (Table 2). CPS im-
munization induced a signicant increase in both the percentage
and integrated geometric mean uorescence intensity (iMFI) of
CD107a
+
CD4 and γδ T cells from the rst immunization until
challenge. Similarly, CD8 T cells expressed a signicantly higher
CD107a iMFI after the second immunization. The proportion of
granzyme Bproducing cells did not change after immunization,
but the granzyme B iMFI was signicantly increased in both CD8
and γδ T cells, returning to baseline on C 1. Production of IFN-
γwas induced in all T-cell subsets, but induction was most
pronounced in CD4 and γδ T cells. There were only weak corre-
lations between cellular responses on C 1 and total blood-stage
parasite exposure, as calculated by the number of parasites per
milliliter after all 3 immunizations (data not shown; Spearman
rho for all, <0.5). None of the responses in the control group
changed signicantly from baseline at any point of time (Table 2),
suggesting that chloroquine alone did not affect P. falciparum
specic T-cell responses.
We next assessed the association of these markers with pro-
tection after challenge (Figure 3). Indeed, complete protection
associated with the proportion of CD107a
+
CD4 T cells (OR,
8.4; 95% CI, 1.5123; P=.011; Figure 3A), the iMFI of
CD107a on CD4 T cells (OR, 11; 95% CI, 1.6188; P= .011;
data not shown), and production of granzyme B by CD8
T cells (OR, 11; 95% CI, 1.9212; P= .004; Figure 3E)at
C1. A subgroup analysis of data from group 3 conrmed
these ndings: the only markers with higher levels in protected
subjects were the proportion of both CD107a
+
CD4 T cells (OR,
4.2; 95% CI, .9140; P= .081) and granzyme Bproducing CD8
T cells (OR = 27; 95% CI, 1.527 687; P= .019). While expres-
sion of CD107 on CD4 T cells and granzyme B in CD8 T cells
predicted protection, with areas under the ROC of 0.73 (95% CI,
.48.98) and 0.81 (95% CI, .63.99), respectively, combining
both markers resulted in only a slight improvement of the
area under the ROC (0.82; 95% CI, .611).
P. falciparumspecic IFN-γproduction by CD4, CD8, or γδ
T cells could not distinguish protected volunteers (Figure 3G,
3H, and 3I). Also pluripotent (IFN-γ
+
IL-2
+
) effector memory
T-cell (CD4
+
CD62L
CD45RO
+
) responses, previously shown
to be signicantly increased by CPS immunization [8], were
again induced (P= .013) but did not differentiate between pro-
tected and unprotected volunteers (OR, 1.6; 95% CI, .54.9;
P= .41; data not shown).
CD107a
+
CD4 T cells presented as the clearest marker asso-
ciated with protection, with values consistently higher in fully
protected subjects from I1 onward (Figure 4A), and was
independent of immunization dose (Figure 4B). A signicant
correlation was found between CD107a expression by CD4
T cells after 1 immunization and prepatent period after chal-
lenge infection in all thick-smear-positive subjects (Spearman
rho, 0.69; P= .013; Figure 4C). The proportion CD107a
+
CD4
T cells in the control subject who developed parasitemia signi-
cantly later than the other controls (ie, day 13.5 vs days 910.5)
was, at baseline, on average 2.8-fold higher than in the other
subjects. Possibly, the inherently higher response in this volun-
teer contributed to delayed patency after challenge.
CD107a
+
CD4 T cells expressed proportionally more gran-
zyme B (7.4% vs 0.39% on C 1; P< .0008) in protected subjects,
indicative of their cytotoxic phenotype, and proportionally more
IFN-γ(13.3% vs 0.39% on C 1; P< .0001) than CD107a
CD4
T cells (Figure 4Dand 4E). CD8 T cells, traditionally considered
the cytotoxic subclass of T cells, contained a larger proportion of
CD107a
+
cells at baseline than CD4 T cells when uninfected
(0.39% vs 0.19%; P< .0001 [all volunteers]). However, the pro-
portion of P. falciparumspecic degranulation of CD8 T cells
was not notably increased by CPS immunization (P= .44), in
contrast to CD4 T cells (P<.0001; Supplementary Figure 2A
and 2B).
Both CD107a expression by CD4 T cells and granzyme B
production by CD8 T cells remained signicantly elevated up
to 20 weeks after the challenge infection (C + 140; P< .05 and
P< .01, respectively; Figure 5Aand 5B), demonstrating longev-
ity of the CPS-induced T-cell response.
DISCUSSION
We show that CPS immunization reproducibly and dose-
dependently induces protection against a homologous
challenge infection. With exposure to a total number of P. fal-
ciparuminfected mosquito bites as low as 30, CPS immuniza-
tion still induces 89% protection in healthy volunteers. We
furthermore demonstrate that markers of cytotoxic T-cell
responses are associated with protection against malaria after
whole-sporozoite immunization.
This study provides further support for the remarkable po-
tency of the CPS-protocol to induce complete protection by
using even lower numbers of P. falciparuminfected mosquitoes
than before [8]. The observed dose-dependent protection is in
line with results from RAS immunization trials with sporozoites
administered either intravenously by needle and syringe [10]or
by bites from irradiated infected mosquitoes [4]. Although the
delay of patency in unprotected CPS-immunized subjects was
not statistically signicant, the patterns of parasitemia indicate
partial protection in some subjects. The unexpectedly delayed
control subject hampered statistical signicance but could be
considered an outlier, possibly because of the inherently high
baseline immune response. The establishment of a suboptimal
CPS immunization regimen inducing protection in 50% of the
1610 JID 2014:210 (15 November) Bijker et al
volunteers immunized with bites from 5 mosquitoes on three
occasions will facilitate further studies of protective immune
mechanisms against P. falciparum malaria.
Our data provide evidence for a role of cytotoxic T-cell re-
sponses in pre-erythrocytic immunity in humans. Because of
obvious practical limitations, we only assessed immune cells
Figure 3. Cytotoxic immune responses upon in vitro Plasmodium falciparuminfected red blood cell stimulation 1 day before challenge infection (C 1).
Each symbol represents a single protected (black symbols) or chemoprophylaxis and sporozoitesimmunized unprotected (gray symbols) individual from
group 1 (exposed to bites from 15 infected mosquitoes on three occasions; dots), group 2 (exposed to bites from 10 infected and 5 uninfected mosquitoes
on three occasions; triangles), or group 3 (exposed to bites from 5 infected and 10 uninfected mosquitoes on three occasions; squares). Horizontal bars and
whiskers represent means and standard errors of the mean (SEM). Panels show CD107a
+
CD4 T cells (A), CD8 T cells (B), and γδ T cells (C); granzyme B
expression by CD4 T cells (D), CD8 T cells (E), and γδ T cells (F); and interferon γ(IFN-γ) expression by CD4 T cells (G), CD8 T cells (H), and γδ T cells (I).
Values are corrected for uninfected red blood cell (uRBC) background and for baseline response before immunization. Mean background responses (±SEM) to
uRBC stimulation for CD4, CD8, and γδ T cells were 0.19 ± 0.01, 0.41 ± 0.02, and 0.61 ± 0.05, respectively, for CD107a; 1.65 ± 0.50, 15.34 ± 1.46, and
64.56 ± 1.74, respectively, for granzyme B; and 0.09 ± 0.00, 0.07 ± 0.00, and 0.14± 0.01, respectively, for IFN-γ(calculated for all volunteers at both baseline
and C 1). High uRBC granzyme B responses in CD8 and γδ T cells indicate that a signicant percentage of these cells contain granzyme B even in a resting
situation. uRBC responses did not change signicantly from baseline for any of the readouts. The differences between responses of protected and unpro-
tected volunteers that are not indicated with a Pvalue are nonsignicant. The differences between protected and unprotected volunteers were evaluated
using logistic regression.
Markers of Protection Against Malaria JID 2014:210 (15 November) 1611
in the peripheral blood, which may not necessarily reect
responses in the liver but rather represent a surrogate. The re-
sults of this exploratory analysis will have to be conrmed in
future trials, and the functional relevance remains to be
investigated.
So-called classic cytotoxic CD8 T cells can be activated by
malaria parasite antigen on infected hepatocytes via major his-
tocompatibility complex (MHC) class I [25] and are associated
with protection in a number of (animal) models [13,14,26].
CD8 T cells are involved in protection in the murine CPS and
RAS models [2729], but their precise effector mechanisms re-
main subject of debate. They might either require direct contact
with infected hepatocytes [13] or be indep endent of granzyme B
and/or other cytotoxic molecules, suggesting a more indirect cy-
tokine-mediated effect by CD8 T cells [12] or other hepatic im-
mune cells [30]. In additi on, a functional role for cytotoxic CD4
T cells is also conceivable because these cells can use cytolytic
pathways, such as those involving granulysin, perforin,
Figure 4. Cytotoxic prole of chemoprophylaxis and sporozoites (CPS)induced CD4 T cells. Aand B, Induction of Plasmodium falciparumspecic
CD107a
+
CD4 T cells was determined in protected and unprotected CPS-immunized subjects (A) and in protected subjects separated for each immunization
dose (B) over the course of immunization. Horizontal bars and whiskers represent mean responses and standard errors of the mean. C, The relationship
between P. falciparumspecic CD107a CD4 T cells on day 27 after immunization 1 (I1) and the prepatent period after challenge for all thick-smear-positive
volunteers (CPS-immunized individuals and controls). Dand E, Among protected CPS-immunized subjects, granzyme B (D) and interferon γ(IFN-γ) production
(E) by CD107a
+
(black dots) and CD107a
(gray dots) CD4 T cells were analyzed at baseline (B) and after CPS immunization (on the day before the challenge
infection [C 1]). Horizontal bars show the mean response. All data were corrected for uninfected red blood cell background for every volunteer at each time
point. *P< .05, **P< .01, and ***P< .001. I2, 27 days after immunization 2; I3, 27 days after immunization 3.
1612 JID 2014:210 (15 November) Bijker et al
granzymes, and FAS-L, as shown mostly in viral infections [31,
32]. The protective role of CD4 T cells in murine malaria has
been suggested, using in vitro experiments [33]andin vivo
depletion [12] or passive transfer [34].Furthermore, functional cy-
totoxic CD4 T cells derived from RAS-immunized or synthetic-
peptide-immunized volunteers are able to lyse autologous B
cells pulsed with a peptide from the circumsporozoite protein
[3537]. We used surface expression of CD107a (LAMP-1), a
marker for cytotoxic degranulation, to phenotypically identify cy-
totoxic CD4 T cells [31]. To directly kill a P. falciparuminfected
hepatocyte, parasite antigens should be presented in the context of
MHC class II to the cytotoxic CD4 T cells. Although hepatocytes
do not express MHC class II in noninammatory circumstances,
the presence of MHC class II on human hepatocytes has been
shown in a small number of patients with chronic hepatitis [38]
and immune-mediated liver disorders [39,40]. Functionally, over-
expression of MHC class II on hepatocytes in a transgenic mice
model showed their capacity for costimulation, antigen presenta-
tion, and CD4 T-cell activation [41]. Only indirect evidence sug-
gests that MHC class II expression on mice hepatocytes may play
a role in murine malaria [33,42], and the presence of MHC class
II on hepatocytes in human malaria has never been studied. Here,
we show for the rst time that degranulating CD4 T cells are as-
sociated with protection in human malaria and are signicantly
induced after 1 immunization.
The observed lack of boosting by the second and third immu-
nizations may reect a saturated response of antigen-specic
memory cells. This raises the possibility that fewer immuniza-
tions may be sufcient to induce protection, supported by the
increased proportion of volunteers without parasitemia after the
second and third immunizations in groups 1 and 2. Moreover,
the observed longevity of the immune response is in line with
long-term protection after CPS immunization in a previous
study [9].
The T-helper type 1 (Th1) cytokine IFN-γhas been repeat-
edly shown to be an important effector molecule in protection
against malaria parasites [43], and the clear induction of Th1
responses in our study corroborates earlier ndings in both an-
imals and humans after whole-sporozoite immunization [8,10,
12,26,27]. We previously showed that a broad range of both in-
nate and adaptive cellular subsets contribute to CPS-induced
P. falciparumspecicIFN-γproduction [16], which is sus-
tained at least up to 2.5 years after immunization [9].IFN- γpro-
duction alone, however, does not correlate with protection in
the RAS model [10] or our CPS model. Also, production of
both IFN-γand IL-2 by effector memory CD4 T cells and pro-
duction of IFN-γby γδ T cells, although clearly increased in im-
munized volunteers [8,16], were not different between
protected and unprotected volunteers.
During CPS immunization, 4 protected subjects did not show
parasitemia by qPCR at any measured time point, not even after
the rst immunization. A possible explanation is that the num-
ber of merozoites released from the liver is too low for qPCR
detection. A strong primary innate immune response may be
responsible for clearing sporozoites and/or killing infected he-
patocytes upon rst encounter. Previous studies in mice indeed
showed that the inammatory cytokines interleukin 1 (IL-1)
and interleukin 6 (IL-6) block pre-erythrocytic development
in mice [16,44]. Alternatively, chloroquine may have contribut-
ed to the decreased (ie undetectable) number of parasites re-
leased from the liver either by direct killing or, indirectly, by
stimulating the immune system.
Antigen recognition and immune cell activation are essential
for an effective response. To investigate pre-erythrocytic cellular
Figure 5. Longevity of cellular immune responses after chemoprophylaxis and sporozoites (CPS) immunization. Plasmodium falciparumspecic cellular
immune responses (corrected for uninfected red blood cell background) were assessed in protected (black dots) and unprotected (gray squares) CPS-
immunized volunteers before CPS immunization (B), and before (C 1) and 20 weeks after (C + 140) challenge infection. Data are shown as mean ± standard
error of the mean for CD107a expression on CD4 T cells (A) and granzyme B production by CD8 T cells (B). Tests are performed separately for protected and
immunized unprotected volunteers, by repeated-measures analysis of variance (including all time points before and after immunizations) and the Dunnett
multiple comparison post test, using B as control column. Only the test results for C + 140, compared with those for baseline, for protected volunteers are
displayed. For immunized unprotected volunteers, all results were nonsignicant. *P< .05 and **P< .01.
Markers of Protection Against Malaria JID 2014:210 (15 November) 1613
immune responses, stimulation with cultured liver-stage P. fal-
ciparum would be preferred, but this is currently impossible.
We therefore used asexual blood-stage parasites for our experi-
ments, and although responses to purely pre-erythrocytic anti-
gens may be missed, the majority of potential memory
responses are likely detected upon P. falcip aru minfected
RBC stimulation, given the large overlap between liver-stage
and blood-stage antigens [45]. Future antigen screening by
stimulation with a comprehensive library of pre-erythrocytic
and cross-stage proteins or peptides, and subsequent functional
studies focusing on cytotoxic T cells, will further identify and
delineate the specicity of protective responses [33,46].
In conclusion, we identied 2 in vitro cellular cytotoxic im-
mune markers that are associated with protection against ma-
laria in a controlled clinical setting. Furthermore, this study
conrms the robustness of CPS immunization as a highly ef-
cient and reproducible immunization strategy for complete ho-
mologous protection. Further exploration of immune responses
induced by CPS immunization will make important contribu-
tions to pre-erythrocytic malaria vaccine development and clin-
ical testing.
Supplementary Data
Supplementary materials are available at The Journal of Infectious Diseases
online (http://jid.oxfordjournals.org/). Supplementary materials consist of
data provided by the author that are published to benet the reader. The
posted materials are not copyedited. The contents of all supplementary
data are the sole responsibilityof the authors. Questions or messages regard-
ing errors should be addressed to the author.
Notes
Acknowledgments. We thank all of the trial volunteers, for their partic-
ipation in this study; K. Suijk-Benschop, J. Fehrmann-Naumann, C. Prins,
E. Jonker, G. Hardeman, and S. ten Velden-Schipper, for blood collection
and their good care of the volunteers; the LUMC Department of Medical
Microbiology, for facilitating parasitological diagnosis; M. Erkens,
T. Arens, J. van der Slot, H. Gerritsma, F. van de Sande, J. van Schie,
E. Brienen, J. Schelfaut, J. Verweij, J.Kromhout, E. van Oorschot, and
M. Beljon, for reading many thick smears; C. Janse, for his unlimited hos-
pitality;M.Bootsma,forhercardiacmonitoringofthetrialvolunteers;
W. Graumans and R. Siebelink-Stoter, for culturing parasites; J. Klaassen,
L. Pelser-Posthumus, J. Kuhnen, and A. Pouwelsen, for generating infected
mosquitoes and for assistance with immunization of and providing chal-
lenge infection to the volunteers; G. Bastiaens and J. Wiersma, for assistance
with the immunizations and the challenge infections; P. Houzé, for chloro-
quine measurements; members of the Safety Monitoring Committee
J. A. Romijn, M. de Boer, and M. Laurensfor their participation, guidance,
and safety recommendations throughout the trial; J. Verweij, for the set up
and supervision of the DNA isolation and qPCR activities; E. Brienen, for
performing DNA isolation and qPCR activities; CromSource, for clinical
monitoring; the staff from the LUMC Central Clinical Biochemistry and
Haematology Laboratories and the staff from the LUMC Pharmacy, for
making this study possible; A. Jansens, and D. Zimmerman, for their ad-
ministrative support; K. Teelen, for help with sample logistics; M. Vos
and B. van Schaijk, for molecular analysis of the parasite batches; and
W. Nahrendorf, for critically reading the manuscript.
Financial support. This work was supported by The Netherlands Or-
ganisation for Health Research and Development (ZonMw) (project
95110086), the Dioraphte Foundation (project 12010100), and a European
Malaria Vaccine Development Association fellowship (to A.C.T.). The re-
search leading to these results has furthermore received funding from the
European Union Seventh Framework Programme (FP7/2007-2013; grant
242095 [EviMalar]).
Potential conicts of interest. All authors: No reported conicts.
All authors have submitted the ICMJE Form for Disclosure of Potential
Conicts of Interest. Conicts that the editors consider relevant to the con-
tent of the manuscript have been disclosed.
References
1. WHO. World malaria report 2013. WHO, 2013.
2. Crompton PD, Pierce SK, Miller LH. Advances and challenges in ma-
laria vaccine development. J Clin Invest 2010; 120:416878.
3. RTSS Clinical Trials Partnership, Agnandji ST, Lell B, et al. A phase 3
trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med
2012; 367:228495.
4. Hoffman SL, Goh LM, Luke TC, et al. Protection of humans against ma-
laria by immunization with radiation-attenuated Plasmodium falcipa-
rum sporozoites. J Infect Dis 2002; 185:115564.
5. Sauerwein RW, Roestenberg M, Moorthy VS. Experimental human
challenge infections can accelerate clinical malaria vaccine develop-
ment. Nat Rev Immunol 2011; 11:5764.
6. McCarthy JS, Good MF. Whole parasite blood stage malaria vaccines: a
convergence of evidence. Hum Vaccin 2010; 6:11423.
7. Butler NS, Vaughan AM, Harty JT, Kappe SH. Whole parasite vaccina-
tion approaches for prevention of malaria infection. Trends Immunol
2012; 33:24754.
8. Roestenberg M, McCall M, Hopman J, et al. Protection against a malaria
challenge by sporozoite inoculation. N Engl J Med 2009; 361:46877.
9. Roestenberg M, Teirlinck AC, McCall MBB, et al. Long-term protection
against malaria after experimental sporozoite inoculation: an open-label
follow-up study. Lancet 2011; 377:17706.
10. Seder RA, Chang LJ, Enama ME, et al. Protection against malaria by
intravenous immunization with a nonreplicating sporozoite vaccine.
Science 2013; 341:135965.
11. Bijker EM, Bastiaens GJ, Teirlinck AC, et al. Protection against malaria after
immunization by chloroquine prophylaxis and sporozoites is mediated by
preerythrocytic immunity. Proc Natl Acad Sci U S A 2013; 110:78627.
12. Doolan DL, Hoffman SL. The complexity of protective immunity
against liver-stage malaria. J Immunol 2000; 165:145362.
13. Overstreet MG, Cockburn IA, Chen YC, Zavala F. Protective CD8
T cells against Plasmodium liver stages: immunobiology of an
unnaturalimmune response. Immunol Rev 2008; 225:27283.
14. Tsuji M. A retrospective evaluation of the role of T cells in the develop-
ment of malaria vaccine. Exp Parasitol 2010; 126:4215.
15. Verhage DF, Telgt DS, Bousema JT, et al. Clinical outcome of experi-
mental human malaria induced by Plasmodium falciparum-infected
mosquitoes. Neth J Med 2005; 63:528.
16. Teirlinck AC, McCall MBB, Roestenberg M, et al. Longevity and com-
position of cellular immune responses following experimental Plasmo-
dium falciparum malaria infection in humans. PLoS Pathog 2011;7:
e1002389.
17. Firth D. Bias reduction of maximum likelihood estimates. Biometrika
1993; 80:2738.
18. Heinze G. A comparative investigation of methods for logistic regression
with separated or nearly separated data. Stat Med 2006;25:421626.
19. R Foundation for Statistical Computing. R: A language and environment
for statistical computing. Vienna, Austria. http://www.R-project.org/.
20. Heinze G, Ploner M, Dunkler D, Southworth H. logistf: Firths bias re-
duced logistic regression. R package version 1.21, 2013.http://CRAN.R-
project.org/package=logistf. Accessed 2 April 2014.
21. Harrell FE Jr. rms: Regression Modeling Strategies. R package version
4.13, 2014.http://CRAN.R-project.org/package=rms. Accessed 2
April 2014.
1614 JID 2014:210 (15 November) Bijker et al
22. Goeman JJ. L-1 Penalized Estimation in the Cox Proportional Hazards
Model. Biom J 2010; 52:7084.
23. Goeman JJ. Penalized R package, version 0.942, 2012.
24. Robin X, Turck N, Hainard A, et al. pROC: an open-source package for
R and S+ to analyze and compare ROC curves. BMC Bioinformatics
2011; 12:77.
25. Ma J, Trop S, Baer S, et al. Dynamics of the major histocompatibility
complex class I processing and presentation pathway in the course of
malaria parasite development in human hepatocytes: implications for
vaccine development. PLoS One 2013; 8:e75321.
26. Epstein JE, Tewari K, Lyke KE, et al. Live attenuated malariavaccine de-
signed to protect through hepatic CD8(+) T cell immunity. Science
2011; 334:47580.
27. Belnoue E, Costa FT, Frankenberg T, et al. Protective T cell immunity
against malaria liver stage after vaccination with live sporozoites under
chloroquine treatment. J Immunol 2004; 172:248795.
28. Nganou-Makamdop K, van Gemert GJ, Arens T, Hermsen CC, Sauer-
wein RW. Long term protection after immunization with P. berghei spo-
rozoites correlates with sustained IFNgamma responses of hepatic
CD8+ memory T cells. PLoS One 2012; 7:e36508.
29. Cockburn IA, Amino R, Kelemen RK, et al. In vivo imaging of CD8+
T cell-mediated elimination of malaria liver stages. Proc Natl Acad Sci
USA2013; 110:90905.
30. Kimura K, Kimura D, Matsushima Y, et al. CD8+ T cells specic for a
malaria cytoplasmic antigen form clusters around infected hepatocytes
and are protective at the liver stage of infection. Infect Immun 2013;
81:382534.
31. Marshall NB, Swain SL. Cytotoxic CD4 T cells in antiviral immunity.
BioMed Research International 2011; 2011:18.
32. Brown DM. Cytolytic CD4 cells: direct mediators in infectious disease
and malignancy. Cell Immunol 2010; 262:8995.
33. Renia L, Marussig MS, Grillot D, et al. In vitro activity of CD4+ and
CD8+ T lymphocytes from mice immunized with a synthetic malaria
peptide. Proc Natl Acad Sci U S A 1991; 88:79637.
34. Tsuji M, Romero P, Nussenzweig RS, Zavala F. CD4+ cytolytic T cell
clone confers protection against murine malaria. J Exp Med 1990;
172:13537.
35. Moreno A, Clavijo P, Edelman R, et al. Cytotoxic Cd4+ T-Cells from a
Sporozoite-Immunized Volunteer Recognize the Plasmodium falcipa-
rum Cs Protein. Int Immunol 1991; 3:9971003.
36. Frevert U, Nardin E. Cellular effector mechanisms against Plasmodium
liver stages. Cell Microbiol 2008; 10:195667.
37. Frevert U, Moreno A, Calvo-Calle JM, Klotz C, Nardin E. Imaging effec-
tor functions of human cytotoxic CD4+ T cells specicforPlasmodium
falciparum circumsporozoite protein. Int J Parasitol 2009;39:11932.
38. Franco A, Barnaba V, Natali P, Balsano C, Musca A, Balsano F. Expres-
sion of class I and class II major histocompatibility complex antigens on
human hepatocytes. Hepatology 1988; 8:44954.
39. Senaldi G, Lobo-Yeo A, Mowat AP, Mieli-Vergani G, Vergani D. Class
I and class II major histocompatibility complex antigens on hepa-
tocytes: importance of the method of detection and expression
in histologically normal and diseased livers. J Clin Pathol 1991;44:
10714.
40. Lobo-Yeo A, Senaldi G, Portmann B, Mowat AP, Mieli-Vergani G,
Vergani D. Class I and class II major histocompatibility complex anti-
gen expression on hepatocytes: a study in children with liver disease.
Hepatology 1990; 12:22432.
41. Herkel J, Jagemann B, Wiegard C, et al. MHC class II-expressing hepa-
tocytes function as antigen-presenting cells and activate specicCD4
T lymphocytes. Hepatology 2003; 37:107985.
42. Renia L, Grillot D, Marussig M, et al. Effector functions of circumspor-
ozoite peptide-primed CD4+ T cell clones against Plasmodium yoelii
liver stages. J Immunol 1993; 150:14718.
43. McCall MB, Sauerwein RW. Interferon-gammacentral mediator of
protective immune responses against the pre-erythrocytic and blood
stage of malaria. J Leukoc Biol 2010; 88:113143.
44. Vreden SG, van den Broek MF, Oettinger MC, Verhave JP, Meuwissen
JH, Sauerwein RW. Cytokines inhibit the development of liver schizonts
of the malaria parasite Plasmodium berghei in vivo. Eur J Immunol
1992; 22:22715.
45. Tarun AS, Peng X, Dumpit RF, et al. A combined transcriptome and
proteome survey of malaria parasite liver stages. Proc Natl Acad Sci
USA2008; 105:30510.
46. Doolan DL, Southwood S, Freilich DA, et al. Identication of Plasmo-
dium falciparum antigens by antigenic analysis of genomic and prote-
omic data. Proc Natl Acad Sci U S A 2003; 100:99527.
47. Adegnika AA, Verweij JJ, Agnandji ST, et al. Microscopic and sub-
microscopic Plasmodium falciparum infection, but not inammation
caused by infection, is associated with low birth weight. Am J Trop
Med Hyg 2006; 75:798803.
48. Lejeune D, Souletie I, Houze S, et al. Simultaneous determination of
monodesethylchloroquine, chloroquine, cycloguanil and proguanil on
dried blood spots by reverse-phase liquid chromatography. J Pharm
Biomed Anal 2007; 43:110615.
Markers of Protection Against Malaria JID 2014:210 (15 November) 1615
... Exposures to organisms during immunization phases are not generally considered to be challenge exposures, such that many CHMI studies do not report whether volunteers develop parasitemia during such phases. However, Bijker et al. [17] retrospectively detected parasitemia in volunteers during immunization phases, demonstrating that volunteers are in fact susceptible to infection while undergoing drug prophylaxis. Although immunization and other non-natural, non-challenge contexts involving intentional exposure to an organism are not done with the purpose of emulating a model of infection, they are still exposures, and should be evaluated and regulated accordingly. ...
... 2. Studies should report the frequency and severity of infections for any context in which human volunteers are exposed to infectious disease organisms, including attenuated pathogens and live virus vaccine candidates. Table 1 presents a completed template using data reported in Bijker et al. [17] as an example of how the minimum data that should be collected in the course of a CHIS should be reported in order to comply with these recommendations. ...
... When challenge-related SAEs or unexpected AEs do occur, they should be reported in as much detail as possible, and publishing separate case reports to describe them sufficiently should be seriously considered. Table 2 presents a completed template using data reported in Bijker et al. [17] as an example of how the minimum data that should be collected in the course of a CHIS should be reported in order to comply with these recommendations. ...
... However, the development of a CD4 + T FH cell response that promotes effective and long-lasting parasite-specific protective Ab results from multiple competing mechanisms orchestrating CD4 + T cell differentiation during infection. Malaria infections, both in humans and in mouse models, drive robust T helper 1 (T H 1) responses as a result of a highly inflammatory environment, in which CD4 + T cells upregulate the T H 1 master transcriptional regulator T-bet, produce IFNγ, and have cytolytic potential [40][41][42] , all of which contribute to protective antimalarial responses 43 . These T cells also express high levels of cell-surface CXCR3 and have poor T FH cell functional characteristics 9,44,45 . ...
... In these reports, the CD4 + T cell cytolytic clones were isolated from the animal model of malaria or human participants that received irradiated sporozoite immunizations. The presence of parasite (CSP)-specific CD4 + T cells with cytolytic activity was more recently confirmed in individuals inoculated with irradiated or live sporozoite immunizations 41,42,74 . To our knowledge, however, no studies have reported the presence of such cells in endemic area residents who have developed immunity against P. falciparum. ...
Article
Full-text available
Clinical immunity against Plasmodium falciparum infection develops in residents of malaria endemic regions, manifesting in reduced clinical symptoms during infection and in protection against severe disease but the mechanisms are not fully understood. Here, we compare the cellular and humoral immune response of clinically immune (0-1 episode over 18 months) and susceptible (at least 3 episodes) during a mild episode of Pf malaria infection in a malaria endemic region of Malawi, by analysing peripheral blood samples using high dimensional mass cytometry (CyTOF), spectral flow cytometry and single-cell transcriptomic analyses. In the clinically immune, we find increased proportions of circulating follicular helper T cells and classical monocytes, while the humoral immune response shows characteristic age-related differences in the protected. Presence of memory CD4⁺ T cell clones with a strong cytolytic ZEB2⁺ T helper 1 effector signature, sharing identical T cell receptor clonotypes and recognizing the Pf-derived circumsporozoite protein (CSP) antigen are found in the blood of the Pf-infected participants gaining protection. Moreover, in clinically protected participants, ZEB2⁺ memory CD4⁺ T cells express lower level of inhibitory and chemotactic receptors. We thus propose that clonally expanded ZEB2⁺ CSP-specific cytolytic memory CD4⁺ Th1 cells may contribute to clinical immunity against the sporozoite and liver-stage Pf malaria.
... Antibodies against PfSPZ induced by CPS and PfSPZ Vaccine have had significant functional activity in vivo in mice with human livers 5,6,22 , and we also report significant functional activity (Fig. 3c). We hypothesize that antibodies that inhibit PfSPZ invasion of hepatocytes contribute to protection, but the primary effectors of protection are cellular immune responses that eliminate parasite-infected hepatocytes during the 5-6 days of parasite maturation in the liver [5][6][7][8][9]11,20,[23][24][25][26][27] . Consistent with this hypothesis, PfSPZ-and PfRBC-specific polyfunctional cytokine-producing memory CD4 T cells correlated with sterile protection in this study, as observed following CPS immunization 7 . ...
Article
Full-text available
A highly protective malaria vaccine would greatly facilitate the prevention and elimination of malaria and containment of drug-resistant parasites. A high level (more than 90%) of protection against malaria in humans has previously been achieved only by immunization with radiation-attenuated Plasmodium falciparum (Pf) sporozoites (PfSPZ) inoculated by mosquitoes; by intravenous injection of aseptic, purified, radiation-attenuated, cryopreserved PfSPZ ('PfSPZ Vaccine'); or by infectious PfSPZ inoculated by mosquitoes to volunteers taking chloroquine or mefloquine (chemoprophylaxis with sporozoites). We assessed immunization by direct venous inoculation of aseptic, purified, cryopreserved, non-irradiated PfSPZ ('PfSPZ Challenge') to malaria-naive, healthy adult volunteers taking chloroquine for antimalarial chemoprophylaxis (vaccine approach denoted as PfSPZ-CVac). Three doses of 5.12 × 10(4) PfSPZ of PfSPZ Challenge at 28-day intervals were well tolerated and safe, and prevented infection in 9 out of 9 (100%) volunteers who underwent controlled human malaria infection ten weeks after the last dose (group III). Protective efficacy was dependent on dose and regimen. Immunization with 3.2 × 10(3) (group I) or 1.28 × 10(4) (group II) PfSPZ protected 3 out of 9 (33%) or 6 out of 9 (67%) volunteers, respectively. Three doses of 5.12 × 10(4) PfSPZ at five-day intervals protected 5 out of 8 (63%) volunteers. The frequency of Pf-specific polyfunctional CD4 memory T cells was associated with protection. On a 7,455 peptide Pf proteome array, immune sera from at least 5 out of 9 group III vaccinees recognized each of 22 proteins. PfSPZ-CVac is a highly efficacious vaccine candidate; when we are able to optimize the immunization regimen (dose, interval between doses, and drug partner), this vaccine could be used for combination mass drug administration and a mass vaccination program approach to eliminate malaria from geographically defined areas.
... Continuous chloroquine coverage is provided to abort blood stages, that follow upon completion of liver-stage development and are responsible for malaria pathology. This protocol mounts sufficient immunity to liver stages to provide solid protection from sporozoite challenge, possibly by the absence of prolonged blood-stage exposure [3][4][5][6][7] . Here, we followed a modified concept of the successful malaria sporozoites under a continuous blood-stage prophylaxis approach (CPS 3 ). ...
Article
Full-text available
Vaccine development for Plasmodium vivax, an important human relapsing malaria, is lagging behind. In the case of the most deadly human malaria P. falciparum, unprecedented high levels of protection have been obtained by immunization with live sporozoites under accompanying chemoprophylaxis, which prevents the onset of blood-stage malaria. Such an approach has not been fully evaluated for relapsing malaria. Here, in the P. cynomolgi-rhesus macaque model for relapsing malaria, we employ the parasites’ natural relapsing phenotype to self-boost the immune response against liver-stage parasites, following a single-shot high-dose live sporozoite vaccination. This approach resulted in sterile protection against homologous sporozoite challenge in three out of four animals in the group that was also exposed for several days to blood stages during primary infection and relapses. One out of four animals in the group that received continuous chemoprophylaxis to abort blood-stage exposure was also protected from sporozoite challenge. Although obtained in a small number of animals as part of a Proof-of-Concept study, these results suggest that limited blood-stage parasite exposure may augment protection in this model. We anticipate our data are a starting point for further research into correlates of protection and extrapolation of the single-shot approach to develop efficacious malaria vaccines against relapsing human malaria.
... We think this net response is an indication of vaccine take, not that these low levels of antibodies mediate protection, which is mediated primarily by T cells. 24 We believe that three factors contributed to vaccine hyporesponsiveness. The first is that the partial anti-parasite immunity resulting from lifelong exposure to Pf parasites may have interdicted the PfSPZ used for immunization before they could invade, replicate and induce protective immune responses, reducing the effective dose to much less than 2.048 £ 10 5 PfSPZ. ...
Article
Full-text available
Background Plasmodium falciparum (Pf) Sporozoite (SPZ) Chemoprophylaxis Vaccine (PfSPZ-CVac) involves concurrently administering infectious PfSPZ and malaria drug, often chloroquine (CQ), to kill liver-emerging parasites. PfSPZ-CVac (CQ) protected 100% of malaria-naïve participants against controlled human malaria infection. We investigated the hypothesis that PfSPZ-CVac (CQ) is safe and efficacious against seasonal, endemic Pf in malaria-exposed adults. Methods Healthy 18–45 year olds were enrolled in a double-blind, placebo-controlled trial in Bougoula–Hameau, Mali, randomized 1:1 to 2.048 × 10⁵ PfSPZ (PfSPZ Challenge) or normal saline administered by direct venous inoculation at 0, 4, 8 weeks. Syringes were prepared by pharmacy staff using online computer-based enrolment that randomized allocations. Clinical team and participant masking was assured by identical appearance of vaccine and placebo. Participants received chloroquine 600mg before first vaccination, 10 weekly 300mg doses during vaccination, then seven daily doses of artesunate 200mg before 24-week surveillance during the rainy season. Safety outcomes were solicited adverse events (AEs) and related unsolicited AEs within 12 days of injections, and all serious AEs. Pf infection was detected by thick blood smears performed every four weeks and during febrile illness over 48 weeks. Primary vaccine efficacy (VE) endpoint was time to infection at 24 weeks. NCT02996695. Findings 62 participants were enrolled in April/May 2017. Proportions of participants experiencing at least one solicited systemic AE were similar between treatment arms: 6/31 (19.4%, 95%CI 9.2-36.3) of PfSPZ-CVac recipients versus 7/31 (22.6%, 95%CI 29.2-62.2) of controls (p value = 1.000). Two/31 (6%) in each group reported related, unsolicited AEs. One unrelated death occurred. Of 59 receiving 3 immunizations per protocol, fewer vaccinees (16/29, 55.2%) became infected than controls (22/30, 73.3%). VE was 33.6% by hazard ratio (p = 0.21, 95%CI -27·9, 65·5) and 24.8% by risk ratio (p = 0.10, 95%CI -4·8, 54·3). Antibody responses to PfCSP were poor; 28% of vaccinees sero-converted. Interpretation PfSPZ-CVac (CQ) was well-tolerated. The tested dosing regimen failed to significantly protect against Pf infection in this very high transmission setting. Funding U.S. National Institutes of Health, Sanaria. Registration number ClinicalTrials.gov identifier (NCT number): NCT02996695.
Article
Full-text available
Introduction: Malaria, a devastating febrile illness caused by protozoan parasites, sickened 247,000,000 people in 2021 and killed 619,000, mostly children and pregnant women in sub-Saharan Africa. A highly effective vaccine is urgently needed, especially for Plasmodium falciparum (Pf), the deadliest human malaria parasite. Areas covered: Sporozoites (SPZ), the parasite stage transmitted by Anopheles mosquitoes to humans, are the only vaccine immunogen achieving > 90% efficacy against Pf infection. This review describes > 30 clinical trials of PfSPZ vaccines in the U.S.A., Europe, Africa, and Asia, based on first-hand knowledge of the trials and PubMed searches of 'sporozoites,' 'malaria,' and 'vaccines.' Expert opinion: First generation (radiation-attenuated) PfSPZ vaccines are safe, well tolerated, 80-100% efficacious against homologous controlled human malaria infection (CHMI) and provide 18-19 months protection without boosting in Africa. Second generation chemo-attenuated PfSPZ are more potent, 100% efficacious against stringent heterologous (variant strain) CHMI, but require a co-administered drug, raising safety concerns. Third generation, late liver stage-arresting, replication competent (LARC), genetically-attenuated PfSPZ are expected to be both safe and highly efficacious. Overall, PfSPZ vaccines meet safety, tolerability, and efficacy requirements for protecting pregnant women and travelers, with licensure for these populations possible within five years. Protecting children and mass vaccination programs to block transmission and eliminate malaria are long-term objectives.
Article
The tropical disease malaria remains a major cause of global morbidity. Once transmitted to the human by a blood-feeding mosquito, the unicellular malaria parasite comes into contact with the complement system and continues to interact with human complement during its intraerythrocytic replication cycles. In the course of infection, both the classical and the alternative pathway of complement are activated, leading to parasite opsonization and lysis as well as the induction of complement-binding antibodies. While complement activity can be linked to the severity of malaria, it remains to date unclear, whether human complement is beneficial for protective immunity or if extensive complement reactions may rather enhance pathogenesis. In addition, the parasite has evolved molecular strategies to circumvent attack by human complement and has even developed means to utilize complement factors as mediators of host cell infection. In this review, we highlight current knowledge on the role of human complement for the progression of malaria infection. We discuss the various types of interactions between malaria parasites and complement factors with regard to immunity and infection outcome and set a special emphasis on the dual role of complement in the context of parasite fitness.
Article
Full-text available
Background: The candidate malaria vaccine RTS,S/AS01 reduced episodes of both clinical and severe malaria in children 5 to 17 months of age by approximately 50% in an ongoing phase 3 trial. We studied infants 6 to 12 weeks of age recruited for the same trial. Methods: We administered RTS,S/AS01 or a comparator vaccine to 6537 infants who were 6 to 12 weeks of age at the time of the first vaccination in conjunction with Expanded Program on Immunization (EPI) vaccines in a three-dose monthly schedule. Vaccine efficacy against the first or only episode of clinical malaria during the 12 months after vaccination, a coprimary end point, was analyzed with the use of Cox regression. Vaccine efficacy against all malaria episodes, vaccine efficacy against severe malaria, safety, and immunogenicity were also assessed. Results: The incidence of the first or only episode of clinical malaria in the intention-to-treat population during the 14 months after the first dose of vaccine was 0.31 per person-year in the RTS,S/AS01 group and 0.40 per person-year in the control group, for a vaccine efficacy of 30.1% (95% confidence interval [CI], 23.6 to 36.1). Vaccine efficacy in the per-protocol population was 31.3% (97.5% CI, 23.6 to 38.3). Vaccine efficacy against severe malaria was 26.0% (95% CI, -7.4 to 48.6) in the intention-to-treat population and 36.6% (95% CI, 4.6 to 57.7) in the per-protocol population. Serious adverse events occurred with a similar frequency in the two study groups. One month after administration of the third dose of RTS,S/AS01, 99.7% of children were positive for anti-circumsporozoite antibodies, with a geometric mean titer of 209 EU per milliliter (95% CI, 197 to 222). Conclusions: The RTS,S/AS01 vaccine coadministered with EPI vaccines provided modest protection against both clinical and severe malaria in young infants. (Funded by GlaxoSmithKline Biologicals and the PATH Malaria Vaccine Initiative; RTS,S ClinicalTrials.gov number, NCT00866619.).
Article
Full-text available
Consistent high-level, vaccine-induced protection against human malaria has only been achieved by inoculation of Plasmodium falciparum (Pf) sporozoites (SPZ) by mosquito bites. We report that the PfSPZ vaccine, composed of attenuated, aseptic, purified, cryopreserved PfSPZ, was safe and well-tolerated when administered 4 to 6 times intravenously (IV) to 40 adults. 0/6 subjects receiving 5 doses, 3/9 subjects receiving 4 doses of 1.35 x 10(5) PfSPZ vaccine, and 5/6 nonvaccinated controls developed malaria following controlled human malaria infection (P = 0.015 in the 5-dose group and P = 0.028 for overall, both versus controls). PfSPZ-specific antibody and T cell responses were dose-dependent. These data indicate that there is a dose-dependent immunological threshold for establishing high-level protection against malaria that can be achieved by IV administration of a vaccine that is safe and meets regulatory standards.
Article
Full-text available
Control of parasite replication exerted by MHC class I restricted CD8+ T-cells in the liver is critical for vaccination-induced protection against malaria. While many intracellular pathogens subvert the MHC class I presentation machinery, its functionality in the course of malaria replication in hepatocytes has not been characterized. Using experimental systems based on specific identification, isolation and analysis of human hepatocytes infected with P. berghei ANKA GFP or P. falciparum 3D7 GFP sporozoites we demonstrated that molecular components of the MHC class I pathway exhibit largely unaltered expression in malaria-infected hepatocytes until very late stages of parasite development. Furthermore, infected cells showed no obvious defects in their capacity to upregulate expression of different molecular components of the MHC class I machinery in response to pro-inflammatory lymphokines or trigger direct activation of allo-specific or peptide-specific human CD8+ T-cells. We further demonstrate that ectopic expression of circumsporozoite protein does not alter expression of critical genes of the MHC class I pathway and its response to pro-inflammatory cytokines. In addition, we identified supra-cellular structures, which arose at late stages of parasite replication, possessed the characteristic morphology of merosomes and exhibited nearly complete loss of surface MHC class I expression. These data have multiple implications for our understanding of natural T-cell immunity against malaria and may promote development of novel, efficient anti-malaria vaccines overcoming immune escape of the parasite in the liver.
Article
Full-text available
Following Anopheles mosquito-mediated introduction into a human host, Plasmodium parasites infect hepatocytes and undergo intensive replication. Accumulating evidence indicates that CD8+ T cells induced by immunization with attenuated Plasmodium sporozoites can confer sterile immunity at the liver stage of infection; however, the mechanisms underlying this protection are not clearly understood. To address this, we generated recombinant Plasmodium berghei ANKA expressing a fusion protein of an ovalbumin epitope and green fluorescent protein in the cytoplasm of the parasite. We have shown that the ovalbumin epitope is presented by infected liver cells in a manner dependent on a transporter associated with antigen processing and becomes a target of specific CD8+ T cells from the T cell receptor transgenic mouse line OT-I, leading to protection at the liver stage of Plasmodium infection. We visualized the interaction between OT-I cells and infected hepatocytes by intravital imaging using two-photon microscopy. OT-I cells formed clusters around infected hepatocytes, leading to the elimination of the intrahepatic parasites and subsequent formation of large clusters of OT-I cells in the liver. Gamma interferon expressed in CD8+ T cells was dispensable for this protective response. Additionally, we found that polyclonal ovalbumin-specific memory CD8+ T cells induced by de novo immunization were able to confer sterile protection, although the threshold frequency of the protection was relatively high. These studies revealed a novel mechanism of specific CD8+ T cell-mediated protective immunity and demonstrated that proteins expressed in the cytoplasm of Plasmodium parasites can become targets of specific CD8+ T cells during liver-stage infection.
Article
Full-text available
Background The candidate malaria vaccine RTS,S/AS01 reduced episodes of both clinical and severe malaria in children 5 to 17 months of age by approximately 50% in an ongoing phase 3 trial. We studied infants 6 to 12 weeks of age recruited for the same trial. Methods We administered RTS,S/AS01 or a comparator vaccine to 6537 infants who were 6 to 12 weeks of age at the time of the first vaccination in conjunction with Expanded Program on Immunization (EPI) vaccines in a three-dose monthly schedule. Vaccine efficacy against the first or only episode of clinical malaria during the 12 months after vaccination, a coprimary end point, was analyzed with the use of Cox regression. Vaccine efficacy against all malaria episodes, vaccine efficacy against severe malaria, safety, and immunogenicity were also assessed. Results The incidence of the first or only episode of clinical malaria in the intention-to-treat population during the 14 months after the first dose of vaccine was 0.31 per personyear in the RTS,S/AS01 group and 0.40 per person-year in the control group, for a vaccine efficacy of 30.1% (95% confidence interval [CI], 23.6 to 36.1). Vaccine efficacy in the per-protocol population was 31.3% (97.5% CI, 23.6 to 38.3). Vaccine efficacy against severe malaria was 26.0% (95% CI, −7.4 to 48.6) in the intention-totreat population and 36.6% (95% CI, 4.6 to 57.7) in the per-protocol population. Serious adverse events occurred with a similar frequency in the two study groups. One month after administration of the third dose of RTS,S/AS01, 99.7% of children were positive for anti-circumsporozoite antibodies, with a geometric mean titer of 209 EU per milliliter (95% CI, 197 to 222). Conclusions The RTS,S/AS01 vaccine coadministered with EPI vaccines provided modest protection against both clinical and severe malaria in young infants. (Funded by GlaxoSmithKline Biologicals and the PATH Malaria Vaccine Initiative; RTS,S ClinicalTrials.gov number, NCT00866619.)
Article
It is shown how, in regular parametric problems, the first-order term is removed from the asymptotic bias of maximum likelihood estimates by a suitable modification of the score function. In exponential families with canonical parameterization the effect is to penalize the likelihood by the Jeffreys invariant prior. In binomial logistic models, Poisson log linear models and certain other generalized linear models, the Jeffreys prior penalty function can be imposed in standard regression software using a scheme of iterative adjustments to the data.
Article
Consistent, high-level, vaccine-induced protection against human malaria has only been achieved by inoculation of Plasmodium falciparum (Pf) sporozoites (SPZ) by mosquito bites. We report that the PfSPZ Vaccine—composed of attenuated, aseptic, purified, cryopreserved PfSPZ—was safe and wel-tolerated when administered four to six times intravenously (IV) to 40 adults. Zero of six subjects receiving five doses and three of nine subjects receiving four doses of 1.35 × 105 PfSPZ Vaccine and five of six nonvaccinated controls developed malaria after controlled human malaria infection (P = 0.015 in the five-dose group and P = 0.028 for overall, both versus controls). PfSPZ-specific antibody and T cell responses were dose-dependent. These data indicate that there is a dose-dependent immunological threshold for establishing high-level protection against malaria that can be achieved with IV administration of a vaccine that is safe and meets regulatory standards.
Article
A CD4+ T cell clone (A1.6) was derived from spleen cells of mice immunized with irradiated sporozoites. This T cell clone recognizes an antigen that is shared by sporozoites and blood forms of Plasmodium berghei and differs from the circumsporozoite protein. Clone A1.6 displays cytotoxic activity, produces IFN-gamma and IL-2 in vitro, and recognizes the plasmodial antigen in the context of the class II I-Ed molecule. Passive transfer of this CD4+ clone into naive mice resulted in a high degree of protection against sporozoite challenge.