review on blood stage vaccine against malarai

Upload: rajkishore-rai

Post on 06-Apr-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    1/14

    REVIEW

    The future for blood-stage vaccines against malaria

    Jack S Richards and James G Beeson

    Malaria is a leading cause of mortality and morbidity globally, and effective vaccines are urgently needed. Malaria vaccine

    approaches can be broadly grouped as pre-erythrocytic, blood stage and transmission blocking. This review focuses on blood-

    stage vaccines, and considers the evidence supporting the development of blood-stage vaccines, the advantages and challenges

    of this approach, potential targets, human vaccine studies and future directions. There is a strong rationale for the development

    of vaccines based on antigens of blood-stage parasites. Symptomatic malaria is caused by blood-stage parasitemia and acquired

    immunity in humans largely targets blood-stage antigens. Several candidate vaccines have proved efficacious in animal models

    and at least one vaccine showed partial efficacy in a clinical trial. At present, all leading candidate blood-stage antigens are

    merozoite proteins, located on the merozoite surface or within the apical organelles. Major challenges and priorities include

    overcoming antigenic diversity, identification of protective epitopes, understanding the nature and targets of protective immune

    responses, and defining antigen combinations that give the greatest efficacy. Additionally, objective criteria and approaches are

    needed to prioritize the large number of candidate antigens, and strong candidates need to be tested in clinical trials as quickly

    as possible.

    Immunology and Cell Biology (2009) 87, 377390; doi:10.1038/icb.2009.27; published online 5 May 2009

    Keywords: Plasmodium; malaria; vaccine; blood stage; merozoite

    Malaria is an important cause of global mortality and morbidity.

    There are an estimated 550 million cases of malaria and 1 million

    deaths each year, and around 2.5 billion people are at risk.14 The

    malaria field has seen major achievements over the last decade,

    including development and implementation of preventive strategies,new treatments and advances in basic research.515 Malaria vaccine

    development has been encouraged particularly with trials of the pre-

    erythrocytic vaccine, RTS,S, showing protective efficacy.1621 Malaria

    vaccines can be considered in three broad groups: pre-erythrocytic,

    blood stage and transmission blocking. This review focuses only on

    blood-stage vaccines, and considers the evidence supporting the

    development of blood-stage vaccines, the advantages and challenges

    of this approach, potential targets, human vaccine studies and future

    directions.

    BACKGROUND

    There are five species of Plasmodium that naturally infect humans:P. falciparum, P. vivax, P. ovale, P. malariae and P. knowlesi. These can

    cause asymptomatic infection in those with existing immunity, or a

    spectrum of clinical disease ranging from mild to severe disease and

    death in those lacking substantial immunity. P. falciparum accounts

    for the great majority of morbidity and mortality, but P. vivax

    also causes considerable symptomatic disease. Estimates suggest that

    P. falciparum may be the largest single cause of mortality in children

    below 5 years of age.22 Therefore, the efforts to develop vaccines have,

    to date, largely focused on P. falciparum.

    After an initial period of incubation and replication in the liver, the

    blood-stage of the lifecycle commences when merozoites are released

    from the infected hepatocytes. The 48 to 72 h-cycle involves mero-

    zoites invading erythrocytes, intra-erythrocytic development and

    asexual reproduction, erythrocyte rupture and the release of newmerozoites to continue the cycle (Figure 1). Erythrocyte invasion is

    thought to occur over several steps involving multiple receptorligand

    interactions. Initial attachment of the merozoite to the erythrocyte

    surface is followed by apical reorientation, tight-junction formation,

    entry into the erythrocyte through an actinmyosin motor, and is

    completed by resealing of the erythrocyte membrane.23 This occurs

    over 12min and is the only time that the parasite is directly exposed

    to the extracellular environment during the blood stage. For most of

    the blood-stage cycle, parasites are partially hidden from immune

    recognition inside the erythrocyte, which lacks major histocompat-ibility complex molecules. The parasite exports proteins to the

    erythrocyte surface, enabling cytoadherence to a range of endothelial

    receptors, facilitating sequestration and reducing splenic clearance,

    and these seem to be important immune targets.

    Clinical illness occurs only during blood-stage infection, with the

    liver stage of the infection being asymptomatic. The pathogenesis of

    disease is complex and is reviewed elsewhere.24,25 Despite the com-

    plexity of the parasite lifecycle, a high level of antigenic diversity and

    parasite mechanisms of immune evasion, naturally acquired immu-

    nity to malaria does develop after repeated exposure. This immunity

    affords protection against symptomatic disease, high-density

    Received 6 February 2009; accepted 15 March 2009; published online 5 May 2009

    Infection and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia

    Correspondence: Dr JG Beeson, Department of Infection and Immunity, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia.

    E-mail: [email protected]

    Immunology and Cell Biology (2009) 87, 377390& 2009 Australasian Society for Immunology Inc. All rights reserved 0818-9641/09 $32.00

    www.nature.com/icb

    http://dx.doi.org/10.1038/icb.2009.27mailto:[email protected]://www.nature.com/icbhttp://www.nature.com/icbmailto:[email protected]://dx.doi.org/10.1038/icb.2009.27
  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    2/14

    parasitemia and death; however, it is less effective at preventing low-

    level parasitemia.26,27 Naturally acquired immunity seems to predo-

    minantly target blood-stage parasites.28

    Effective immunity involves both humoral- and cell-mediated

    immune responses, which gives some cross-species and cross-strain

    protection, though this seems to be limited.29,30 The importance of

    antibodies has been supported by the passive transfer of immunoglo-

    bulin from immune individuals conferring protective benefit to non-immune individuals.31,32 Immune effector mechanisms are poorly

    understood and the relative importance of different mechanisms has

    not been quantified. Antibodies may have a role in preventing

    merozoite invasion, clearance of infected erythrocytes, prevention of

    adhesion and sequestration in the vasculature, and prevention of

    schizont rupture. Cytophilic IgG subclasses seem to be particularly

    important.3336 They probably facilitate parasite clearance in the

    spleen through opsonization for phagocytosis, antibody-dependent

    cell-mediated cytotoxicity, and complement-mediated lysis.26,3739

    Antibodies do not act alone and there is increasing evidence of the

    importance of T cells, not only in providing B cell help, but in

    activating Th1 responses, which help mediate antibody effector

    mechanisms, as well as cell-mediated effector mechanisms.

    ADVANTAGES AND CHALLENGES OF A BLOOD-STAGE VACCINE

    It is likely that a highly effective malaria vaccine will need to be

    multivalent and may need to incorporate antigens of several lifecycle

    stages. There is a strong rationale for developing blood-stage vaccines

    as part of this strategy. Pathogenesis of malarial disease results fromblood-stage infection and studies in humans and animal models have

    clearly established that immune responses targeting blood-stage anti-

    gens can protect against disease or facilitate control of parasite-

    mia.31,32,40,41 Immunization with blood-stage antigens, mainly

    merozoite antigens, has been shown to be protective in a number of

    animal models using different antigens and there was some protective

    effect with one blood-stage vaccine tested in humans.4246 At present,

    the leading blood-stage vaccine candidates are all merozoite proteins,either located on the merozoite surface or contained within the apical

    organelles (Figure 2).

    1

    2 3 4

    1

    2 3

    4

    5

    Figure 1 The blood-stage lifecycle of Plasmodium. (a) Merozoites are an

    extracellular form of Plasmodium (1) that attach and invade erythrocytes

    (2). The parasite then matures and divides through asexual replication inside

    the erythrocyte (3). The expression of parasite proteins on the surface of the

    infected erythrocyte enables interactions with receptors on the endothelial

    surface, facilitating sequestration of parasite-infected erythrocytes in various

    organs. After asexual replication, the schizont form ruptures and releases

    new merozoites into the circulation (4). (b) Merozoite invasion of

    erythrocytes. Merozoites are initially free in the intravascular space after

    schizont rupture (1), and the process of erythrocyte invasion commences

    when a merozoite binds to the surface of an erythrocyte (2). The merozoite

    then reorients its apical end to come into contact with the erythrocyte

    surface (3). A series of irreversible high-affinity ligandreceptor interactions

    then occur between the parasite and the erythrocyte, enabling tight-junction

    formation (4) and entry of the erythrocyte through an actinmyosin motor. As

    it does so, the parasite invaginates the erythrocyte membrane to form theparasitophorous vacuole. Outer surface proteins are partially cleaved as the

    merozoite enters the erythrocyte (5).

    Micronemes

    Merozoite surface

    Dense granules

    Nucleus

    Rhoptries

    Apicoplast

    Mitochondrion

    Apical end

    MSP1, 2, 4, 5, 10 (GPI anchored)MSP3, 6, 7, 9 (ABRA)GLURPSERAS-antigen6-cys family (GPI anchored)

    AMA1

    EBA175, 140, 181MTRAPPTRAMPASP

    Rh 1, 2a, 2b, 4, 5RAP1, 2, 3RhopH1, 2, 3RAMA

    Merozoite surface proteins

    Micronemes

    Rhoptries

    Figure 2 The structure and major antigens of the P. falciparum merozoite. The apical end of the merozoite has specific organelles involved in erythrocyte

    invasion, including the paired rhoptries and micronemes, which are thought to expel proteins to bind to erythrocyte receptors during invasion. The merozoite

    also has dense granules, and an apicoplast (a plastid remnant). Merozoite ligands and vaccine candidate antigens are present in the organelles and on the

    surface of the merozoite. Surface proteins may be GPI anchored or associated with GPI-anchored proteins by molecular interactions. Listed are known

    proteins of the merozoite surface and organelles, but many others remain to be identified or characterized.

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    378

    mmunology and Cell Biology

    http://www.nature.com/icbhttp://www.nature.com/icb
  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    3/14

    One hurdle for the continued development of blood-stage vaccines

    is the perceived lack of progress despite many years of research. There

    are many reasons why development of blood-stage vaccines has not

    progressed more rapidly, but slow progress has been common to all

    strategies including pre-erythrocytic stage candidates. Clearly greater

    effort and funding is needed to facilitate the development and

    evaluation of promising vaccines. The Combination B vaccine,

    which showed protection against parasitemia, was conducted over10 years ago. However, no further testing of this vaccine or any of its

    components in phase II studies has been conducted since. At the time

    of writing, only four blood-stage vaccines have been tested in phase II

    trials. Of the two that are published, Combination B showed some

    degree of efficacy, whereas merozoite surface protein 1 (MSP1)-42 did

    not. Although RTS,S, which is based on the circumsporozoite protein,

    has been efficacious in several trials, another pre-erythrocytic vaccine,multi-epitope thrombospondin-related adhesion protein (ME-

    TRAP)47,48 was not protective. It is likely that few of the vaccines

    that are eventually tested in clinical trials will prove to be sufficiently

    efficacious; therefore, poor outcomes with specific antigens should not

    justify abandonment of an overall strategy.

    Recently, there has been a renewed call for the eradication ofmalaria.49,50 An effective vaccine will probably be needed to achieve

    eradication, in addition to other existing measures. Part of the

    renewed push for eradication of malaria is an emphasis on approaches

    to reduce malaria transmission and not just clinical illness. Therefore,

    vaccines inducing sterile immunity and/or transmission-reducing

    activity would be preferred. The role of blood-stage vaccines as partof this strategy has been questioned, as the objective of blood-stage

    vaccines is to reduce parasitemia and prevent clinical illness. However,

    data from animal models and clinical studies suggests that controlling

    parasite density reduces the generation of gametocytes in the blood

    stream. Therefore, it seems likely that an effective blood-stage vaccine

    will have benefit in reducing transmission, in addition to preventing

    clinical illness.

    Perhaps the greatest challenge in developing a blood-stage vaccine isovercoming antigenic diversity. Most, if not all, important antigens

    show substantial polymorphism.5154 Antigenic diversity has evolved

    to facilitate immune evasion and vaccine approaches need to account

    for this such that they will cover the majority of strains causing

    infection and disease. This approach may be difficult or impossible if a

    large number of different allelic forms of an antigen are required in a

    single vaccine. However, the degree of polymorphism is much less for

    some antigens than others, and most antigens have domains or

    regions that are less polymorphic or are conserved. These may be

    suitable targets of vaccines to overcome the problem of antigenicdiversity. Some antigens, such as P. falciparum erythrocyte membrane

    protein 1 (PfEMP1), are encoded by large multigene families and

    parasites can switch the expression of different genes to facilitate

    immune evasion. Recent studies have also shown that some merozoite

    protein families can be differentially expressed to facilitate immune

    evasion.5557

    Further challenges are presented by potential mechanisms of

    immune interference seen during natural exposure to Plasmodium

    infection. As natural immunity is principally directed against blood-

    stage antigens, this may be more important than for pre-erythrocytic

    and transmission-blocking vaccines. For example, non-functionalantibodies that impair the binding of functionally important anti-

    bodies to merozoite antigens have been identified.58,59 There are also

    likely to be effects of earlier exposure or maternally transferred

    antibodies in altering immune responses to a vaccine.60,61 Some

    data suggests that there may be compromised dendritic cell function

    and suppression or deletion of memory B cells, and long-lived plasma

    cells during infection.62

    VACCINE APPROACHES

    Current day immunization evolved from the live-attenuated or whole-

    killed vaccines of Jenner, Pasteur and Koch. Even today, the majority

    of licensed vaccines use whole organisms. Recent advances have led to

    renewed interest in using genetically attenuated parasites as a basis fora malaria vaccine. Most malaria vaccine research, however, has

    followed subunit approaches in which antigens are identified, purified,

    characterized and then immunologically evaluated.63 This approach

    lends itself to using recombinant antigens, peptides and, more

    recently, viral vectors.

    Whole blood-stage parasites

    The whole parasite approach for malaria has partly evolved from

    studies of radiation-attenuated sporozoite studies, reviewed else-

    where.64 Some information on blood-stage immunity can be gleaned

    from early studies in which deliberate infection with Plasmodia was

    used to treat paresis in humans. These studies indicated that infection

    led to predominantly strain-specific immunity that limited sympto-matic disease, but did not prevent re-infection.65 Other studies in

    animal models also suggest that immunity induced in this way is

    largely strain-specific.66,67 More recently a study administered live P.

    falciparum parasites intravenously at low dose to five patients,

    followed by early anti-malarial treatment.68 Four patients were rechal-

    lenged with live parasites of the same strain, and three did not developany detectable parasitemia before final anti-malarial treatment. Sur-

    prisingly, these patients developed cell-mediated immune responses,

    but a limited antibody response.

    Whole organism approaches have a range of potential advantages

    and these are more thoroughly reviewed elsewhere.6971 In short, they

    have the benefit of not requiring the precise determination of epitopes

    or antigens. Rather the whole organism enables a vast array of antigens

    to be delivered, essentially providing a multi-epitope vaccine, with ahigh probability of antigens being in their native conformation. There

    are several important challenges to this approach. Safety is a major

    concern, as current technology requires that parasites are cultured in

    human erythrocytes, which is accompanied by a risk of serious blood-

    borne infections. Large-scale production of whole parasites ensuring

    consistent quality and dose is more challenging than for recombinant

    or synthetic vaccines. With live-attenuated parasites there is also the

    concern that attenuated parasites might revert to a non-attenuated

    state, although genetic attenuation by targeting multiple genes would

    make this unlikely. There are likely to be significant challenges inproducing and delivering live blood-stage parasites for widespread

    use. In vitro culture of P. vivax is difficult to sustain and large-scale

    production is not currently feasible.

    Recombinant antigens

    An advantage of this approach is the ability to direct immune

    responses to a specified region or epitope. This has the potential to

    maximize protective responses whilst minimizing undesired responses.

    A further advantage is the suitability for large-scale antigen produc-

    tion under good manufacturing practice. The inherent difficulty is

    identifying protective target antigens for development, or specificdomains of proteins, and the likelihood that multiple antigens

    would be required to induce an effective response and overcome

    antigenic diversity. Expression of many Plasmodium proteins is

    problematic because they are often conformationally complex and/

    or large. Most recombinant antigens require some form of adjuvant to

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    379

    Immunology and Cell Biology

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    4/14

    elicit sufficient immune responses. There is much uncertainty about

    which proteinadjuvant combinations will be well-tolerated, yet elicit

    an effective immune response with sustained duration.

    Peptide vaccines

    Peptide vaccines have the potential to deliver precisely defined

    epitopes with a large scale of production at relatively low cost. 72,73

    Using a totally synthetic approach is perhaps the safest manufacturingmethod, and largely avoids concerns of microbiological contamina-

    tion. There is also the potential to synthesize or conjugate multiple

    epitopes into a single construct. However, there are major challenges to

    synthesizing conformational epitopes; peptide antigens with limited

    epitopes may not elicit adequate immune responses in children and in

    certain human leukocyte antigen (HLA) subgroups.74 There are

    approaches to overcome some of these barriers in which peptidescan be conjugated to T-cell epitopes, lipid moieties, or suspended in

    liposomes or virus-like particles.75 Similarly, peptides are more amen-

    able to engineering to potentially improve immunogenicity and can be

    built into scaffolds to mimic native conformation.72,76 At present, a lack

    of knowledge regarding functionally relevant or protective epitopes of

    major antigens is a major impediment for the development of peptide-based vaccines, and clearly represents an area for further studies.

    DNA and viral-vectored vaccines

    The application of DNA and viral-vectored approaches for malaria

    vaccines is beginning to emerge and is reviewed elsewhere.7781 Most

    employ prime-boost strategies, but use different viral vectors and arange of different formulations, including liposomes, virosomes,

    microspheres and nanoparticles.82 They offer the advantage of includ-

    ing multiple antigens and have been shown to effectively induce both

    cell-mediated immunity and specific antibodies.

    POTENTIAL TARGETS AND THEIR PRIORITIZATION

    Prioritizing antigens

    Until recently, research efforts have focused on a handful of individualantigens. Perhaps the greatest advance for malarial research in the past

    decade has come from completion of major genome projects, includ-

    ing P. falciparum, P. vivax, P. knowlesi, P. berghei, P. chabaudi and P.

    yoelii.15,8388 Thousands of novel proteins have been identified,5,89

    providing new opportunities for vaccine development. However, at

    present, little is known about their structure and function or their role

    as targets of protective immunity.

    Several criteria can be used to objectively prioritize known and

    predicted antigens for further study.15,85 Table 1 lists some important

    properties that could be used in this process, and rates five vaccinecandidates against these criteria as an example. Targeting proteins that

    have an important function seems obvious, but the function is known

    for very few antigens. In some instances structural features may be

    suggestive of function (for example, epidermal growth factor domains

    in MSP1-19), and others have been classified as essential or non-

    essential on the basis of whether they can be genetically disrupted or

    not.90 Surface exposure or location within the apical organelles of the

    merozoite is likely to be highly relevant, but defining surface-exposed

    epitopes within antigens has proven to be more challenging. An

    increasing amount of data is emerging about polymorphisms, which

    may indicate targets under the pressure of immune selection, and

    regions or proteins of limited diversity. Abundance of proteins varies

    widely (for example, MSP1 vs MSP4), which may have implicationsfor the extent and nature of immune responses. Structural features

    and the size of antigens have implications for vaccine manufacture.

    Studies of naturally acquired immunity can facilitate the identification

    of antigen-specific immune responses involved in protective immunity

    and the identification and characterization of immune effector

    mechanisms. At present, there is an over reliance on using standardimmunoassays of uncertain functional significance, and the develop-

    ment and application of functional assays is greatly needed.

    Potential targets and results from vaccine trials

    Human trials of blood-stage malaria vaccines have only been carried

    out with whole blood-stage parasites and merozoite antigens to date.

    Table 2 summarizes clinical trials that have been conducted or are in

    progress. Here, we discuss findings from these trials and the propertiesof vaccine antigens. Unfortunately, it was beyond the scope of this

    review to comprehensively include vaccine testing in animal models.

    Table 1 Pre-clinical criteria for prioritizing candidate antigens for vaccine development

    Criteria

    Current vaccine candidatesa

    MSP1-42 MSP2 MSP3 LSP AMA1 EBA175 RII

    Known functionb No No No No Yes

    Essential or important functionc Yes Yes No/unknown Yes Yes

    Location Merozoite surface Merozoite surface Merozoite surface Apical Apical

    Abundance of antigen High High High Low Low

    Limited diversityd No No Yes No Yes

    Possible target of protective immunity in humanse Yes Yes Yes Yes Yes

    Antibodies inhibit parasite growth in vitrof Yes Unknown Yesg Yes Yes

    Vaccination induces protective immunity in non-human primate models h Yes Unknown Yes Yes Yes

    Phase II vaccination in humansi Not protective Protective Trial ong oing Trial ong oing No t tested

    Abbreviations: AMA, apical membrane antigen; EBA, erythrocyte-binding antigens; LSP, long synthetic peptide; MSP, merozoite surface protein.aFive leading vaccine antigens were selected as illustrative examples.bEBA175 binds to glycophorin A on the erythrocyte surface.cEssential function defined as an inability to knock out the gene in transfection studies. MSP3 is not essential, and its function is unknown.dMSP3 is polymorphic, but the LSP construct is highly conserved.eStudies of endemic populations have found naturally acquired antibodies associated with protection from symptomatic disease (this does not mean that antibodies are causally responsible forprotection).fStandard assay is the growth inhibition assay without immune cells.gMSP3 antibodies do not have direct growth inhibitory activity but show antibody-dependent cellular inhibition.hMSP2 has not been tested in non-human primates. It is not possible to test it in murine models.iPhase II vaccination in humans is meant to serve as a comparison of how the criteria predict the final outcomes.

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    380

    mmunology and Cell Biology

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    5/14

    Table 2 Summary of vaccine studies in humansa

    Stage Antigen Vaccine Trial

    phase

    Site Participants Status References

    Erythrocytic Whole parasite Low-dose live parasites/drug cureb I/II Australia five adults Completed 68

    MSP1-42 FMP1/ASO2 I USA 15 adults Completed 108

    FMP1/ASO2 I Kenya 40 adults Completed 109

    FMP1/ASO2 I Mali 40 Completed 110

    FMP1/ASO2 I Kenya 135 children

    14 years

    Completed 242

    FMP1/ASO2c II Kenya 400 children

    14 years

    Completed NP

    MSP1-42 (3D7 or FVO)/alum I USA 60 adults Completed 112,243

    MSP1-42-C1/alhydrogel (+/CPG 7909) I USA 60 adults Completed NP

    MSP1 FMP010/ASO1 I USA 25 adults Completed NP

    MSP2 MSP2 (3D7+FC27) I Australia 45 adults Completed NP

    MSP3 MSP3 LSP/alum/ISA720 I Switzerland 35 adults Completed 131,132

    MSP3 LSP/alum I Burkina Faso 30 adults Completed 135

    MSP3 LSP/alum I Tanzania 45 children

    12 years

    Recruiting NP

    MSP3 LSP/alum I Burkina Faso 45 children

    12 years

    Recruiting NP

    MSP3 LSP/alumc II Mali 378 children

    12 years

    Recruiting NP

    AMA1 AMA1-C1/alhydrogel I USA 30 adults Completed 147

    AMA1-C1/alhydrogel I Mali 54 adults Completed 148

    AMA1-C1/alhydrogel I Mali 36 children

    23 years

    Completed 149

    AMA1-C1/alhydrogelc II Mali 900 children

    23 years

    Completed NP

    AMA1-C1/alhydrogel (short schedule) I USA 18 adults Completed NP

    C1/alhydrogel +/CPG 7909 I USA 75 adults Completed 150

    C1/alhydrogel +/CPG 7909 I Mali 24 adults Completed NP

    C1/alhydrogel +/CPG 7909 I Mali 200 children

    13 years

    Recruiting NP

    C1/alhydrogel +CPG 7909b I/II UK 21 adults Not started NP

    C1/alhydrogel +/CPG 7909 (high/low) I USA No. not stated-adults Completed NP

    C1/alhydrogel +/CPG 7909 (saline/phosphate) I USA 24 adults Completed NP

    C1/alhydrogel/ISA720 I USA 28 adults Completed NP

    FMP2.1/ASO2 I USA 23 adults Completed 151

    FMP2.1/ASO2 I Mali 60 adults Completed NP

    FMP2.1/ASO2 I Mali 100 children

    16 years

    Completed NP

    FMP2.1/ASO2c II Mali 400 children

    16 years

    Active NP

    FMP2.1/ASO2/ASO1 I USA 35 adults Completed NP

    PfAMA1-FVO/alhydrogel/ISA720/ASO2 I Netherlands 56 adults Completed 155

    PfAMA1-FVO/alhydrogel/ISA720/ASO2 I Mali 60 adults Recruiting NP

    EBA175 EBA175 RII-NG I USA 80 adults Completed NP

    GLURP GLURP LSP I Netherlands 36 adults Completed 187

    SERA5 SE36 I Japan Not specified Completed 244

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    381

    Immunology and Cell Biology

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    6/14

    Merozoite surface antigens. Merozoite surface protein 1 coats

    the merozoite surface and is GPI (glycosylphosphatidylinositol)

    anchored.91,92 It undergoes several proteolytic processing steps creat-

    ing several fragments, of which the 42 kDa (MSP1-42) and 19 kDafragment (MSP1-19) have been most studied.93 The crystal structure

    for MSP1-19 has been elucidated.94,95 Its function seems to be

    essential, probably for initial attachment of merozoites to the ery-

    throcyte surface through a complex with MSP9.96,97 There are two

    major allelic variants, MAD20 and K1, with highly polymorphic (for

    example, Block 2) and relatively conserved regions (for example,

    MSP1-19).98 Antibodies to MSP1-19 have been variably associated

    with protection from symptomatic disease.99103 Vaccine-induced

    antibodies have invasion inhibitory activity and may also inhibit

    MSP1 processing.103,104 Vaccination of mice and primates protected

    from subsequent challenge in some studies.43,105107 Human vaccinestudies include MSP1-42 (3D7) formulated with adjuvant, ASO2A,

    (GlaxoSmithKline, Brentford, Middlesex, UK) that induced growth

    inhibitory antibodies.108110 However, a Phase II trial among 12 to 47-

    month-old children in Kenya found no protective effect.111 Phase I

    studies of MSP1-42 (3D7 and FVO) in Alhydrogel (Sigma-Aldrich,

    St Louis, MO, USA) generated moderate antibody responses, but

    insufficient in vitro inhibitory activity.112 MSP1-19 has only been

    tested in human vaccine studies as part of the chimeric vaccine,

    PfCP2.9, which combines domain 3 of apical membrane antigen 1(AMA1) (3D7).113,114 In phase I studies, adjuvanted with Montanide

    ISA720, (SEPPIC SA, Paris, France) there was high immunogenicity,

    but significant reactogenicity.115,116 Antibody invasion inhibitory activ-

    ity was weak, as were T-cell responses. Human trials with P. vivax

    antigens have not yet been carried out. However, immunization using

    PvMSP1-19 had some protective effect in a non-human primate

    challenge.117

    Merozoite surface protein 2 (MSP2) is also GPI anchored to the

    merozoite surface. Its function remains unclear, but also seems to be

    essential.90 There are two major allelic families (FC27 and 3D7) each

    with a highly variable central region consisting of several repeats andconserved flanking regions.54 Natural immunity results in high levels

    of antibodies, predominantly IgG3 and a moderate association with

    protection from symptomatic disease.36,118 MSP2 (3D7) was included

    in the Combination A vaccine, adsorbed onto Alum and found to be

    safe, but poorly immunogenic.119 Five patients were challenged by

    Table 2 Continued

    Stage Antigen Vaccine Trial

    phase

    Site Participants Status References

    MSP1/MSP2/RESA Combination B/ISA720 I Australia 32 adults Completed 120

    Combination B/ISA720 I Australia 36 adults Completed 120

    Combination B/ISA720b I/II Australia 17 adults Completed 121

    Combination B/ISA720 I Papua New Guinea 12 adults Completed 122

    Combination B/ISA720c I/II Papua New Guinea 120 children

    59 years

    Completed 42,123125

    AMA1/MSP1-19 PfCP2.9/ISA720 I China 52 adults Completed 115

    PfCP2.9/ISA720 I China 70 adults Completed 116

    MSP3/GLURP GMZ2/alum I Germany 30 adults Recruiting NP

    GMZ2/alum I Gabon 40 adults Recruiting NP

    GMZ2/alum I Gabon 30 children

    15 years

    Recruiting NP

    SPf66d

    Multistage Polyprotein

    (six antigens)

    FP9-PP, MVA-PP I UK 35 adults Completed NP

    FP9-PP, MVA-PPe II UK 26 adults Completed NP

    AMA1/CSP PEV301 (AMA1), PEV302 (CSP),

    PEV3A (PEV301+PEV302)

    I Switzerland 46 adults Completed 159

    PEV3A (AMA1+CSP), FP9 ME-TRAPe I/II UK 30 adults Completed 158

    A MA1/CS P A MA1/CS P/Adeno5f I/II USA 70 adults Recruiting NP

    NYVAC-Pf7 CSP, SSP2, LSA1, MSP1, AMA1, SERA, Pfs25e I/II USA 59 adults Completed 220

    MSP2/ CSP Combi nation A/alume I/II Switzerland 39 adults Completed 119

    Abbreviations: AMA, apical membrane antigen; EBA, erythrocyte-binding antigens; GLURP, glutamate-rich protein; LSP, long synthetic peptide; MSP, merozoite surface protein; NP, Not published;

    SERA, serine repeat antigen; SSP, sporozoite surface protein; TRAP, thrombospondin-related adhesion protein.aSome of this information is gathered from www.clinicaltrials.gov and is the most accurate publically available data at the time of writing.bBlood stage challenge.cNatural exposure.dSPf66 underwent numerous clinical studies which are comprehensively reviewed elsewhere.eSporozoite challenge.

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    382

    mmunology and Cell Biology

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    7/14

    mosquito bite, but no protective benefit was detected. Combination

    B comprised of MSP1 (190LCS.T3, N terminal end, K1 allele), MSP2

    (3D7) and a part of the ring-infected erythrocyte surface antigen

    (RESA) in Montanide ISA720. Phase I/II studies among children in

    Papua New Guinea reported lower parasite densities in the vaccine

    recipients.42,120123 Furthermore, the risk of infection with the MSP2

    allelic variant included in the vaccine (3D7) was much lower than the

    risk with the alternate allelic variant (FC27), suggesting that theantigen mediating the major protective effect was MSP2.124,125

    Merozoite surface protein 3 (MSP3) has neither a GPI anchor nor

    typical transmembrane domain, but is non-covalently attached to the

    merozoite surface. Its function is unknown, but it seems to interact

    with acidic basic repeat antigen (ABRA, also known as MSP9) and can

    be genetically disrupted.126 It has allelic dimorphism (K1 and 3D7)

    with the C-terminal end being conserved and the N terminal endbeing highly polymorphic.127129 Antibodies have been associated with

    protective immunity in studies of acquired immunity in humans.130

    MSP3 vaccines based on the conserved C-terminal end of the protein

    expressed as a long synthetic peptide have been tested in clinical

    trials.131,132 The vaccine was immunogenic, mostly of IgG3 subclass,

    but antibody titers were not sustained. Antibodies have not beenshown to directly inhibit invasion, but instead seem to act in

    cooperation with monocytes to inhibit parasite replication in

    vitro.131,133,134 A phase I study in Burkina Faso failed to show any

    vaccine-related boosting of MSP3 antibody responses above the high

    level seen at enrollment.135 There are two phase I studies and a phase

    II study examining this vaccine in children from endemic countries.There are also three phase I studies currently recruiting using a MSP3/

    GLURP (glutamate-rich protein) chimera.136

    Proteins of the apical organelles of merozoites. The apical organelles of

    the merozoite contain a number of known or predicted invasionligands. One of these, AMA1 is thought to play an essential role in

    erythrocyte invasion, however, its precise function is not known.137 Its

    crystal structure has recently been solved and it is thought to form acomplex with rhoptry neck protein 4 (RON4).138,139 AMA1 is highly

    polymorphic and allele-specific antibodies are not cross-protective;

    presenting a major challenge to vaccine development.52,140143

    Acquired antibodies have been associated with protection from

    symptomatic disease in humans.36,144 Antibodies have been shown

    to inhibit invasion in functional assays, although non-functional

    antibodies are also prevalent.141,145 Immunization with AMA1 was

    protective in primate studies.146

    There have been numerous AMA1 human vaccine studies, mainly

    phase I studies to date. The AMA-C1 (3D7 and FVO strains) vaccine,adjuvanted with Alhydrogel, was safe and immunogenic, and gener-

    ated antibodies that inhibited parasite growth in vitro.141,147 This was

    confirmed in Malian adults and children, but antibody levels were not

    maintained.148,149 A phase II study is presently underway. There are a

    range of studies examining other adjuvants. Adding CPG 7909 to the

    Alhydrogel formulation led to three- to fourfold higher antibody

    titers.150 There was significant invasion inhibition, but this was

    relatively variant-specific. In phase I trials, the FMP2.1 vaccine (3D7

    strain) adjuvanted with ASO2A generated high-titre antibodies that

    inhibited growth and AMA1 processing.144,151,152 It also resulted in

    cell-mediated immune responses. Phase II studies are currently under-way. ASO1B is also being explored as an alternative adjuvant (unpub-

    lished). The PfAMA1-FVO construct has been formulated with a

    range of adjuvants, including Alhydrogel, Montanide ISO720 and

    ASO2.153155 Antibody titers were highest with ASO2, and were

    maintained for longer and inhibited in vitro invasion. This vaccine

    is being studied in Malian adults. The NMRC-M3V-Ad-PfCA vaccine

    contains CSP (circumsporozoite protein) and AMA1 (3D7) using an

    adenovirus 5 vector. The PEV301, PEV302, PEV3A (AMA1/CSP

    peptide) vaccines use influenza virosomes to deliver an AMA1-

    containing peptide from loop I of domain III (K1 strain), as well as

    a double loop of NPNA repeats of the CSP peptide. 156,157 Phase I and

    II studies showed good safety and immunogenicity with high antibody

    levels, high avidity and recognition of blood-stage parasites.158,159

    Sporozoite challenge failed to afford sterile protection or to prolong

    time to parasitemia, but vaccine recipients had lower parasite growth

    rates and had morphologically altered parasites detected.

    The erythrocyte-binding antigens (EBAs) are a family of antigens,

    which are orthologues of the Duffy-binding protein of P. vivax and

    include EBA175, EBA140 (BAEBL), EBA181 (JESEBL), MAEBL, EBL1

    and EBA165 (pseudogene). On each of these proteins, there is areceptor-binding domain (region II), which is structurally related

    between proteins, but differs in sequence; the crystal structure was

    recently solved for EBA175.160 They are located in the micronemes

    and are secreted onto the merozoite surface just before invasion.161163

    It is known that EBA175 and EBA140 interact with glycophorin A and

    C, respectively.164167

    Vaccine-induced antibodies in animals inhibitinvasion.165,168,169 There is limited sequence polymorphism in the

    receptor-binding domains, but this does not seem to affect the activity

    of vaccine-induced antibodies.170 Overall, there is little polymorphism

    throughout the protein. Acquired antibodies among children have

    been associated with protection from symptomatic disease in some

    studies.171,172 Parasites can vary the expression and use of these ligandsduring invasion, which seems to facilitate immune evasion, and the

    EBAs seem to function in a complementary manner with the

    reticulocyte binding-like homologs (PfRHs).56 The only human vac-

    cine study is a phase I study of EBA175 region 2 adsorbed onto

    aluminum phosphate (unpublished). An EBA175 vaccine was shown

    to have efficacy in a primate challenge.173

    Very little work has been carried out on the development of P vivax

    vaccines. However, the Duffy-binding protein (DBP) is a strongvaccine candidate. DBP binds to DARC (Duffy antigen/receptor for

    chemokines) during parasite invasion of reticulocytes.174,175 DBP

    seems essential for invasion, and resistance to P. vivax in humans

    is conferred by a lack of DARC expression.176,177 DBP is localized in

    the micronemes,174 and is thought to be essential for the formation of

    the tight junction. The receptor-binding face of the protein has

    little polymorphism suggesting that it may be possible to develop a

    vaccine that predominantly targets the conserved region to block the

    DBPDARC interaction. Studies have shown that vaccine-induced

    antibodies in animals and acquired human antibodies can inhibitreticulocyte invasion and inhibit the binding of DBP to DARC.178,179

    Furthermore, DBP-binding inhibitory antibodies have been

    associated with acquired protective immunity in humans and immu-

    nization with DBP was partially protective in a primate challenge

    model.180,181

    Other merozoite antigens. Glutamate-rich protein is expressed on the

    merozoite surface and in liver stage schizonts,182 but its function is

    unknown. It has two repeat regions (R1 and R2), and an N-terminal

    non-repeat region which has limited diversity (R0).183 Naturally

    acquired antibodies of IgG1 and IgG3 subclasses (depending on theantigenic region) are variably associated with protection.184186

    Human vaccine studies have focused on a long synthetic peptide

    derived from the R0 region. A phase I study with glutamate-rich

    protein long synthetic peptide adjuvanted in alum or Montanide

    ISA720 generated antibodies that inhibited parasite growth in coop-

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    383

    Immunology and Cell Biology

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    8/14

    eration with monocytes.187 It has also been examined in combination

    with MSP3.

    Future malaria vaccine research will need to assess the potential of

    other antigens, which have been identified, but have not yet been

    tested in humans. The reticulocyte binding-like homologs (PfRHs) are

    a family of invasion ligands that have homology with the reticulocyte-

    binding proteins of P. vivax. They are found in the neck of the

    rhoptries and are known to bind to erythrocytes, but receptors remainto be identified. Antibodies are acquired naturally and have been

    strongly associated with protection.188 Antibodies to PfRH1 and

    PfRH2 have been shown to inhibit invasion, and polymorphism in

    the PfRH proteins is limited. MSP4 is GPI anchored to the merozoite

    surface, has limited diversity and elicits naturally acquired antibodies,

    though these are not associated with protection.189191 Vaccination

    with the P. yoelii homologue, MSP4/5, is protective in murinechallenge.192,193 There many other promising candidates, amongst

    these are the SERA (serine repeat antigen) family, RhopH1-3, and the

    6-cysteine domain family.

    Variant surface antigens. Antibodies against variant surface antigens

    (VSAs) expressed on the surface of the infected erythrocyte arethought to contribute to strain-specific protection in humans and

    primate models.66,194 Studies in monkeys also suggest that VSAs are

    important targets of protective antibodies. VSAs include PfEMP1,

    rifins, subtelomeric variable open reading frame (STEVOR), and

    others;25 to date, only PfEMP1 has been established as a major target

    of antibodies.195,196 PfEMP1 is encoded by the var multigene family,

    and different var genes encode PfEMP1 variants with different anti-

    genic properties.197 The immune response against VSAs is thought to

    contribute to strain-specific protection, but only PfEMP1 has been

    established as a major target of antibodies.194 There is a high level of

    polymorphism between different var genes within a single genome

    and between vargenes in different genomes. Rifins and STEVOR are

    also encoded by large polymorphic gene families. The major barrier to

    developing VSAs as vaccines is their very high degree of antigenic

    diversity and capacity for clonal antigenic variation. Studies are

    beginning to identify subsets of var genes associated with severe

    disease that are likely to be important immune targets.198200 Some

    studies have suggested that cross-reactive antibodies can be generateddespite extensive polymorphism and a vaccine trial in primates

    showed protection against homologous parasites.201,202 A specific

    variant of PfEMP1 (var2csa) is expressed by P. falciparum-infected

    erythrocytes that mediates adhesion to the placental lining during

    pregnancy and seems to be an important target of acquired anti-

    bodies.203 Var2csa is less antigenically diverse than other PfEMP1

    variants and antibodies that cross-react with different placental-bind-

    ing isolates have been shown in humans and animal models. There-

    fore, it may be possible to develop a vaccine inducing broad coverageagainst different placental-binding variants by targeting conserved

    and/or common epitopes of var2csa.204206

    Other antigens. Glycosylphosphatidylinositol is a glycolipid that

    anchors a number of erythrocytic stage proteins to the membrane,

    including MSP1, MSP2 and serine proteases. It has been reported to

    have toxin-like effects and induce pro-inflammatory responses and

    clinical symptoms in murine models (reviewed in Schofield andGrau207). Immunization with a synthetic version of the GPI glycan

    showed some protection against clinical illness in mice, providing a

    proof-of-concept for this type of approach.208 The vaccine did not

    have any affect on parasite replication and it is likely that GPI would need

    to be included in a multivalent vaccine rather than being used alone.

    Other human vaccine studies. The SPf66 vaccine consists of a 45

    amino-acid peptide, which is a chimera of three merozoite-derived

    epitopes interspersed with the PNANP sporozoite peptide of CSP.

    Most studies have examined it adjuvanted with aluminum hydroxide.

    It showed mild protective efficacy in a South American and a

    Tanzanian study, but failed to show any benefit in the studies

    conducted in Gambia or Thailand.209217 Much controversy sur-

    rounded these studies and it was the subject of a Cochrane review,which concluded that there was no justification for further trials using

    the same formulation.218

    The NYVAC-Pf7 vaccine incorporates seven genes into an attenu-

    ated vaccinia virus. These genes included blood-stage antigens, MSP1,

    AMA1, SERA as well CSP, liver-stage antigens 1 (LSA1), sporozoite

    surface protein 2(SSP2) and the 25 kDa sexual-stage antigen, SPf25.219

    In a human sporozoite challenge model, antibody levels in vaccinated

    individuals were found to have a slight delay in the prepatent

    period.220 The FP9-PP, MVA-PP vaccine is principally a pre-erythro-

    cytic vaccine, using the fowlpox 9 (FP9) and the modified vaccinia

    virus Ankara (MVA) polypeptide (PP) vaccines, which contain

    proteins that are also expressed during the erythrocytic stage.221

    RECENT ADVANCES AND CONTINUING CHALLENGES

    Determining structure and function

    A more complete understanding of the structure and function of

    Plasmodium proteins, particularly those involved in host-cell adhesion

    and invasion will greatly facilitate the identification, rationalization

    and evaluation of candidate antigens. As expected, there will be a lagbetween advances in genomics to that of proteomics, and particularly

    functional molecular biology. Remarkably little is known about the

    molecular interactions that occur during merozoite invasion, with the

    binding partners of most merozoite antigens yet to be identified.222224

    This lack of knowledge limits more targeted approaches against

    functionally important domains, the significance of sequence poly-

    morphism in antigens and the development of functional immuno-

    logical assays for vaccine trials. Structural modeling is beginning toinform molecular interactions, including antibodies.95,138,160,225230

    This has the potential to assist in epitope mapping and determining

    the mechanisms of immune evasion.231,232

    Identifying correlates of immunity and developing

    functional assays

    The identification of robust correlates of immunity and assays that

    measure functionally relevant immune responses would be of

    enormous benefit for the development of blood-stage vaccines.

    Unfortunately there is still much to be done to achieve this goal.Studies of acquired human immunity have largely focused on only a

    small number of antigens using standard immunoassays that provide

    little indication of the functional relevance of the responses. Very little

    is known about functional effector mechanisms and there are few

    assays examining these issues.38,39,131,233 Future studies will need to

    accommodate the testing of responses to a large number of candidate

    antigens that are now being identified, and greater effort is required to

    define immune effector mechanisms and their contribution to immu-

    nity in humans. For example, the predominant assays for merozoite

    vaccines measure direct antibody growth inhibition or antibody-

    dependent cellular inhibition.38,39 These assays have been carriedout for many years, but have rarely been applied to large population

    studies. The further refinement and optimization of these assays and

    the development of high-throughout methods, such as using flow

    cytometry, and transgenic parasites to identify and quantify antigen-

    specific responses will advance our ability to understand and

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    384

    mmunology and Cell Biology

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    9/14

    measure immune responses in studies of acquired and vaccine-

    induced immunity.56,59,103,180,234240

    Pathway to vaccine development: animal models

    and human challenge

    In many instances, malaria vaccine research has taken candidate

    antigens into rodent studies to determine immunogenicity and

    evaluate efficacy using challenge models. Although this approachhas the benefits of being readily available and relatively affordable,

    there are considerable limitations. Human malarias cannot infect mice

    and rodent malarias lack many important antigens of P. falciparum

    and P. vivax, which means that murine challenge models cannot be

    used to assess their efficacy. Additionally, there are important differ-

    ences between mice and humans in the nature of immune and

    pathogenic responses to malaria. More recently, there have been effortsto humanize the mouse model to make it more representative of

    human immune responses.131,241 These models may provide an

    informative approach to evaluate blood-stage vaccines, but further

    studies are needed to establish their relevance. Vaccine testing often

    proceeds to challenge studies in non-human primates. This is

    expensive, has limited availability, and for P. falciparum, is largelyrestricted to Aotus and Saimiri models. Given the limitations of animal

    models for testing malaria vaccines, the safety and immunogenicity of

    candidate vaccines in humans should be determined as rapidly as

    possible. It could be argued that vaccine candidates that have a very

    strong rationale for development, based on a set of widely accepted

    biological and immunological criteria, could proceed through tohuman trials without the need for evaluation in animal models.

    With the need to progress candidate vaccines more quickly into

    human trials, there is an increasing interest in testing vaccines in

    humans using a live parasite challenge; this has been carried out either

    by direct inoculation of blood-stage parasites or with sporozoites

    through mosquito bite. There are a number of significant concerns

    about the relevance of this model for evaluating the efficacy of blood-

    stage vaccines, and it remains to be established that it is a validapproach. A principle problem with this approach is that participants

    must be treated as soon as blood-stage parasitemia is detected for

    ethical reasons. Blood-stage immunity acts primarily by limiting

    parasite density and protecting against symptomatic illness rather

    than preventing parasitization per se. However, it is not possible to

    assess clinical illness or high-density parasitemia as an outcome using

    this approach, and there is only a limited ability to measure parasite

    replication rates before treatment. On-going studies might shed light

    on these issues and clarify whether non-human primate challenge orthe human challenge model is more informative for evaluating blood-

    stage vaccines. Of interest, the Combination B vaccine did not show

    any effect on blood-stage replication in a challenge carried out in

    vaccinated malaria-nave individuals, but did show protective effect in

    a phase II trial in children in Papua New Guinea. 42,121

    CONCLUSIONS AND PERSPECTIVES

    In this review, we have highlighted a number of issues surrounding the

    development of vaccines against malaria based on antigens expressed

    in the blood-stage, as well as summarized recent progress and the

    current state of the field. Effective blood-stage vaccines will almost

    certainly need to be comprised of several different antigens, or severaldifferent alleles of a single antigen, to overcome antigenic diversity and

    the capacity ofP. falciparum for immune evasion or perhaps to achieve

    more potent anti-parasite activity by combining antigens that induce

    antibodies with synergistic activities against parasite growth. There are

    presently a substantial number of blood-stage vaccine candidates and

    a multitude of new antigens are likely to emerge from recent genomic

    and proteomic insights. Therefore, there is a strong need for

    approaches to validate and prioritize potential vaccine candidates

    for further development.

    The recent success of the RTS, S vaccine in several phase II trials

    conducted in African children has changed the landscape of malaria

    vaccine development. RTS,S is now entering phase III studies and it

    seems likely that it will eventually become licensed for broad use.RTS,S represents a first generation vaccine against malaria, and the

    challenge ahead will be to develop the next generation of vaccines that

    have greater efficacy and provide more sustained protection. This may

    be achieved by combining the RTS,S antigen with other antigens, or

    the development of vaccines targeting different antigens altogether.

    There is a strong rationale for the continued development and testing

    of blood-stage vaccines highlighted in this review, and for investigatingthe inclusion of blood-stage antigens in combination with pre-

    erythrocytic antigens, such as the RTS,S antigen. The importance of

    blood-stage antigens as targets of acquired immunity in humans

    together with the demonstrated efficacy of different blood-stage

    antigens in animal models and the partial efficacy of at least one

    blood-stage vaccine in humans provides reasonable cause to beoptimistic about the development of blood-stage vaccines against

    malaria.

    ACKNOWLEDGEMENTSJGB is supported by a Clinical Career Development Award, and JSR by a

    Medical Postgraduate Research Scholarship from the National Health and

    Medical Research Council of Australia. Thanks to Creepy Crawley Cartoons for

    preparing the figures.

    1 Snow RW, Guerra CA, Noor AM, Myint HY, Hay SI. The global distribution of clinical

    episodes of Plasmodium falciparum malaria. Nature 2005; 434: 214217.

    2 Guerra CA, Gikandi PW, Tatem AJ, Noor AM, Smith DL, Hay SI et al. The limits andintensity of Plasmodium falciparum transmission: implications for malaria control and

    elimination worldwide. PLoS Med 2008; 5: e38.

    3 Hay SI, Snow RW. The Malaria Atlas Project: developing global maps of malaria risk.

    PLoS Med 2006; 3: e473.

    4 World Health Organization. World Health Report: Make Every Mother and Child Count.

    WHO: Geneva, 2005; cited 15/3/06. Available from: http://www.who.int/whr/2005/en/

    index.html.

    5 Gardner MJ, Hall N, Fung E, White O, Berriman M, Hyman RW et al. Genome

    sequence of the human malaria parasite Plasmodium falciparum. Nature 2002;

    419: 498511.

    6 Carlton JM, Adams JH, Silva JC, Bidwell SL, Lorenzi H, Caler E et al. Comparative

    genomics of the neglected human malaria parasite Plasmodium vivax. Nature 2008;

    455: 757763.

    7 Carlton JM, Angiuoli SV, Suh BB, Kooij TW, Pertea M,Silva JC etal. Genome sequence

    and comparative analysis of the model rodent malaria parasite Plasmodium yoelii

    yoelii. Nature 2002; 419: 512519.

    8 Pain A, Bohme U, Berry AE, Mungall K, Finn RD, Jackson AP et al. The genome of the

    simian and human malaria parasite Plasmodium knowlesi. Nature 2008; 455:799803.

    9 Holt RA, Subramanian GM, Halpern A, Sutton GG, Charlab R, Nusskern DR et al. The

    genome sequence of the malaria mosquito Anopheles gambiae. Science 2002; 298:

    129149.

    10 Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J et al. Initial

    sequencing and analysis of the human genome. Nature 2001; 409: 860921.

    11 Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, Sutton GG et al. The sequence of

    the human genome. Science 2001; 291: 13041351.

    12 Noor AM, Mutheu JJ, Tatem AJ, Hay SI, Snow RW. Insecticide-treated net coverage in

    Africa: mapping progress in 20002007. Lancet 2009; 373: 5867.

    13 Karunajeewa HA, Mueller I, Senn M, Lin E, Law I, Gomorrai PS et al. A trial of

    combination antimalarial therapies in children from Papua New Guinea. N Engl J Med

    2008; 359: 25452557.

    14 Malarias watershed. Nature 2008; 455: 707.

    15 Hall N, Karras M, Raine JD, Carlton JM, Kooij TW, Berriman M et al. A comprehensive

    survey of the Plasmodium life cycle by genomic, transcriptomic, and proteomic

    analyses. Science 2005; 307: 8286.

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    385

    Immunology and Cell Biology

    http://www.who.int/whr/2005/en/index.htmlhttp://www.who.int/whr/2005/en/index.htmlhttp://www.who.int/whr/2005/en/index.htmlhttp://www.who.int/whr/2005/en/index.html
  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    10/14

    16 Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, Aide P et al. Duration of

    protection with RTS,S/AS02A malaria vaccine in prevention of Plasmodium falci-

    parum disease in Mozambican children: single-blind extended follow-up of a rando-

    mised controlled trial. Lancet 2005; 366: 20122018.

    17 Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, Milman J et al. Efficacy of the

    RTS,S/AS02A vaccine against Plasmodium falciparuminfection and disease in young

    African children: randomised controlled trial. Lancet 2004; 364: 14111420.

    18 Bejon P, Lusingu J, Olotu A, Leach A, Lievens M, Vekemans J et al. Efficacy of RTS,S/

    AS01E vaccine against malaria in children 517 months of age. N Engl J Med 2008;

    359: 25212532.19 Abdulla S, Oberholzer R, Juma O, Kubhoja S, Machera F, Membi C et al. Safety and

    immunogenicity of RTS,S/AS02D malaria vaccine in infants. N Engl J Med 2008;

    359: 25332544.

    20 Bojang KA, Milligan PJ, Pinder M, Vigneron L, Alloueche A, Kester KE et al. Efficacy

    of RTS,S/AS02 malaria vaccine against Plasmodium falciparum infection in

    semi-immune adult men in The Gambia: a randomised trial. Lancet 2001; 358:

    19271934.

    21 Bojang KA, Olodude F, Pinder M, Ofori-Anyinam O, Vigneron L, Fitzpatrick S et al.

    Safety and immunogenicty of RTS,S/AS02A candidate malaria vaccine in Gambian

    children. Vaccine 2005; 23: 41484157.

    22 Elliott SR, Beeson JG. Estimating the burden of global mortality in children agedo5

    years by pathogen-specific causes. Clin Infect Dis 2008; 46: 17941795.

    23 Gilson PR, Crabb BS. Morphology and kinetics of the three distinct phases of red

    blood cell invasion by Plasmodium falciparum merozoites. Int J Parasitol 2009; 39:

    9196.

    24 Miller LH, Baruch DI, Marsh K, Doumbo OK. The pathogenic basis of malaria. Nature

    2002; 415: 673679.

    25 Beeson JG, Brown GV. Pathogenesis of Plasmodium falciparum malaria: the roles of

    parasite adhesion and antigenic variation. Cell Mol Life Sci 2002; 59: 258271.26 Marsh K, Kinyanjui S. Immune effector mechanisms in malaria. Parasite Immunol

    2006; 28: 5160.

    27 Michon P, Cole-Tobian JL, Dabod E, Schoepflin S, Igu J, Susapu M et al. The risk of

    malarial infections and disease in Papua New Guinean children. Am J Trop Med Hyg

    2007; 76: 9971008.

    28 Doolan DL, Dobano C, Baird JK. Acquired immunity to malaria. Clin Microbiol Rev

    2009; 22: 1336.

    29 Beeson JG, Osier FH, Engwerda CR. Recent insights into humoral and cellular

    immune responses against malaria. Trends Parasitol 2008; 24: 578584.

    30 Douradinha B, Mota MM, Luty AJ, Sauerwein RW. Cross-species immunity in malaria

    vaccine development: two, three, or even four for the price of one? Infect Immun

    2008; 76: 873878.

    31 Cohen S, McGregor I, Carrington S. Gamma-globulin and acquiredimmunity tohuman

    malaria. Nature 1961; 192: 733737.

    32 McGregor IA. The passive transfer of human malarial immunity. Am J Trop Med Hyg

    1964; 13 (Suppl): 237239.

    33 Tongren JE, Drakeley CJ, McDonald SL, Reyburn HG, Manjurano A, Nkya WM et al.

    Target antigen, age, and duration of antigen exposure independently regulate immu-noglobulin G subclass switching in malaria. Infect Immun 2006; 74: 257264.

    34 Taylor RR, Smith DB, Robinson VJ, McBride JS, Riley EM. Human antibody response

    to Plasmodium falciparum merozoite surface protein 2 is serogroup specific and

    predominantly of the immunoglobulin G3 subclass. Infect Immun 1995; 63:

    43824388.

    35 Nebie I, Diarra A, Ouedraogo A, Soulama I, Bougouma EC, Tiono AB et al. Humoral

    responses to Plasmodium falciparum blood-stage antigens and association with

    incidence of clinical malaria in children living in an area of seasonal malaria

    transmission in Burkina Faso, West Africa. Infect Immun 2008; 76: 759766.

    36 Stanisic DI, Richards JS, McCallum FJ, Michon P, King CL, Schoepflin S et al. IgG

    subclass-specific responses against Plasmodium falciparum merozoite antigens are

    associated with control of parasitemia and protection from symptomatic illness. Infect

    Immun2009; 77: 11651174.

    37 Bergmann-Leitner ES, Duncan EH, Mullen GE, Burge JR, Khan F, Long CA et al.

    Critical evaluation of different methods for measuring the functional activity of

    antibodies against malaria blood stage antigens. Am J Trop Med Hyg 2006; 75:

    437442.

    38 Bouharoun-Tayoun H, Attanath P, Sabchareon A, Chongsuphajaisiddhi T, Druilhe P.

    Antibodies that protect humans against Plasmodium falciparum blood stages do noton their own inhibit parasite growth and invasion in vitro, but act in cooperation with

    monocytes. J Exp Med 1990; 172: 16331641.

    39 Bouharoun-Tayoun H, Oeuvray C, Lunel F, Druilhe P. Mechanisms underlying the

    monocyte-mediated antibody-dependent killing of Plasmodium falciparum asexual

    blood stages. J Exp Med 1995; 182: 409418.

    40 Diggs CL, Osler AG. Humoral immunity in rodent malaria. III: studies on the site of

    antibody action. J Immunol 1975; 114: 12431247.

    41 Mitchell GH, Butcher GA, Voller A, Cohen S. The effect of human immune IgG on the

    in vitro development of Plasmodium falciparum. Parasitology 1976; 72: 149162.

    42 Genton B, Betuela I, Felger I, Al-Yaman F, Anders RF, Saul A et al. A recombinant

    blood-stage malaria vaccine reduces Plasmodium falciparum density and exerts

    selective pressure on parasite populations in a phase 1-2b trial in Papua New Guinea.

    J Infect Dis 2002; 185: 820827.

    43 Ling IT, Ogun SA, Holder AA. Immunization against malaria with a recombinant

    protein. Parasite Immunol1994; 16: 6367.

    44 Perera KL, Handunnetti SM, Holm I, Longacre S, Mendis K. Baculovirus merozoite

    surface protein 1 C-terminal recombinant antigens are highly protective in a natural

    primate model for human Plasmodium vivax malaria. Infect Immun 1998; 66:

    15001506.

    45 Crewther PE, Matthew ML, Flegg RH, Anders RF. Protective immune responses to

    apical membrane antigen 1 of Plasmodium chabaudi involve recognition of strain-

    specific epitopes. Infect Immun 1996; 64: 33103317.

    46 Collins WE, Pye D, Crewther PE, Vandenberg KL, Galland GG, Sulzer AJ et al.

    Protective immunity induced in squirrel monkeys with recombinant apical membrane

    antigen-1 of Plasmodium fragile. Am J Trop Med Hyg 1994; 51: 711719.

    47 Moorthy VS, Imoukhuede EB, Milligan P, Bojang K, Keating S, Kaye P et al.

    A randomised, double-blind, controlled vaccine efficacy trial of DNA/MVA ME-TRAPagainst malaria infection in Gambian adults. PLoS Med 2004; 1: e33.

    48 Bejon P, Mwacharo J, Kai O, Mwangi T, Milligan P, Todryk S et al. A phase 2b

    randomised trial of the candidate malaria vaccines FP9 ME-TRAP and MVA ME-TRAP

    among children in Kenya. PLoS Clin Trials 2006; 1: e29.

    49 Feachem R, Sabot O. A new global malaria eradication strategy. Lancet 2008; 371:

    16331635.

    50 World Health Organization. The Role Back Malaria Partnership. Global Malaria Action

    Plan. WHO: Geneva, 2008.

    51 Volkman SK, Hartl DL, Wirth DF, Nielsen KM, Choi M, Batalov S et al. Excess

    polymorphisms in genes for membrane proteins in Plasmodium falciparum. Science

    2002; 298: 216218.

    52 Polley SD, Conway DJ. Strong diversifying selection on domains of the Plasmodium

    falciparum apical membrane antigen 1 gene. Genetics 2001; 158: 15051512.

    53 Miller LH, Roberts T, Shahabuddin M, McCutchan TF. Analysis of sequence diversity

    in the Plasmodium falciparum merozoite surface protein-1 (MSP-1). Mol Biochem

    Parasitol 1993; 59: 114.

    54 Smythe JA, Peterson MG, Coppel RL, Saul AJ, Kemp DJ, Anders RF. Structural

    diversity in the 45-kilodalton merozoite surface antigen of Plasmodium falciparum.

    Mol Biochem Parasitol 1990; 39: 227234.55 Cortes A. Switching Plasmodium falciparumgenes on and off for erythrocyte invasion.

    Trends Parasitol 2008; 24: 517524.

    56 Persson KE, McCallum FJ, Reiling L, Lister NA, Stubbs J, Cowman AF et al. Variation

    in use of erythrocyte invasion pathways by Plasmodium falciparum mediates evasion

    of human inhibitory antibodies. J Clin Invest 2008; 118: 342351.

    57 Baum J, Maier AG, Good RT, Simpson KM, Cowman AF. Invasion by P. falciparum

    merozoites suggestsa hierarchy ofmolecular interactions. PLoSPathog2005; 1: e37.

    58 Guevara Patino JA, Holder AA, McBride JS, Blackman MJ. Antibodies that inhibit

    malaria merozoite surface protein-1 processing and erythrocyte invasion are blocked

    by naturally acquired human antibodies. J Exp Med 1997; 186: 16891699.

    59 Miura K, Zhou H, Moretz SE, Diouf A,TheraMA, Dolo A etal. Comparison of biological

    activity of human anti-apical membrane antigen-1 antibodies induced by natural

    infection and vaccination. J Immunol 2008; 181: 87768783.

    60 Stanisic DI, Martin LB, GattonML, Good MF. Inhibition of 19-kDaC-terminal region of

    merozoite surface protein-1-specific antibody responses in neonatal pups by mater-

    nally derived 19-kDa C-terminal region of merozoite surface protein-1-specific

    antibodies but not whole parasite-specific antibodies. J Immunol 2004; 172:

    55705581.61 Wykes MN, Good MF. What really happens to dendritic cells during malaria? Nature

    Rev 2008; 6: 864870.

    62 Wykes MN, Liu XQ, Beattie L, Stanisic DI, Stacey KJ, Smyth MJ et al. Plasmodium

    strain determines dendritic cell function essential for survival from malaria. PLoS

    Pathog 2007; 3: e96.

    63 Xiao L, Rafi-Janajreh A, Patterson P, Zhou Z, Lal AA. Adjuvants and malaria vaccine

    development. Chem Immunol 2002; 80: 343365.

    64 Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP, Doolan DL et al. Protection of

    humans against malaria by immunization with radiation-attenuated Plasmodium

    falciparum sporozoites. J Infect Dis 2002; 185: 11551164.

    65 Collins WE, Jeffery GM. A retrospective examination of secondary sporozoite- and

    trophozoite- induced infections with Plasmodium falciparum: development of para-

    sitologic and clinical immunity following secondary infection. Am J Trop Med Hyg

    1999; 61: 2035.

    66 Brown KN, Brown IN. Immunity to malaria: antigenic variation in chronic infections of

    Plasmodium knowlesi. Nature 1965; 208: 12861288.

    67 Jones TR, Obaldia III N, Gramzinski RA, Hoffman SL. Repeated infection of Aotus

    monkeys with Plasmodium falciparum induces protection against subsequent chal-

    lenge with homologous and heterologous strains of parasite. Am J Trop Med Hyg2000; 62: 675680.

    68 Pombo DJ, Lawrence G, Hirunpetcharat C, Rzepczyk C, Bryden M, Cloonan N et al.

    Immunity to malaria after administration of ultra-low doses of red cells infected with

    Plasmodium falciparum. Lancet 2002; 360: 610617.

    69 Renia L, Gruner AC, Mauduit M, Snounou G. Vaccination against malaria with live

    parasites. Expert Rev Vaccines 2006; 5: 473481.

    70 Wykes M, Good MF. A case for whole-parasite malaria vaccines. Int J Parasitol 2007;

    37: 705712.

    71 Pinzon-Charry A, Good MF. Malaria vaccines: the case for a whole-organism approach.

    Expert Opin Biol Ther 2008; 8: 441448.

    72 Patarroyo ME, Patarroyo MA. Emerging rules for subunit-based, multiantigenic,

    multistage chemically synthesized vaccines. Acc Chem Res 2008; 41: 377386.

    73 Corradin G, Spertini F, Verdini A. Medicinal application of long synthetic peptide

    technology. Expert Opin Biol Ther 2004; 4: 16291639.

    74 Satterthwait AC, Chiang LC, Arrhenius T, Cabezas E, Zavala F, Dyson HJ et al. The

    conformational restriction of synthetic vaccines for malaria. Bull World Health Organ

    1990; 68: 1725.

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    386

    mmunology and Cell Biology

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    11/14

    75 Jackson DC, Purcell AW, Fitzmaurice CJ, Zeng W, Hart DN. The central role played by

    peptides in the immune response and the design of peptide-based vaccines against

    infectious diseases and cancer. Curr Drug Targets 2002; 3: 175196.

    76 Riley EM, Olerup O, Bennett S, Rowe P, Allen SJ, Blackman MJ et al. MHC and

    malaria: the relationship between HLA class II alleles and immune responses to

    Plasmodium falciparum. Int Immunol 1992; 4: 10551063.

    77 Doolan DL, Hoffman SL. DNA-based vaccines against malaria: status and promise of

    the multi-stage malaria DNA vaccine operation. Int J Parasitol 2001; 31: 753762.

    78 Dunachie SJ, Hill AV. Prime-boost strategies for malaria vaccine development. J Exp

    Biol 2003; 206: 37713779.79 Moore AC, Hill AV. Progress in DNA-based heterologous prime-boost immunization

    strategies for malaria. Immunol Rev 2004; 199: 126143.

    80 Li S, Locke E, Bruder J, Clarke D, Doolan DL, Havenga MJ et al. Viral vectors for

    malaria vaccine development. Vaccine 2007; 25: 25672574.

    81 Reyes-Sandoval A, Harty JT, Todryk SM. Viral vector vaccines make memory T cells

    against malaria. Immunology 2007; 121: 158165.

    82 Tyagi RK, Sharma PK, Vyas SP, Mehta A. Various carrier system(s)-mediated genetic

    vaccination strategies against malaria. Expert Rev Vaccines 2008; 7: 499520.

    83 Sanders PR, Cantin GT, Greenbaum DC, Gilson PR, Nebl T, Moritz RL et al.

    Identification of protein complexes in detergent-resistant membranes of Plasmodium

    falciparum schizonts. Mol Biochem Parasitol 2007; 154: 148157.

    84 Sanders PR, Gilson PR, Cantin GT, Greenbaum DC, Nebl T, Carucci DJ et al. Distinct

    protein classes including novel merozoite surface antigens in Raft-like membranes of

    Plasmodium falciparum. J Biol Chem 2005; 280: 4016940176.

    85 Mu J, Awadalla P, Duan J, McGee KM, Keebler J, Seydel K et al. Genome-wide

    variation and identification of vaccine targets in the Plasmodium falciparum genome.

    Nature genet 2007; 39: 126130.

    86 Doolan DL, Aguiar JC, Weiss WR, Sette A, Felgner PL, Regis DP et al. Utilization of

    genomic sequence information to develop malaria vaccines. J Exp Biol 2003; 206:37893802.

    87 Winzeler EA. Applied systems biology and malaria. Nature Rev 2006; 4: 145151.

    88 Winzeler EA. Malaria research in the post-genomic era. Nature2008; 455: 751756.

    89 Florens L, Washburn MP, Raine JD, Anthony RM, Grainger M, Haynes JD et al. A

    proteomic view of the Plasmodium falciparum life cycle. Nature 2002; 419:

    520526.

    90 Cowman AF, Crabb BS. Invasion of red blood cells by malaria parasites. Cell 2006;

    124: 755766.

    91 Gerold P, Schofield L,Blackman MJ, Holder AA, Schwarz RT. Structural analysis of the

    glycosyl-phosphatidylinositol membrane anchor of the merozoite surface proteins-1

    and -2 of Plasmodium falciparum. Mol Biochem Parasitol 1996; 75: 131143.

    92 Holder AA, Freeman RR. Immunization against blood-stage rodent malaria using

    purified parasite antigens. Nature 1981; 294: 361364.

    93 Blackman MJ, Ling IT, Nicholls SC, Holder AA. Proteolytic processing of the

    Plasmodium falciparum merozoite surface protein-1 produces a membrane-bound

    fragment containing two epidermal growth factor-like domains. Mol Biochem Parasitol

    1991; 49: 2933.

    94 Chitarra V, Holm I, Bentley GA, Petres S, Longacre S. The crystal structure ofC-terminal merozoite surface protein 1 at 1.8 A resolution, a highly protective malaria

    vaccine candidate. Mol Cell 1999; 3: 457464.

    95 Pizarro JC, Chitarra V, Verger D, Holm I, Petres S, Dartevelle S et al. Crystal structure

    of a Fab complex formed with PfMSP1-19, the C-terminal fragment of merozoite

    surface protein 1 from Plasmodium falciparum: a malaria vaccine candidate. J Mol

    Biol 2003; 328: 10911103.

    96 Goel VK, Li X, Chen H, Liu SC, Chishti AH, Oh SS. Band 3 is a host receptor binding

    merozoite surface protein 1 during the Plasmodium falciparum invasion of erythro-

    cytes. Proc Natl Acad Sci USA 2003; 100: 51645169.

    97 Li X, Chen H, Oo TH, Daly TM, Bergman LW, Liu SC et al. A co-ligand complex anchors

    Plasmodium falciparummerozoites to the erythrocyte invasion receptor band 3. J Biol

    Chem 2004; 279: 57655771.

    98 Polley SD, Tetteh KK, Cavanagh DR, Pearce RJ, Lloyd JM, Bojang KA et al. Repeat

    sequences in block 2 of Plasmodium falciparum merozoite surface protein 1 are

    targets of antibodies associated with protection from malaria. Infect Immun 2003;

    71: 18331842.

    99 Riley EM, Allen SJ, Wheeler JG, Blackman MJ, Bennett S, Takacs B et al. Naturally

    acquired cellular and humoral immune responses to the major merozoite surface

    antigen (PfMSP1) of Plasmodium falciparum are associated with reduced malariamorbidity. Parasite Immunol1992; 14: 321337.

    100Branch OH, Udhayakumar V, Hightower AW, Oloo AJ, Hawley WA, Nahlen BL et al.

    A longitudinal investigation of IgG and IgM antibody responses to the merozoite

    surface protein-1 19-kiloDalton domain of Plasmodium falciparumin pregnantwomen

    and infants: associations with febrile illness, parasitemia, and anemia. Am J Trop Med

    Hyg 1998; 58: 211219.

    101Egan AF, Burghaus P, Druilhe P, Holder AA, Riley EM. Human antibodies to the 19 kDa

    C-terminal fragment of Plasmodium falciparum merozoite surface protein 1 inhibit

    parasite growth in vitro. Parasite Immunol1999; 21: 133139.

    102Dodoo D, Theander TG, Kurtzhals JA, Koram K, Riley E, Akanmori BD et al. Levels of

    antibody to conserved parts of Plasmodium falciparum merozoite surface protein 1 in

    Ghanaian children are not associated with protection from clinical malaria. Infect

    Immun1999; 67: 21312137.

    103ODonnell RA, de Koning-Ward TF, Burt RA, Bockarie M, Reeder JC, Cowman AF et al.

    Antibodies against merozoite surface protein (MSP)-1(19) are a major component of

    the invasion-inhibitory response in individuals immune to malaria. J Exp Med 2001;

    193: 14031412.

    104Blackman MJ, Scott-Finnigan TJ, Shai S, Holder AA. Antibodies inhibit the protease-

    mediated processing of a malaria merozoite surface protein. J Exp Med 1994; 180:

    389393.

    105Singh S, Kennedy MC, Long CA, Saul AJ, Miller LH, Stowers AW. Biochemical and

    immunological characterization of bacterially expressed and refolded Plasmodium

    falciparum 42-kilodalton C-terminal merozoite surface protein 1. Infect Immun2003;

    71: 67666774.

    106Daly TM, Long CA. A recombinant 15-kilodalton carboxyl-terminal fragment of

    Plasmodium yoelii yoelii 17XL merozoite surface protein 1 induces a protective

    immune response in mice. Infect Immun 1993; 61: 24622467.107Kumar S, Yadava A, Keister DB, Tian JH, Ohl M, Perdue-Greenfield KA et al.

    Immunogenicity and in vivoefficacy of recombinant Plasmodium falciparummerozoite

    surface protein-1 in Aotus monkeys. Mol Med 1995; 1: 325332.

    108Ockenhouse CF, Angov E, Kester KE, Diggs C, Soisson L, Cummings JF et al. Phase I

    safety and immunogenicity trial of FMP1/AS02A, a Plasmodium falciparum MSP-1

    asexual blood stage vaccine. Vaccine 2006; 24: 30093017.

    109Stoute JA, Gombe J, Withers MR, Siangla J, McKinney D, Onyango M et al. Phase 1

    randomized double-blind safety and immunogenicity trial of Plasmodium falciparum

    malaria merozoite surface protein FMP1 vaccine, adjuvanted with AS02A, in adults in

    western Kenya. Vaccine 2007; 25: 176184.

    110Thera MA, Doumbo OK, Coulibaly D, Diallo DA, Sagara I, Dicko A et al. Safety and

    allele-specific immunogenicity of a malariavaccine in Malian adults: results of a phase

    I randomized trial. PLoS Clin Trials 2006; 1: e34.

    111Ogutu BR, Apollo OJ, McKinney D, Okoth W, Siangla J, Dubovsky F et al. Blood stage

    malaria vaccine eliciting high antigen-specific antibody concentrations confers no

    protection to young children in western Kenya. PLoS ONE 2009; 4: e4708.

    112Malkin E, Long CA, Stowers AW, Zou L, Singh S, Macdonald NJ et al. Phase 1 study of

    two merozoite surface protein 1 (MSP1(42)) vaccines for Plasmodium falciparum

    malaria. PLoS Clin Trials 2007; 2: e12.113Pan W, Huang D, Zhang Q, Qu L, Zhang D, Zhang X et al. Fusion of two malaria vaccine

    candidate antigens enhances product yield, immunogenicity, and antibody-mediated

    inhibition of parasite growth in vitro. J Immunol 2004; 172: 61676174.

    114Langermans JA, Hensmann M, van Gijlswijk M, Zhang D, Pan W, Giersing BK et al.

    Preclinical evaluation of a chimeric malaria vaccine candidate in montanide ISA

    720((R)): immunogenicity and safety in Rhesus macaques. Human Vaccin 2006; 2:

    222226.

    115Hu J, Chen Z, Gu J, Wan M, Shen Q, Kieny MP et al. Safety and immunogenicity of a

    malaria vaccine, Plasmodium falciparum AMA-1/MSP-1 chimeric protein formulated

    in montanide ISA 720 in healthy adults. PLoS ONE 2008; 3: e1952.

    116Malkin E, Hu J, Li Z, Chen Z, Bi X, Reed Z et al. A Phase 1 trial of PfCP2.9: An AMA1/

    MSP1 chimeric recombinant protein vaccine for Plasmodium falciparum malaria.

    Vaccine 2008; 26: 68646873.

    117Valderrama-Aguirre A, Quintero G, Gomez A, Castellanos A, Perez Y, Mendez F et al.

    Antigenicity, immunogenicity, and protective efficacy of Plasmodium vivax MSP1

    PV200l: a potential malaria vaccine subunit. Am J Trop Med Hyg 2005; 73: 1624.

    118Polley SD, Conway DJ, Cavanagh DR, McBride JS, Lowe BS, Williams TN et al. High

    levels of serum antibodies to merozoite surface protein 2 of Plasmodium falciparumare associated with reduced risk of clinical malaria in coastal Kenya. Vaccine 2006;

    24: 42334246.

    119Sturchler D, Berger R, Rudin C, Just M, Saul A, Rzepczyk C et al. Safety, immuno-

    genicity, and pilot efficacy of Plasmodium falciparum sporozoite and asexual blood-

    stage combination vaccine in Swiss adults. Am J Trop Med Hyg1995; 53: 423431.

    120Saul A, Lawrence G, Smillie A, Rzepczyk CM, Reed C, Taylor D et al. Human phase I

    vaccine trials of 3 recombinant asexual stage malaria antigens with Montanide

    ISA720 adjuvant. Vaccine 1999; 17: 31453159.

    121Lawrence G, Cheng QQ, Reed C, Taylor D, Stowers A, Cloonan N et al. Effect of

    vaccination with 3 recombinant asexual-stage malaria antigens on initial growth

    rates of Plasmodium falciparum in non-immune volunteers. Vaccine 2000; 18:

    19251931.

    122Genton B, Al-Yaman F, Anders R, Saul A, Brown G, Pye D et al. Safety and

    immunogenicity of a three-component blood-stage malaria vaccine in adults living

    in an endemic area of Papua New Guinea [In Process Citation]. Vaccine 2000; 18:

    25042511.

    123Genton B, Al-Yaman F, Betuela I, Anders RF, Saul A, Baea K et al. Safety and

    immunogenicity of a three-component blood-stage malaria vaccine (MSP1, MSP2,

    RESA) against Plasmodium falciparum in Papua New Guinean children. Vaccine2003; 22: 3041.

    124Fluck C, Schopflin S, Smith T, Genton B, Alpers MP, Beck HP et al. Effect of the

    malaria vaccine Combination B on merozoite surface antigen 2 diversity. Infect Genet

    Evol 2007; 7: 4451.

    125Fluck C, Smith T, Beck HP, Irion A, Betuela I, Alpers MP et al. Strain-specific humoral

    response to a polymorphic malaria vaccine. Infect Immun 2004; 72: 63006305.

    126 Mills KE, Pearce JA, Crabb BS, Cowman AF. Truncation of merozoite surface protein 3

    disrupts its trafficking and that of acidic-basic repeat protein to the surface of

    Plasmodium falciparum merozoites. Mol Microbiol 2002; 43: 14011411.

    127McColl DJ, Anders RF. Conservation of structural motifs and antigenic diversity in the

    Plasmodium falciparum merozoite surface protein-3 (MSP-3). Mol Biochem Parasitol

    1997; 90: 2131.

    128McColl DJ, Silva A, Foley M, Kun JF, Favaloro JM, Thompson JK et al. Molecular

    variation in a novel polymorphic antigen associated with Plasmodium falciparum

    merozoites. Mol Biochem Parasitol 1994; 68: 5367.

    129Escalante AA, Lal AA, Ayala FJ. Genetic polymorphism and natural selection in the

    malaria parasite Plasmodium falciparum. Genetics 1998; 149: 189202.

    Blood-stage vaccines against malariaJS Richards and JG Beeson

    387

    Immunology and Cell Biology

  • 8/2/2019 Review on Blood Stage Vaccine Against Malarai

    12/14

    130Soe S, Theisen M, Roussilhon C, Aye KS, Druilhe P. Association between protection

    against clinical malaria and antibodies to merozoite surface antigens in an area of

    hyperendemicity in Myanmar: complementarity between responses to merozoite sur-

    face protein3 and the 220-kilodalton glutamate-rich protein. InfectImmun2004; 72:

    247252.

    131Druilhe P, Spertini F, Soesoe D, Corradin G, Mejia P, Singh S et al. A malaria vaccine

    that elicits in humans antibodies able to kill Plasmodium falciparum. PLoS Med

    2005; 2: e344.

    132Audran R, Cachat M, Lurati F, Soe S, LeroyO, Corradin G et al. Phase I malaria vaccine

    trial with a long synthetic peptide derived from the merozoite surface protein 3antigen. Infect Immun 2005; 73: 80178026.

    133Oeuvray C, Bouharoun-Tayoun H, Gras-Masse H, Bottius E, Kaidoh T, Aikawa M et al.

    Merozoite surface protein-3: a malaria protein inducing antibodies that promote

    Plasmodium falciparum killing by cooperation with blood monocytes. Blood 1994;

    84: 15941602.

    134Singh S, Soe S, Mejia JP, Roussilhon C, Theisen M, Corradin G et al. Identification of a

    conserved region of Plasmodium falciparum MSP3 targeted by biologically active

    antibodies to improve vaccine design. J Infect Dis 2004; 190: 10101018.

    135Sirima SB, Nebie I, Ouedraogo A, Tiono AB, Konate AT, Gansane A et al. Safety and

    immunogenicity of the Plasmodium falciparum merozoite surface protein-3 long

    synthetic peptide (MSP3-LSP) malaria vaccine in healthy, semi-immune adult

    males in Burkina Faso, West Africa. Vaccine 2007; 25: 27232732.

    136Theisen M, Soe S, Brunstedt K, Follmann F, Bredmose L, Israelsen H et al.

    A Plasmodium falciparum GLURP-MSP3 chimeric protein; expression in Lactococcus

    lactis, immunogenicity and induction of biologically active antibodies. Vaccine 2004;

    22: 11881198.

    137Remarque EJ, Faber BW, Kocken CH, Thomas AW. Apical membrane antigen 1: a

    malaria vaccine candidate in review. Trends Parasitol 2008; 24: 7484.138Pizarro JC, Vulliez-Le Normand B, Chesne-Seck ML, Collins CR, Withers-Martinez C,

    Hackett F et al. Crystal structure of the malaria vaccine candidate apical membrane

    antigen 1. Science 2005; 308: 408411.

    139Alexander DL, Arastu-Kapur S, Dubremetz JF, Boothroyd JC. Plasmodium falciparum

    AMA1 binds a rhoptry neck protein homologous to TgRON4, a component of the

    moving junction in Toxoplasma gondii. Eukaryot cell 2006; 5: 11691173.

    140Marshall VM, Zhang L, Anders RF, Coppel RL. Diversity of the vaccine candidate AMA-

    1 of Plasmodium falciparum. Mol Biochem Parasitol 1996; 77: 109113.

    141Kennedy MC, Wang J, Zhang Y, Miles AP, Chitsaz F, Saul A et al. In vitro studies with

    recombinant Plasmodium falciparum apical membrane antigen 1 (AMA1): production

    and activity of an AMA1 vaccine and generation of a multiallelic response. Infect

    Immun 2002; 70: 69486960.

    142Escalante AA, Grebert HM, Chaiyaroj SC, Magris M, Biswas S, Nahlen BL et al.

    Polymorphism in the gene encoding the apical membrane antigen-1 (AMA-1) of

    Plasmodium falciparum. X. Asembo Bay Cohort Project. Mol Biochem Parasitol

    2001; 113: 279287.

    143Cortes A, Mellombo M, Mueller I, Benet A, Reeder JC, Anders RF. Geographical

    structure of diversity and differences between symptomatic and asymptomatic infec-

    tions for Plasmodium falciparum vaccine candidate AMA1. Infect Immun 2003; 71:

    14161426.

    144Polley SD, Mwangi T, Kocken CH, Tho