Sei sulla pagina 1di 9

MINIREVIEW

Vaccines against malaria—still a long way to go


Kai Matuschewski
Department of Molecular Parasitology, Institute of Biology, Humboldt University Berlin, Germany

Keywords Several species of Plasmodium cause a broad spectrum of human disease


gametocyte; immunization; malaria; that range from nausea and fever to severe anemia, cerebral malaria, and
merozoite; Plasmodium falciparum;
multiorgan failure. In malaria-endemic countries, continuous exposure to
Plasmodium vivax; sporozoite; vaccine
Plasmodium sporozoite inoculations and subsequent blood infections elicit
Correspondence only partial and short-lived immunity, which gradually develops over many
K. Matuschewski, Department of Molecular years of parasite exposure and multiple clinical episodes. The ambitious
Parasitology, Institute of Biology/Faculty of goal of malaria vaccinology over the past 70 years has been to develop an
Life Sciences, Humboldt University Berlin, immunization strategy that mounts protection superior to naturally
Philippstr. 13 #14, 10115 Berlin, Germany acquired immunity. Herein, three principal concepts in evidence-based
Fax: +49 30 2093 6051
malaria vaccine development are compared. Feasible leads are typically
Tel: +49 30 2093 6053
stand-alone subunit vaccine approaches that block Plasmodium parasite life
E-mail: kai.matuschewski@hu-berlin.de
cycle progression or parasite/host interactions, and they constitute the
(Received 31 January 2017, revised 9 April majority of candidates in preclinical research and early clinical testing.
2017, accepted 10 May 2017) Integrated approaches incorporate malaria antigen(s) into licensed or
emerging pediatric vaccine formulations. This strategy can complement the
doi:10.1111/febs.14107 malaria control portfolio even if the antimalarial component is only par-
tially effective and has led to the development of the only candidate vac-
cine to date, namely RTS,S-AS01. Experimental whole parasite vaccine
approaches have been repeatedly shown to elicit sterile and lasting protec-
tion against identical parasite strains, but mass production, proof of broad
protection against different parasite strains, and routes of vaccine delivery
remain significant translational road blocks. Global access to an effective
and affordable malaria vaccine will critically depend on innovative transla-
tional research that builds on a better molecular understanding of Plasmod-
ium biology and host immunity.

Introduction
[2]. What distinguishes Plasmodium is their unidirec-
Malaria remains the most important mosquito-borne tional population expansion first in the liver and then
infectious disease and is caused by blood infection in red blood cells before differentiation into sexual
with Plasmodium parasites [1]. These obligate intracel- stages that colonize Anopheles mosquitoes [3].
lular pathogens belong to the phylum Apicomplexa, Plasmodium stage conversion over the course of the
which are single-cell eukaryotes that coevolved with human infection offers multiple targets for vaccine-
their respective hosts across the entire animal kingdom induced immunity (Fig. 1), but the parasite is tailored to

Abbreviations
BCG, Bacillus Calmette-Gue rin; CelTOS, cell-traversal protein for ookinetes and sporozoites; ChAD63, chimpanzee adenovirus 63; CSP,
circumsporozoite protein; GLURP, glutamate rich protein; GMO, genetically modified organism; MSP, merozoite surface protein; MVA,
modified vaccinia Ankara virus; PfEMP1, Plasmodium falciparum erythrocyte membrane protein 1; RTS,S/AS01, proprietary vaccine
formulation by GlaxSmithKline Biologicals; SSP, sporozoite surface protein; TRAP, thrombospondin-related anonymous protein.

The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies 1


Malaria vaccine perspectives K. Matuschewski

reinfect the host lifelong. Naturally acquired immunity A hypothetical malaria vaccine is highly unlikely to
develops only gradually after > 20 clinical episodes over replace existing malaria control tools, unless it achieves
the course of many years [4], but it is never sterile [5]. Nat- complete sterilizing immunity. Rather a vaccine will be
ural immunity mostly comprises plasma B cells that pro- an integral part of malaria control programs, which
duce antibodies against variant parasite-encoded include access to rapid diagnosis and treatment with
erythrocyte surface antigens, such as Plasmodium falci- Artemisinin-based combination therapies, ownership
parum erythrocyte membrane proteins (PfEMP1s) and, to of long-lasting insecticide treated nets, and regular
a lesser extent, merozoite proteins (Fig. 1) [5]. In marked campaigns of indoor residual spraying with a suitable
contrast, acute infections by viral and some bacterial insecticide. Malaria control has been successful over
pathogens elicit lasting and frequently lifelong protection the past 10 years because of exceptional donor com-
against reinfection, often after a single infection. This mitment. Continuation of effective malaria manage-
immunological memory is the basis for protective vaccines ment and further reduction in the malaria disease
[6] and clearly missing in malaria and other chronic infec- burden in high-risk endemic areas critically depends
tious diseases, such as tuberculosis. Accordingly, simply on sustained recognition of malaria as a global health
extrapolating successful antimicrobial vaccines to malaria priority. Furthermore, widespread tolerance against
is likely inadequate, and despite intense efforts in malaria insecticides in Anopheles vector populations and para-
vaccinology over the past 70 years the goal to develop an site resistance against every approved antimalarial
effective malaria vaccine that performs superior to natu- drug on the market emphasize the urgent need for a
rally acquired immunity has not yet been achieved. safe and effective vaccine.

Fig. 1. Adaptive immune responses during Plasmodium life cycle progression in humans. (1) During an infectious Anopheles bite
sporozoites (green) are injected and migrate through the skin to enter a blood capillary and eventually invade a suitable hepatocyte.
Plasma cells (purple) originating from activated memory B cells and secreting antibodies against SSPs can immobilize sporozoites and
reduce the infectious parasite dose. (2) During the first pre-erythrocytic asexual replication phase in the liver a single successful
sporozoite forms thousands of infectious merozoites. Cytolytic effector memory CD8+ T cells (T) can specifically recognize and lyse
infected hepatocytes. Merozoites initiate the second asexual replication phase inside erythrocytes. (3) Plasmodium falciparum, unlike
other Plasmodium species, extensively remodels the erythrocyte surface resulting in sequestration of infected erythrocytes to the
vascular endothelium. This so-called cytoadherence is a major cause of severe disease. A large range of plasma cells (yellow) originating
from activated memory B cells and secreting antibodies against the P. falciparum-encoded surface protein repertoire correlates with
gradual acquisition of naturally acquired immunity in malaria-endemic areas. (4) Plasma cells (blue) originating from activated memory B
cells and secreting antibodies against MSPs can immobilize merozoites and reduce parasitemia in the blood circulation. A subset of
merozoites differentiates into female (blue) and male (pink) gametocytes, which may be ingested by an Anopheline mosquito and lead to
completion of the life cycle.

2 The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies


K. Matuschewski Malaria vaccine perspectives

arrested, liver stages [22–25]. Although work in murine


Vaccine opportunities to break the
malaria models has consistently recovered antigen-spe-
Plasmodium cycle
cific cytolytic effector memory CD8+ T cells as the
Parasitic eukaryotes are complex pathogens and some main cell type that recognizes and lyses infected hepa-
of the finest artists in escaping host defense mecha- tocytes [26–29], a similar important role remains to be
nisms, including adaptive immune responses [7,8]. No shown for human malaria.
correlates of protection against malaria, that is, Merozoites are tailor-made to actively and rapidly
immune signatures or antigens that permit distinction invade red blood cells (Fig. 1). This second asexual
between previous parasite exposures vs. protective replication phase inside erythrocytes is the exclusive
immunity, have been identified yet [5,9]. Therefore, cause of malaria and an ideal target to reduce the inci-
malaria vaccine development still relies on empirical dence of clinical attacks. However, antibody-mediated
testing in preclinical models [10] and small human inhibition and destruction of merozoites [30] is limited
challenge studies [11]. Four principal, and ideally com- to a very brief (~ 30 s) moment, when the parasite is
plementary, biological targets, namely sporozoites, outside its host cell and in the process of invading a
liver stages, and asexual and sexual blood stages new erythrocyte. Many candidate antigens that per-
(Fig. 1), can be exploited for vaccine development to form vital functions in the invasion process are stored
fast-forward naturally acquired immunity to curtail— in specialized organelles, termed micronemes, rhop-
in the order of priority—(a) severe disease, (b) clinical tries, and dense granules, and are sequentially released
malaria episodes, (c) parasite load, and (d) community upon intimate merozoite contact with the erythrocyte
transmission [12–16]. surface, thus further limiting exposure to inhibitory
Sporozoites are the infectious agents that are antibodies [31]. A population-based study of antibody
injected during the probing phase of an infectious responses in malaria-endemic countries did not return
Anopheles bite (Fig. 1). They actively migrate through any signatures of protection, and many candidate anti-
the skin in search of a blood capillary to be flushed to gens were recognized to a similar extent by protected
the final target organ, the liver [3,17]. Antibody- and susceptible individuals [32]. In addition, increasing
mediated inhibition of sporozoite infection of the liver evidence suggests that memory B cells, which are the
can reduce the initial parasite inoculum and requires principal antibody-secreting cells that target blood
the induction of long-lived antibody responses [18]. stage antigens upon vaccine-induced immunity, appear
Accordingly, antisporozoite vaccines should stimulate to contract remarkably early [33].
the formation of germinal center B cells, which can Severe malaria in a proportion of children infected
maintain an effector phenotype and initiate rapid with P. falciparum remains a leading cause of death in
plasma cell differentiation through interaction with Afrotropical regions [34]. After invasion, P. falciparum
memory T follicular helper cells [19]. However, also in extensively refurbishes the host erythrocyte and
the secondary response, upon parasite antigen rechal- exports PfEMP1s and other variant antigens to the
lenge, high levels of inhibitory antibodies are only surface. Binding of infected erythrocytes to cognate
formed within a few days. In the meantime, the sporo- receptors on the vascular endothelium, a process ter-
zoite is already in full liver stage development. Accord- med cytoadhesion, is crucial to avoid splenic clearance
ingly, antisporozoite vaccine strategies that elicit and results in a large pathological spectrum, ranging
antibodies can only achieve substantial reduction in from capillary occlusion and hypoxia to tissue inflam-
Plasmodium transmission in subsequent infections and mation and multiorgan failure [35]. A broad range of
have an impact over time. However, they consistently memory B cells that target the P. falciparum-encoded
fail in preventing malaria infections [20] and, hence, surface protein repertoire is gradually acquired during
are not considered in travel medicine. infancy and childhood and reflects the slow acquisition
During the obligate pre-erythrocytic phase in the of antidisease immunity in malaria-endemic areas.
liver, Plasmodium undergoes an enormous population Whether other human Plasmodium species, including
expansion resulting in the formation of thousands of Plasmodium vivax, express parasite-encoded surface
first-generation merozoites (Fig. 1). Only during liver markers that can be targeted by inhibitory antibodies
stage maturation is the parasite susceptible to direct T- remains an unsolved matter.
cell-mediated killing, and this period is long enough A proportion of merozoites eventually differentiate
(~ 7 days) for secondary recall responses [3,21]. Last- into sexual stages, which can recombine and complete
ing and robust antiliver stage immunity is the principal the life cycle upon ingestion by an Anopheles mos-
mechanism for the experimental success of vaccine quito. Although of no immediate benefit to the vacci-
strategies that lead to live, but developmentally nated individual high-titer antibodies against

The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies 3


Malaria vaccine perspectives K. Matuschewski

gametocytes and the diploid zygote might aid in reduc- that any of the Plasmodium antigens is a signature of
tion of Plasmodium transmission on a community-level protective immunity as opposed to parasite exposure
[36]. However, when prioritizing a small subset of can- remains elusive [32].
didate antigens for the most vulnerable population, A landmark study established that a recombinant
infants and pregnant women, gamete and ookinete sur- malaria vaccine, comprising MSP1, MSP2, and ring-
face proteins are negligible. infected erythrocyte surface antigen, could exert selec-
Together, two of the five parasite stages in the mam- tive pressure on parasite populations [38]. Although
malian host, namely liver stages and parasite antigens this finding signifies selective immunity against some
on the infected erythrocyte surface, represent promis- antigens, in this case MSP2, it highlights swift parasite
ing targets for vaccine strategies. Liver stages can be evasion of vaccine-induced selection pressure. A recent
targeted in all Plasmodium species and an effective vac- completion of the largest field trial of a blood stage
cine can elicit complete protection against blood infec- vaccine candidate formulation, consisting of recombi-
tion. But how to accurately induce a range of effector nant MSP3 and glutamate-rich protein (GLURP) in
memory CD8+ T cells that home to the liver upon re- alum, in 1,735 children revealed no efficacy [39], reiter-
infection remains a principal problem in vaccinology ating enormous challenges for merozoite vaccine
[37]. Antibody-mediated inhibition of P. falciparum- approaches. These examples illustrate that antigenic
encoded erythrocyte surface proteins can prevent dis- variation and polymorphic antigens inevitably remain
ease and targets the long replication phase inside red the largest obstacles for subunit vaccine development.
blood cells. But management of antigenic variation is Only informed design of a multivalent vaccine incor-
a persistent challenge. On the other hand, induction of porating several invariant antigens can surmount this
antibody-mediated inhibition of sporozoite and mero- persisting challenge.
zoite invasion or gamete fusion in the mosquito mid- An alternative approach is the delivery of recombi-
gut is less demanding. While either one may offer nant virus particles to express Plasmodium antigens, in
synergistic benefits to an antiliver stage or anticytoad- this case TRAP/SSP2 [40]. Multiple rounds of opti-
hesion vaccine, none are likely to curtail Plasmodium mization returned a heterologous prime/boost protocol
life cycle progression on their own. It follows that with chimpanzee adenovirus 63 (ChAd63) and modi-
malaria vaccine research aims at developing a multi- fied vaccinia Ankara virus (MVA), respectively. A
component, multistage vaccine, similar to pediatric recent small phase II study with 115 participants in
combination vaccines against multiple childhood Senegal showed no efficacy of this vaccine, and com-
diseases. bined analysis with earlier data from Kenia returned
only a small, albeit significant, delay until patency [40].
A number of candidate antigens are still in the
Feasible leads: testing one antigen at
exploratory, preclinical phase or phase I safety testing.
a time
One example is a formulation with the cell-traversal
Until date, the vast majority of candidate antigens are protein for ookinetes and sporozoites (CelTOS), which
designed as stand-alone malaria subunit vaccines was tested in an elegant murine infection model using
(Table 1). Major advantages of a distinct antigen for- transgenic chimeric parasites that express the P. falci-
mulation include the potential feasibility to produce a parum protein [41]. Efficacy was very modest leading
defined malaria vaccine under good manufacturing only to a twofold reduction in parasite liver load,
procedures, at reasonable cost, and in a formulation which does not translate in any detectable effect on
that is thermostable and easy to deliver intramuscu-
larly to human subjects. Accordingly, identification of
a protective antigen and its translation to a safe and Table 1. Exemplary candidate subunit preparations.
protective malaria subunit vaccine would be the most
desirable solution. Over the past 30 years, a large Target antigen Delivery platform Reference
range of candidate antigen formulations often included Phase II clinical trials
parasite antigens in the historic order of recombinant GLURP, MSP3 Protein/alum [39]
cloning. They encompassed initially the major sporo- TRAP ChAd63/MVA [40]
zoite surface proteins (SSPs) circumsporozoite protein Phase I trials/ preclinial
(CSP/SSP1) and thrombospondin-related anonymous CelTOS Protein/AS10B adjuvant [41]
Var2-CSA Protein/alhydrogel [42]
protein (TRAP/SSP2), the major merozoite surface
s25, s230 ChAd63/MVA [43]
proteins 1–3 (MSP1–3), and the sexual stage antigens
25 and 230 (s25, s230). However, experimental proof s25, s230, sexual stage antigen 25, 230.

4 The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies


K. Matuschewski Malaria vaccine perspectives

blood infection. Therefore, such a vaccine needs to be Table 2. Integrated vaccination approaches.
considerably re-engineered before any further testing, Integrated Plasmodium
including in clinical trials. An attractive candidate is Original vaccine version antigen Reference
the VAR2CSA surface protein on infected erythro-
Phase III clinical trials
cytes, which mediates binding to chondroitin sulfate
Hepatitis B RTS,S/AS01 PfCSP fragment [44,47,48]
A (CSA) on the placenta and is a major factor for
vaccine
placental malaria in pregnant women [42]. Many Preclinical evaluation
obstacles remain, but the development of a tailored Yellow fever YF17D/PyCS PyCSP fragment [49]
vaccine for young women will be an important step- 17D
ping-stone toward anticytoadhesion vaccines. Finally, BCG Glaxo BCG-CS PfCSP full length [50]
sexual stage antigens to induce transmission-blocking
Pf, Plasmodium falciparum; Py, Plasmodium yoelii.
immunity can be tested in membrane feeding assays
that partly mimic natural transmission to Anopheline
mosquitoes. A systematic analysis returned two anti- but in addition a smaller fraction consisting of a por-
gens, s25 and s230, as lead candidates for further tion of the P. falciparum CSP, including the repeat
investigation [43]. Together, feasible subunit vaccine region (R) and a T-cell epitope (T), fused to the S
approaches often deliver disappointing results in effi- antigen. After multiple rounds of safety, immuno-
cacy testing in the field, and conserved, nonredun- genicity and efficacy testing and improvements of a
dant antigens that are indispensable for parasite life formulation with a potent liposome-based adjuvant,
cycle progression need to be targeted to avoid termed AS01 [46], was selected for large-scale studies
counterselection [38]. in at-risk populations [47]. In all studies, there was an
initial short phase of protection against clinical malar-
ia, in good correlation with exceptionally high anti-
Integrated vaccines: taking advantage
body titers resulting from the vaccination scheme. As
of existing pediatric vaccines
expected [20], protection disappeared with the concur-
As a valuable addition to sustain affordable malaria rent decrease in antibody titers after few months.
control, a candidate vaccine needs to perform safely, Since initial protection was offset in later years the
and ideally efficiently, during neonatal and early life overall result was no efficacy [48]. These disappointing
vaccination in resource-poor settings. Accordingly, results reflect the fundamental differences in infection
such a malaria childhood vaccine needs to be tailored biology between viral and parasitic pathogens and
for integration into the expanded program of immu- cast doubt whether integrated approaches work in
nization. A powerful answer is to swing aboard principle.
licensed pediatric vaccines and add-on a malaria anti- Nevertheless, the importance of the RTS,S/AS01 tri-
gen. When combined as prime-boost strategy employ- als cannot be overestimated. They achieved the first
ing various vaccine approaches Plasmodium infections advancement of a candidate vaccine against any of the
may be targeted in addition to the original pathogen three most important infectious diseases, HIV/AIDS,
(Table 2). Most importantly, because they are not tuberculosis, and malaria, to phase III clinical trials,
stand-alone formulations integrated malaria vaccines the set-up of clinical trials capacity in malaria-ende-
are not required to perform to perfection from the mic countries, and a robust foundation for further
beginning and can be improved iteratively, while developments. Important research topics are to
always maintaining their original purpose, for instance uncover whether (a) CD4+ and CD8+ T-cell
complete protection against hepatitis B. responses were induced in addition to antibody
It is, therefore, no surprise that only one candidate responses, (b) the selected Plasmodium antigen is cen-
malaria vaccine, which followed this approach from tral to pre-erythrocytic immunity, (c) alternative liver
the original design, ever advanced to pivotal phase III stage antigens might perform superior, and (d)
clinical trials, namely RTS,S/AS01 [44]. This vaccine biomarkers other than antibody titers might be better
formulation resembles an advanced hepatitis B vac- predictors of vaccine efficacy.
cine, which was the world’s first recombinant vaccine It will also be interesting to explore whether combi-
[45]. The vaccine consists of virus-like particles that nation with other integrated formulations can improve
can be produced in large fermenters by recombinant the outcome (Table 2). A recombinant yellow fever
expression of the hepatitis B surface (S) antigen in vaccine has been designed for preclinical studies in the
Saccharomyces cerevisiae. The modified version still murine malaria model Plasmodium yoelii [49], a per-
contains the S antigen as the largest protein portion, haps critical intermediate step toward translation,

The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies 5


Malaria vaccine perspectives K. Matuschewski

which was omitted entirely in the development of Early proof of principle studies demonstrated sterile
RTS,S/AS01 [10]. The yellow fever vaccine is based on immunity in humans that were immunized by exposure
a live, attenuated virus and a single dose protects for to c-irradiated P. falciparum-infected mosquitoes, and
life. It will be interesting to explore potential protec- these findings could be consistently repeated [51]. Mos-
tion against malaria once a suitable P. falciparum anti- quito delivery of c-irradiated sporozoites (cspz) was
gen is selected. The recombinant expression of soon recognized unrealistic, but these studies were an
P. falciparum CSP in the childhood tuberculosis vac- impressive testimonial for the validity of the original
cine strain Bacillus Calmette-Guerin (BCG) is an inter- concept [22], the important contribution of murine
esting research direction, since this approach builds on malaria models for evidence-based malaria vaccine
the robust safety and efficacy of the only live, attenu- development [10], and, perhaps most critical, the feasi-
ated antibacterial pediatric vaccine [50]. Immunity bility to develop a malaria vaccine on the basis of
against the complex intracellular bacterium Mycobac- arrested liver stage development.
terium tuberculosis is likely to resemble anti-Plasmodium Limitations of the cspz production include strict
immunity much closer than antiviral immunity and also dependence on biosafety level 3 facilities and batch-to-
requires only one immunization dose. However, experi- batch variations, because c irradiation induces uncon-
mental testing of vaccine safety and efficacy proves diffi- trolled DNA double-strand breaks. Moreover, the full
cult, since a preclinical animal model was not range of liver stage-induced immunity may be missed,
considered [50]. Other pediatric vaccines that are because cspz arrest very early in liver infection, imme-
routinely delivered, including against measles, might diately after sporozoite transformation [21]. A strong
also serve as suitable platforms for integrated malaria advantage of the cspz technology is the universal cov-
vaccine strategies. erage of every human Plasmodium species and the abil-
ity to implement multiple vaccine strains, which can
cover regional parasite diversity in the tropics [52].
Attenuated liver stages: benchmark
Some of the restrictions of cspz were overcome by
experimental malaria vaccines
targeted removal of stage-specific Plasmodium genes
In recognition of the complex molecular make-up and that perform vital functions for liver stage maturation
intriguing escape mechanisms of Plasmodium parasites [24]. Genetically arrested parasites (GAPs) can now be
as well as the persistent challenges to subunit malaria engineered as precision malaria vaccines, tailored to
vaccines, major efforts now return to the origins of arrest at any given point in pre-erythroyctic develop-
malaria vaccine development [22]. The unifying princi- ment and harboring additional components, such as
ple is to elicit lasting, sterile immunity by immuniza- blood stage antigens and ligands to stimulate antigen
tion with live Plasmodium sporozoites, which cannot presentation. Of note, occupational exposure or acci-
progress to blood infection (Table 3). dental mosquito release no longer pose health risks,
greatly facilitating potential production in malaria-
endemic countries. P. falciparum GAPs have already
Table 3. Live, metabolically active Plasmodium sporozoite advanced to clinical testing, and first safety and
inoculations. immunogenicity data are encouraging [53]. However,
in marked contrast to cspz, GAP technology requires
Approach Advantages Challenges Reference
re-engineering by reverse genetics for every Plasmod-
In clinical trials ium species and strain.
Chloroquine Superior immunity Patient [23,54] An alternative approach to stop blood infection is
cover compliance sporozoite delivery under simultaneous drug cover
c-Irradiation Applicable to all Low immunity [22,51]
[21,23,25]. Again, the success of a preclinical model,
(cspz) species and strains
where mice were given normal sporozoites together
Gene deletion Precision liver GMO strains [24,53]
(GAP) stage arrests, with the antimalarial drug chloroquine and enjoyed
no BSL3 facility lasting protection against challenge infection [23], were
required repeated successfully in small human trials [54]. Drug
Preclinical validation cover can also be done with the antibiotic azithromy-
Azithromycin Superior immunity Patient [25] cin, which leads to release of attenuated liver stage
cover compliance
merozoites and superior protection [25], and the 8-ami-
Primaquine Elimination of Low immunity [55]
noquinoline primaquine, which eliminates developing
cover liver stages
and quiescent liver stages [55] (Table 3). Major safety
BSL3, biosafety level 3; GMO, genetically modified organism. concerns with all drug cover approaches are patient

6 The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies


K. Matuschewski Malaria vaccine perspectives

compliance and a range of pharmacokinetics leading viral and bacterial pathogens. Eliciting strong recall
to inconsistent liver stage arrest in different individu- antibody responses alone appear insufficient against
als. Plasmodium parasites. These eukaryotic cells evolved
sophisticated strategies to escape invasion inhibition
by sequential secretion of redundant parasite ligands
Whole parasite vaccines: terminal or
and to overcome recognition of visible antigens by
roadmap from bench to field?
antigenic diversity. Integration of candidate Plasmod-
Four significant hurdles need to be overcome before ium antigens into licensed vaccines for neonatal and
whole parasite vaccines can be reasonably implemented childhood immunization remains largely underex-
in malaria-endemic countries. (a) Mass rearing of plored. The historic momentum gained by multicenter
infected Anopheles mosquitoes, hand-dissection of sali- clinical trials of RTS,S/AS01 should be utilized to
vary glands, and sporozoite purification are labor- advance this vaccine further and initiate immunologi-
intense processes. Broad access and affordability will cal studies in murine malaria models to gain mechanistic
critically depend on replacement by automated mos- insights into the potential effects against invading sporo-
quito-free, axenic sporozoite culture systems. (b) Wide- zoites and developing liver stages, which remain inacces-
ranging protection against different parasite strains is sible in human subjects. Since the choice of target antigen
vital and was addressed only very recently. Protection is essential a better molecular understanding of what dis-
against challenge with a heterologous P. falciparum tinguishes a protective immune response from immune
strain was incomplete in subjects immunized by three recognition of parasite exposure is important. For any
doses of sporozoite inoculations under chloroquine subunit vaccine approach the necessary evidence to
cover [56] and negligible in individuals immunized with advance to clinical testing should be based on robust
five high doses of Pfcspz [57]. These unsatisfactory indications for signatures of protection. Whole sporo-
results raise the alarming, yet plausible, possibility that zoite vaccines need to be 100% effective to be successful,
robust sterile protection elicited by whole parasite vacci- but can also provide the missing roadmap and assist in
nes is not strain transcending. If confirmed, a potential prioritizing Plasmodium pre-erythrocytic antigens.
solution could be vaccine cocktails containing different Pioneering work beyond classical vaccine development,
strains, but selection for vaccine-resistant strains could readiness of each stakeholder to start all over again, and
emerge as a major risk. (c) Vaccines need to be delivered public awareness of one of the most important challenges
safely by trained field workers, and the preferred route in medicine are vital to further accelerate malaria vacci-
of vaccine delivery is intramuscular or subcutaneous nology. Yet, the discovery, development, and licensing of
needle injection. Thus far, lasting protection was only a malaria vaccine formulation, which meets all necessary
achieved by intravenous sporozoite injection, which is requirements, including safety, affordability, accessibility,
an unacceptable administration route for preventive applicability, and efficacy, is far beyond the horizon.
vaccines. (d) As with all eukaryotic cells, long- and
short-term storage of viable parasites needs to be done
in liquid nitrogen and dry ice, respectively. Since main- Acknowledgements
taining a cold chain for vaccine delivery in the tropics is Diane Schad (Max Planck Institute for Infection Biol-
already an enormous logistic challenge, deep-frozen ogy) designed the illustration. Apologies to the many
chains to some of the world’s most remote communities researchers in this field whose work could not be cited
seem too visionary, and alternative long-term ther- directly in this mini-review due to space constrains.
mostabilization techniques need to be developed.
To overcome all four roadblocks and reroute the
development of whole sporozoite vaccines from a References
deadlock to a convincing road map, extensive bioengi- 1 Tuteja R (2007) Malaria – an overview. FEBS J 274,
neering and basic science investments have to be made. 4670–4679.
But the complex infection biology of Plasmodium para- 2 Keeling PJ & Rayner JC (2015) The origins of malaria:
sites might require complex solutions, such as whole there are more things in heaven and earth. . .
parasite vaccines. Parasitology 142(Suppl. 1), S16–S25.
3 Hafalla JCM, Silvie O & Matuschewski K (2011) Cell
biology and immunology of malaria. Immunol Rev 240,
Conclusion and outlook
297–316.
In the past malaria vaccinology has focused on con- 4 Trape J-F, Rogier C, Konate L, Diagne N, Bouganali
cepts that were developed against acute infections with H, Canque B, Legros F, Badji A, Ndiaye G, Ndiaye P

The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies 7


Malaria vaccine perspectives K. Matuschewski

et al. (1994) The Dielmo project: a longitudinal study of X-irradiated sporozoites of Plasmodium berghei. Nature
natural malaria infection and the mechanisms of 216, 160–162.
protective immunity in a community living in a 23 Belnoue E, Costa FTM, Frankenberg T, Vigario AM,
holoendemic area of Senegal. Am J Trop Med Hyg 51, Voza T, Leroy N, Rodrigues MM, Landau I, Snounou
123–137. G & Renia L (2004) Protective T cell immunity against
5 Langhorne J, Ndungu FM, Sponaas AM & Marsh K malaria liver stage after vaccination with live
(2008) Immunity to malaria: more questions than sporozoites under chloroquine treatment. J Immunol
answers. Nat Immunol 9, 725–732. 172, 2487–2495.
6 Zinkernagel RM (2003) On natural and artificial 24 Mueller AK, Labaied M, Kappe SHI & Matuschewski
vaccinations. Annu Rev Immunol 21, 515–546. K (2005) Genetically modified Plasmodium parasites as
7 Casares S & Richie TL (2009) Immune evasion by a protective experimental malaria vaccine. Nature 433,
malaria parasites: a challenge for vaccine design. Curr 164–167.
Opin Immunol 21, 321–330. 25 Friesen J, Silvie O, Putrianti ED, Hafalla JCR,
8 Renia L & Goh YS (2016) Malaria parasites: the great Matuschewski K & Borrmann S (2010) Natural
escape. Front Immunol 7, 463. immunization against malaria: causal prophylaxis with
9 Riley EM & Stewart VA (2013) Immune mechanisms in antibiotics. Sci Transl Med 2, ra49.
malaria: new insights in vaccine development. Nat Med 26 Romero P, Maryanski JL, Corradin G, Nussenzweig
19, 168–178. RS, Nussenzweig V & Zavala F (1989) Cloned
10 Matuschewski K (2013) Murine infection models for cytotoxic T cells recognize an epitope in the
vaccine development: the malaria example. Hum Vaccin circumsporozoite protein and protect against malaria.
Immunother 9, 450–465. Nature 341, 323–326.
11 Sauerwein RW, Rostenberg M & Moorthy VS (2011) 27 Cockburn IA, Amino R, Kelemen RK, Kuo SC,
Experimental human challenge infections can accelerate Radtke A, Mac-Daniel L, Ganusov VV, Zavala F &
clinical malaria vaccine development. Nat Rev Immunol Menard R (2013) In vivo imaging of CD8+ T cell-
11, 57–64. mediated elimination of malaria liver stages. Proc Natl
12 malERA Consultative Group on Vaccines (2011) A Acad Sci USA 110, 9090–9095.
research agenda for malaria eradication: vaccines. PLoS 28 Hafalla JCR, Bauza K, Friesen J, Gonzalez-
Med 8, e1000398. Aseguinolaza G, Hill AVS & Matuschewski K (2013)
13 Crompton PD, Pierce SK & Miller LH (2010) Identification of targets of CD8+ T cell responses to
Advances and challenges in malaria vaccine malaria liver stages by genome-wide epitope profiling.
development. J Clin Invest 120, 4168–4178. PLoS Pathog 9, e1003033.
14 Arama C & Troye-Blomberg M (2014) The path of 29 Fernandez-Ruiz D, Ng WY, Holz LE, Ma JZ, Zaid A,
malaria vaccine development: challenges and Wong YC, Lau LS, Mollard V, Cozijnsen A, Collins N
perspectives. J Intern Med 275, 456–466. et al. (2016) Liver-resident memory CD8+ T cells form
15 Matuschewski K (2006) Vaccine development against a front-line defense against malaria liver-stage infection.
malaria. Curr Opin Immunol 18, 449–457. Immunity 45, 889–902.
16 Matuschewski K & Mueller AK (2007) Vaccines 30 Teo A, Feng G, Brown GV, Beeson JG & Rogerson SJ
against malaria – an update. FEBS J 274, 4680–4687. (2016) Functional antibodies and protection against
17 Frischknecht F & Matuschewski K (2017) Plasmodium blood-stage malaria. Trends Parasitol 32, 888–898.
sporozoite biology. Cold Spring Harb Perspect Med 7, 31 Cowman AF, Berry D & Baum J (2012) The cellular
a025478. https://doi.org/10.1101/cshperspect.a025478. and molecular basis for malaria parasite invasion of the
18 Dups JN, Pepper M & Cockburn IA (2014) Antibody human red blood cell. J Cell Biol 198, 961–971.
and B cell responses to Plasmodium sporozoites. Front 32 Crompton PD, Kayala MA, Traore B, Kayentao K,
Microbiol 5, 625. Ongoiba A, Weis GE, Molina DM, Burk CR, Waisberg
19 Kurosaki T, Kometani K & Ise W (2015) Memory B M, Jasinskas A et al. (2010) A prospective analysis of
cells. Nat Rev Immunol 15, 149–159. the Ab response to Plasmodium falciparum before and
20 Snounou G, Gr€ uner AC, M€ uller-Graf CD, Mazier D after a malaria season by protein microarray. Proc Natl
& Renia L (2005) The Plasmodium sporozoite Acad Sci USA 107, 6958–6963.
survives RTS, S vaccination. Trends Parasitol 21, 33 Portugal S, Pierce SK & Crompton PD (2013) Young
456–461. lives lost as B cells falter: what we are learning about
21 Borrmann S & Matuschewski K (2011) Targeting antibody responses in malaria. J Immunol 190, 3039–
Plasmodium liver stages: better late than never. Trends 3046.
Mol Med 17, 527–536. 34 Greenwood B, Marsh K & Snow R (1991) Why do
22 Nussenzweig RS, Vanderberg J, Most H & Orton C some African children develop severe malaria? Parasitol
(1967) Protective immunity produced by the injection of Today 7, 277–281.

8 The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies


K. Matuschewski Malaria vaccine perspectives

35 Haldar K, Murphy SC, Milner D Jr & Taylor TE 48 Olotu A, Fegan G, Wambua J, Nyangweso G, Leach
(2007) Malaria: mechanisms of erythrocytic infection A, Lievens M, Kaslow DC, Njuguna P, Marsh K &
and pathological correlates of severe disease. Annu Rev Bejon P (2016) Seven-year efficacy of RTS, S/AS01
Pathol 2, 217–249. malaria vaccine among young African children. N Engl
36 Wu Y, Sinden RE, Churcher TS, Tsuboi T & Yusibov J Med 374, 2519–2529.
V (2015) Development of malaria transmission-blocking 49 Stoyanov CT, Boscardin SB, Deroubaix S, Barba-
vaccines: from concept to product. Adv Parasitol 89, Spaeth G, Franco D, Nussenzweig RS, Nussenzweig M
109–152. & Rice CM (2010) Immunogenicity and protective
37 Pulendran B & Ahmed R (2011) Immunological efficacy of a recombinant yellow fever vaccine against
mechanisms of vaccination. Nat Immunol 12, 509–517. the murine malarial parasite Plasmodium yoelii. Vaccine
38 Genton B, Betuela I, Felger I, Al-Yaman F, Anders RF, 28, 4644–4652.
Saul A, Rare L, Baisor M, Lorry K, Brown GV et al. 50 Arama C, Waseem S, Fernandez C, Assefaw-Redda Y,
(2002) A recombinant blood-stage malaria vaccine You L, Rodriguez A, Radosevic K, Goudsmit J,
reduces Plasmodium falciparum density and exerts Kaufmann SH, Reece ST et al. (2012) A recombinant
selective pressure on parasite populations in a phase 1-2b bacille Calmette-Guerin construct expressing the
trial in Papua New Guinea. J Infect Dis 185, 820–827. Plasmodium falciparum circumsporozoite protein
39 Sirima SB, Mordm€ uller B, Milligan P, Ateba Ngoa U, enhances dendritic cell activation and primes for
Kironde F, Atugguba F, Tiono AB, Issifou S, circumsporozoite-specific memory cells in BALB/c mice.
Kaddumukasa M, Bangre O et al. (2016) A phase 2b Vaccine 30, 5578–5584.
randomized, controlled trial of the efficacy of the 51 Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP,
GMZ2 malaria vaccine in African children. Vaccine 34, Doolan DL, Sacci J, de la Vega P, Dowler M, Paul C
4536–4542. et al. (2002) Protection of humans against malaria by
40 Mensah VA, Gueye A, Ndiaya M, Edwards NJ, Wright immunization with radiation-attenuated Plasmodium
D, Anagnostou NA, Syll M, Ndaw A, Abiola A, Bliss falciparum sporozoites. J Infect Dis 185, 1155–1164.
C et al. (2016) Safety, immunogenicity and efficacy of 52 Borrmann S & Matuschewski K (2011) Protective
prime-boost vaccination with ChAd63 and MVA immunity against malaria by ‘natural immunization’: a
encoding ME-TRAP against Plasmodium falciparum question of dose, parasite diversity, or both? Curr Opin
infection in adults in Senegal. PLoS One 11, e0167951. Immunol 23, 500–508.
41 Espinosa DA, Vega-Rodriguez J, Flores-Garcia Y, Noe 53 Kublin JG, Mikolajczak SA, Sack BK, Fishbaugher ME,
AR, Munoz C, Coleman R, Bruck T, Haney K, Stevens Seilie A, Shelton L, VonGoedert T, Firat M, Magee S,
A, Retallack D et al. (2017) The Plasmodium falciparum Fritzen E et al. (2017) Complete attenuation of genetically
cell-traversal protein for ookinetes and sporozoites as a engineered Plasmodium falciparum sporozoites in human
candidate for preerythrocytic and transmission-blocking subjects. Sci Transl Med 9, eaad9099.
vaccines. Infect Immun 85, e00498–16. 54 Roestenberg M, Teirlinck AC, McCall MB, Teelen K,
42 Fried M & Duffy PE (2015) Designing a VAR2CSA- Makamdop KN, Wiersma J, Arens T, Beckers P, van
based vaccine to prevent placental malaria. Vaccine 33, Gemert G, van de Vegte-Bolmer M et al. (2011) Long-
7483–7488. term protection against malaria after experimental
43 Kapulu MC, Da DF, Miura K, Li Y, Blagborough sporozoite inoculation: an open-label follow-up study.
AM, Churcher TS, Nikolaeva D, Williams AR, Lancet 377, 1770–1776.
Goodman AL, Sangare I et al. (2015) Comparative 55 Putrianti ED, Silvie O, Kordes M, Borrmann S &
assessment of transmission blocking vaccine candidates Matuschewski K (2009) Vaccine-like immunity against
against Plasmodium falciparum. Sci Rep 5, 11193. malaria by repeated causal-prophylactic treatment of
44 Cohen J, Nussenzweig V, Nussenzweig R, Vekemans J & liver-stage Plasmodium parasites. J Infect Dis 199,
Leach A (2010) From the circumsporozoite protein to 899–903.
the RTS, S/AS candidate vaccine. Hum Vaccin 6, 90–96. 56 Schats R, Bijker EM, van Gemert GJ, Graumans W,
45 McAleer WJ, Buynak EB, Maigetter RZ, Wampler DE, van de Vegte-Bolmer M, van Lieshout L, Hkas MC,
Miller WJ & Hilleman MR (1984) Human hepatitis B Hermsen CC, Scholzen A, Visser LG et al. (2015)
vaccine from recombinant yeast. Nature 307, 178–180. Heterologous protection against malaria after
46 Didierlaurent AM, Laupeze B, Di Pasquale A, Hergli immunization with Plasmodium falciparum sporozoites.
N, Collignon C & Garcßon N (2017) Adjuvant system PLoS One 10, e0124243.
AS01: helping to overcome the challenges of modern 57 Epstein JE, Paolino K, Richie TL, Sedegah M, Singer
vaccines. Expert Rev Vaccines 16, 55–63. A, Ruben AJ, Chakravarty S, Stafford A, Ruck RC,
47 RTS,S Clinical Trials Partnership (2011) First results of Eappen AG et al. (2017) Protection against Plasmodium
phase 3 trial of RTS, S/AS01 malaria vaccine in falciparum malaria by PfSPZ vaccine. JCI Insights 2,
African children. N Engl J Med 365, 1863–1875. e89154.

The FEBS Journal (2017) ª 2017 Federation of European Biochemical Societies 9

Potrebbero piacerti anche