1,833
Views
56
CrossRef citations to date
0
Altmetric
Review

Can growth inhibition assays (GIA) predict blood-stage malaria vaccine efficacy?

, &
Pages 706-714 | Received 10 Nov 2011, Accepted 15 Feb 2012, Published online: 20 Apr 2012

Abstract

An effective vaccine against P. falciparum malaria remains a global health priority. Blood-stage vaccines are an important component of this effort, with some indications of recent progress. However only a fraction of potential blood-stage antigens have been tested, highlighting a critical need for efficient down-selection strategies. Functional in vitro assays such as the growth/invasion inhibition assays (GIA) are widely used, but it is unclear whether GIA activity correlates with protection or predicts vaccine efficacy. While preliminary data in controlled human malaria infection (CHMI) studies indicate a possible association between in vitro and in vivo parasite growth rates, there have been conflicting results of immunoepidemiology studies, where associations with exposure rather than protection have been observed. In addition, GIA-interfering antibodies in vaccinated individuals from endemic regions may limit assay sensitivity in heavily malaria-exposed populations. More work is needed to establish the utility of GIA for blood-stage vaccine development.

Introduction

P.falciparum malaria is the pre-eminent tropical parasitic infection, causing approximately 300 million infections and around 800,000 deaths per year (World Malaria Report, WHO, 2010). Effective control strategies such as insecticide-treated bed-nets (ITNs), and artemesinin-combination therapies (ACT) have contributed to considerable and impressive reductions in malaria incidence in some countries,Citation1 prompting renewed calls for malaria eradication.Citation2 Yet the evolution of parasite resistance to drugsCitation3 and vector resistance to insecticidesCitation4 continues to challenge control efforts, and the development of an effective malaria vaccine is a global public health priority.Citation5,Citation6

While a partially effective vaccine is aiming for licensure in 2015,Citation7 a highly effective vaccine against P. falciparum malaria remains elusive. There are many challenges to overcome,Citation8,Citation9 including considerable parasite genetic diversity, a lack of suitable animal models, and an incomplete understanding of the effector mechanisms that determine natural immunity in humans.Citation10 A variety of vaccine strategies targeting all stages of the parasite lifecycle have been pursued, including recombinant protein-in-adjuvant preparations,Citation11 replication-deficient viral vectors encoding malaria antigensCitation12 and attenuated whole parasites.Citation13 Fewer than 0.5% of malaria proteins have been explored as potential candidate vaccine antigens,Citation9 but the presence of naturally acquired immunity (in contrast to other important pathogens such as HIV), together with evidence of experimentally-induced immunity in humans,Citation14 offers the promise that better understanding of protective immune effector mechanisms might accelerate the vaccine development process.Citation9

With so many potential vaccine candidates and platforms, robust down-selection strategies are required for candidate antigens. In the case of vaccines to the asexual blood-stage, the most commonly employed strategy for candidate antigen down-selection has been the detection of antibody with in vitro activity in growth inhibition assays (GIA).Citation15,Citation16 In primate challenge models induced antibodies with high levels of GIA activity against blood-stage antigens such as apical membrane antigen-1 (AMA-1) and merozoite surface protein-1 (MSP-1) have been associated with protection against lethal challenge.Citation17,Citation18 But can the in vitro GIA activity of induced antibodies predict blood-stage vaccine efficacy in humans? Here we attempt to address this question using data from published immunoepidemiological, CHMI, and field efficacy studies.

Parasite growth and invasion

The malaria lifecycle is complex, involving several stages. Infected Anopheles mosquitoes inject sporozoites of P. falciparum present in their salivary glands when taking a blood meal. These sporozoites migrate to and invade liver cells, setting up the liver (or pre-erythrocytic) stage of infection. After around seven days each infected liver cell releases approximately 30,000 merozoites into the bloodstream. These merozoites invade and replicate asexually within red blood cells (erythrocytes), leading to an exponential increase in parasites in the blood (parasitemia). This is the blood-stage of infection - the only stage at which clinical disease occurs. Later in blood-stage replication a few parasites develop into male and female gametocytes, and these in turn may be taken up by feeding mosquitoes, leading to sexual reproduction in the mosquito that produces a new generation of sporozoites.

Invasion of erythrocytes by merozoites is rapidCitation19 and involves three main phases: 1) attachment, 2) apical re-orientation and 3) invasion.Citation20 Various merozoite antigens are involved in these processes; such as the merozoite surface proteins (MSPs, particularly MSP-1) in attachment; the apical membrane antigen 1 (AMA-1) in re-orientation; and two families termed the erythrocyte binding antigens (EBAs) and the Rh proteins in invasion.Citation21 Some of these proteins are leading BS vaccine targets (e.g., AMA-1 and MSP-1), although many more are untested.Citation15

Blood-stage immunity

Although the precise immunological mechanisms underpinning malaria immunity are unresolved, its natural history is well established.Citation22 Immunity develops over time with repeated exposure to the malaria parasite (providing death does not occur), first to severe disease in infants, then to clinical disease in children and young adults. Immunity is rarely ‘sterilizing’ (i.e., asymptomatic parasitemia is often observed in adults), suggesting that naturally acquired immunity occurs mainly at the blood-stage.Citation10 Attempting to accelerate and improve upon this naturally acquired immunity is the major goal of blood-stage vaccines.Citation15,Citation16 Malaria immunity is maintained by continued exposure to parasite antigens, and the ideal BS vaccine will be similarly boosted by, but will not require, natural exposure.Citation16 Studies of experimentally induced human malaria (used as a therapy for neurosyphilis in the pre-antibiotic era) have demonstrated that the immunity that develops is both strain and species specific.Citation23 Even in a single host, substantial variation of surface antigen expression may enable evasion of host immunity.Citation24

A few BS vaccine candidates have demonstrated limited evidence of strain-specific efficacy,Citation25-Citation28 however the most effective malaria vaccine to date has been the protein-adjuvant anti-sporozoite (pre-erythrocytic stage) vaccine RTS,S, which is the only candidate malaria vaccine to have progressed to phase III efficacy trials.Citation7 Yet RTS,S is only partially effective (30–50% efficacy against clinical malaria in phase IIb studies,Citation29-Citation32 50% in an interim analysis of the phase III trialCitation7) and recent data suggest that by reducing the frequency of exposure to blood stage parasites, RTS,S may actually increase the probability of clinical malaria (vs. asymptomatic parasitemia) in those who are subsequently infected.Citation33 This increased probability of clinical disease is outweighed by the vaccine-induced reductions in exposure that reduce the incidence of clinical malaria overall.Citation33 Nevertheless, the impact of changing transmission patterns on disease severity is uncertain,Citation34 and the desirability of a complementary BS vaccine to mop-up leaky pre-erythrocytic immunity, and potentially deal with epidemic transmission patterns, is generally acknowledged.Citation8,Citation9,Citation15

Antibodies are central to BS immunity. The role of antibodies was first identified by passive transfer experiments in rhesus macaques in 1937.Citation35 Seminal experiments by Cohen and colleagues established the protective role of antibody against clinical disease by transferring purified IgG from semi-immune adults to young children with acute malaria.Citation36 Additional immune responses may also be involved in BS immunity.Citation10 Th1 CD4+ effector memory responses to BS parasitesCitation37,Citation38 are associated with long-term protection after repeat sporozoite exposure under drug treatment, although the precise mechanisms of this protection are unclear. A previous study had also proposed T-cell mediated protection resulted from repeated exposure to ultra-low dose BS parasite inoculation followed by drug treatment,Citation39 but these protection data were confounded by residual antimalarial activity at the final challenge.Citation40 In vitro growth inhibition assay (GIA)

The activity of immune seraCitation41 or purified immunoglobulinCitation42-Citation44 in parasite growth inhibition assays (GIA) has been recognized for many years. Cohen and colleagues published the first report of the in vitro growth inhibitory activity of P. knowlesi antibody in 1969.Citation45 Antibodies with GIA activity against P. falciparum can be effectively induced by immunization in humans using protein-in-adjuvant and vectored subunit vaccine approaches (reviewed in refs.Citation15,Citation16), although the magnitude of vaccine-induced GIA responses are dependent on the population group immunizedCitation46 and the vaccine antigen.Citation47 For the best-studied BS antigens AMA-1 and MSP-1, significant antigen-specific variation is observed in the GIA response, with AMA1 vaccines providing greater GIA activity than MSP-142 based vaccines.Citation47 The vaccine-induced GIA response is also highly parasite strain-dependent, particularly for AMA-1,Citation48 which mirrors the strain-specificity of the AMA-1 vaccine-induced protective immunity in vivo.Citation25 In addition, vaccine-induced increases in GIA responses in adults in endemic regions appear less pronounced.Citation46

The development of the growth inhibition assay (GIA) is indebted to the in vitro P. falciparum culture methods developed by Tragar and Jensen.Citation49 Various GIA techniques have been used to qualitatively assess antibody-mediated effects on parasite growth and/or invasion in vitro, from labor-intensive microscopyCitation42-Citation44,Citation50 and high-throughput biochemical assays of parasite proliferation and viability,Citation48,Citation51,Citation52 to the use of chimeric murine parasitesCitation53 and GFP-expressing parasites in flow cytometric assays.Citation54,Citation55 The recent development of isolation and culture methods for P. falciparum merozoites has also allowed the development of a highly sensitive merozoite invasion assay.Citation19

It is important to recognize that these various functional assays measure distinct components of the antibody response.Citation52 Most microscopy and flow cytometry assays measure growth as a function of invasion capacity (in other words they are invasion inhibition assays - IIA - but confusingly this term is often used synonymously with GIA). These assays do not measure parasite viability, and some cannot distinguish dead from live parasites. On the other hand, viability assays (e.g., biochemical measurement of parasite lactate dehydrogenase (pLDH)) measure intracellular growth characteristics as well as the invasion capacity of the parasite.Citation52

These different methodologies produce functional readouts that do appear to correlate,Citation52,Citation56 although head-to-head comparisons of different methods are infrequently employed in clinical trials.Citation27,Citation56 Standardization of the functional assay chosen is extremely important, since the read-outs can be influenced by methodological factors such as the use of whole sera or purified immunoglobulin, methods of immunoglobulin purification, the use of dialysis to remove potential contaminating antimalarial drugs, the number of growth cycles, and heat-inactivation of complement.Citation52,Citation55,Citation57-Citation61 Furthermore, GIA activity is rarely normalized to the concentration of purified immunoglobulin or the sera dilution, and this creates further heterogeneity which limits comparability between studies: for example, AMA-1 vaccine-induced GIA was quoted as 63% in one study using 10mg/mL of purified IgG,Citation62 and 70–77% in another using 4mg/mL of purified IgG.Citation27 Therefore headline GIA activity may be misleading without appreciating the antibody concentration tested, yet this is rarely reported prominently. Although a somewhat more labor intensive approach, titration of sera/immunoglobulin and presentation of the 50% inhibitory concentration (IC50)Citation63 would greatly improve inter-study comparability. Further moves to standardize the GIA have been taken, including the establishment of the PATH-Malaria Vaccine Initiative funded GIA reference center at the NIH in 2004, yet various iterations of the GIA continue to be employed in clinical studies. For example, in a recent clinical vaccine and challenge study, there was a 2–3 fold difference in the magnitude of GIA activity when the same samples were tested by different methodologies, although the outputs were well correlated.Citation27 We suggest that vaccine developers reporting GIA should endeavor to report in parallel results on their samples from the GIA reference center, to improve comparability of outcomes for the field. A potential limiting factor to its wide application in field vaccine studies is that GIA often requires a prohibitive quantity of sera considering the bleed volumes permitted in most infant studies in endemic regions, and moves to down-scale assays are welcomed.Citation51

In addition to growth or invasion inhibition assays, promising assays to measure additional Fc receptor (FcR) dependent functional antibody activities have been developed. A limited number of field studies have identified that clinical protection or parasitemia correlated with antibody parasite inhibitor activity in the presence of monocytesCitation64,Citation65 or neutrophils,Citation66 whereas antibody alone did not. Although highly promising as a functional readout,Citation67 such assays are technically challengingCitation66,Citation68 and have not been widely replicated by other groups, therefore their utility to the broader vaccine development field has been limited to date. Another approach takes advantage of transgenic murine parasite lines expressing P. falciparum antigens and transgenic mice expressing human FcRs to examine FcR-mediated effector functions in detail.Citation69 Together these functional assays are likely to contribute significantly to future vaccine development efforts.Citation16 However, we focus here on the most widely used and repeatable functional assays in blood-stage vaccine development, the growth/invasion inhibition assays (GIA).

Acquisition of inhibitory antibodies

Before examining the utility of GIA in BS vaccine development, it is important to understand more about the kinetics of inhibitory antibody development. Although the well-standardized controlled human malaria infection (CHMI) models in malaria-naïve volunteers provide an appropriate framework to explore such questions, no CHMI studies have reported GIA post-challenge, meaning it is unclear whether a single episode of malaria infection during CHMI results in detectable functional antibody in GIA.Citation26,Citation27,Citation38,Citation39,Citation62,Citation70-Citation74 In macaques challenged with P. knowlesi, substantial increases in GIA activity have been noted post-challenge.Citation75

Uncertainty also surrounds the acquisition of inhibitory antibodies after naturally acquired infection. Eisen and colleagues identified that invasion inhibitory antibodies developed rapidly in non-immune travelers who had recently acquired P. falciparum,Citation76 yet in transmigrants the development of MSP-119 inhibitory antibodies required two or more infections.Citation77 Incremental boosting of the memory B cell compartment by repeated infection has been observed in individuals in malaria transmission settings,Citation78 but GIA activity appears to be acquired at an early age in high transmission settings,Citation79 does not appear to be boosted by repeated infections and in fact often decreases with age (see ). Using sera from semi-immune Sudanese adults, inhibition of intraerythrocytic parasite growth, but not invasion activity, was temporally associated with transmission.Citation50 In addition, an increase in autologous parasite inhibition activity was observed over a two-week period in 57% of infected individuals in Burkina Faso.Citation80 A similar kinetic of increased invasion inhibition activity was observed one month after infection in Vietnam, but by contrast, MSP-119 specific invasion activity fell rapidly, and the end of the transmission season was also associated with a fall in MSP-119 invasion inhibition activity.Citation81 Overall the kinetics of acquisition of GIA activity suggest an association with parasite exposure, although this does not necessarily imply a protective role.

Table 1. GIA/IIA field studies

Immunoepidemiology studies

Many prospective studies have examined the association between protection against malaria infection or clinical disease and antibodies to merozoite surface proteins, and this topic has been subject to a recent comprehensive systematic review.Citation82 Despite considerable heterogeneity in study design and clinical endpoints there is evidence for a positive relationship between the levels of antibodies (measured by ELISA) to several leading blood-stage malaria vaccine candidate antigens (including AMA-1, MSP-119, MSP-3 and glutamate-rich protein (GLURP)) and protection in malaria-exposed populations.Citation82

Simply measuring the magnitude of immune responses yields no information on the qualitative nature of the induced antibodies. Unfortunately, far fewer studies have focused on functional characteristics of the antibody response and whether these correlate with protection.Citation33,Citation56,Citation68,Citation79-Citation81,Citation83-Citation88 Those studies published to date are summarized in .

It can be seen from that the conclusions of these studies in terms of the role of GIA activity in protection are highly conflicting. While some studies suggest a protective role for GIA activity,Citation68,Citation83,Citation86,Citation88 most do not.Citation33,Citation56,Citation79,Citation81,Citation84,Citation85,Citation87 These conflicting data have several possible origins. Cross-sectional studies are less informative than prospective studies. In addition, the choice of trial endpoints is influential. A problem common to all field studies is the difficulty in differentiating between the absence of malaria exposure and “protection.”Citation89 Whether this has a significant impact on the data are dependent on the intensity of transmission and other important confounding factors such as the use of ITNs.Citation90 Misclassification of unexposed individuals as “protected” will significantly bias interpretation of the immune response measured.Citation33,Citation90 This is a particular problem for studies using GIA since the kinetics of acquisition of growth inhibitory antibodies appears to be linked to exposure (see ). The use of more clinically relevant endpoints such as protection against clinical disease (defined as parasitemia with clinical symptoms vs. asymptomatic parasitemia) may be more informative. However, the definition of clinical disease is also subject to inter-study variability and a lack of agreed standardization.Citation82

Additional heterogeneity is introduced by the use of different assays, which as discussed above, measure distinct aspects of the functional antibody response; however Dent and colleagues used three different GIA methodologies and found similar associations.Citation68 Moreover, even with very similar assays, there can be conflicting data from different populations,Citation81,Citation86 suggesting the presence of additional confounding effects of transmission intensity and/or host genetic variation.

There is also significant correlation between age and protection against malaria,Citation91 and to account for this prospective studies should be performed in well-defined age groups and analysis corrected for age.Citation68,Citation88 Interestingly, it appears from most prospective studies that GIA activity is present in young children and then reduces with increasing age,Citation33,Citation68,Citation79,Citation84,Citation87,Citation88 in contrast to the age-related progression of clinical immunity. This could suggest that GIA activity is unrelated to clinical disease immunity, although some authors have suggested a possible contribution to early protection against severe disease.Citation79,Citation88 This seems unlikely, since severe disease immunity is generally acquired after one or two infections and asymptomatic high-density parasitemia is very common in young children in endemic areas. On the other hand, malaria antibodies that interfere with vaccine-induced GIA activity are found in adultsCitation58 and children in endemic areas,Citation92 and are not found in malaria-naïve individuals.Citation58 This suggests that GIA “interfering” antibodies may also develop with malaria exposure. The acquisition of these interfering antibodies could partially explain the lack of a positive association between GIA and age or protection. The implications of such interfering antibodies are not yet clear, but these data imply that the sensitivity of GIA may be reduced significantly in certain groups,Citation46 and raise concerns about the use of GIA as a marker of BS vaccine responses in malaria-experienced populations.

These observations highlight the importance of the fine specificity of the antibody response. For the leading vaccine candidates (MSP-1 and AMA-1), evidence suggests that antibodies to conformational epitopes are critical in mediating GIA activity (reviewed in refs.Citation11,Citation93). Monoclonal antibodies (mAb) with GIA activity against AMA-1 and MSP-1 have been shown in structural studies to inhibit proteolytic processing of the precursor proteins (e.g., 88-kDa AMA1/MSP142), a step required for merozoite invasion, through binding to discontinuous (conformational) epitopes.Citation11,Citation93 Individuals with MSP119 antibodies displaying cross-competition against the inhibitory monoclonal antibody (mAb) 12.10 had significantly lower parasite densities in cross-sectional studies.Citation85,Citation94 However, the “interfering” antibodies described above can, through steric inhibition, block the binding of growth/invasion inhibitory antibodies and therefore interfere with this functional activity. Binding affinity, an additional aspect of the fine specificity of the antibody response, may interestingly influence the functional phenotype of the antibody, with recent suggestions that higher affinity antibodies may be associated with inhibitory activity, while lower affinity antibodies generate ADCI.Citation11 Finally, as mentioned briefly before, the specificity of protective antibody can be influenced by the degree of parasite polymorphism. In pre-clinical studies significant diversity of the target epitopes in AMA-1 and MSP-1 attenuates the response against heterologous (non-vaccine) strains.Citation95 The clinical importance of this target antigen polymorphism was recently illustrated in a phase IIb study of an AMA-1 vaccine based on recombinant 3D7 strain AMA-1, where 3D7 strain-specific protective effects were observed in immunized individuals, but no protection was conferred against heterologous (non-vaccine) strains.Citation25 A similar pattern was observed in the phase IIb field study of the Combination B vaccine.Citation28 Therefore parasite antigen polymorphism may significantly influence the association (or lack thereof) between GIA activity and clinical protection in observational studies. Moreover, it should be remembered that GIA is often tested against well-characterized laboratory strains in vitro that may not reflect the parasite strains circulating in the community, further reducing the likelihood of a relationship (if present) being observed.

With such a complex pathogen as P. falciparum it seems unlikely that a single immune mediator will ever account entirely for immunological protection.Citation9 Assessment of a greater breadth of functional immune responses, combined with integrative systems biology approaches, will likely be required to achieve more substantial insights into the major components of protective immunity. This also applies to CHMI studies.

Controlled human malaria infections (CHMI)

CHMI by either sporozoite or blood-stage inoculation of healthy malaria-naïve volunteers can accelerate candidate vaccine development by providing rapid and robust efficacy readouts.Citation5,Citation96 These studies are performed in a small number of centers worldwide.Citation97 Whether efficacy in CHMI predicts field efficacy in target populations remains an open question, since so few candidates have demonstrated convincing efficacy in CHMIs, but the indications are that CHMI can accurately down-select potential candidates, since very few phase IIb studies have detected efficacy in the absence of an efficacy signal in a phase IIa CHMICitation5 (the only possible exception is the Combination B vaccineCitation28,Citation71). Pre-erythrocytic vaccine efficacy readouts are unambiguous (i.e., sterilizing protection or delays to patency), although mathematical modeling of parasitemia (measured by highly-sensitive quantitative polymerase chain reaction assays) also detects important reductions in liver-to-blood inocula.Citation98 Blood-stage vaccine CHMI efficacy readouts include delays to microscopic patency by blood-film microscopy, and/or reductions of the in vivo parasite multiplication rate.Citation70,Citation99,Citation100 Relatively few blood-stage vaccine candidates have been tested by CHMICitation96 (see ), although more studies have been performed recentlyCitation27,Citation62 (and clinicaltrials.gov/NCT01142765) as the utility of the CHMI model is increasingly recognized.Citation97 Until recently the BS vaccine goal-posts in CHMI had not been experimentally defined. PMR in semi-immune individuals appears to be considerably lower than in malaria-naïve individuals,Citation101 and is also considerably lower than has been achieved in any vaccine CMHI trial to dateCitation26,Citation27,Citation62,Citation71 implying that the first generation of BS vaccines was not sufficiently immunogenic to impact on in vivo PMR. PMR by CHMI is a more clinically relevant study endpoint than in vitro GIACitation70 and is most appropriate to determine which candidates to take to the field,Citation97 but CHMI studies may be impractical in some settings due to logistics, regulatory hurdles and prohibitive costs.Citation15 In addition, by CHMI no consistent association has emerged between significant reductions in PMR and clinical indicators of BS protection such as delays in pre-patent period.Citation26,Citation27,Citation62,Citation71 Robust CHMI models (e.g., those involving twice-daily qPCR monitoring of blood-stage parasitemia) should be better capable of predicting field efficacy for BS vaccines as more immunogenic BS vaccine candidates are developed.

Table 2. Published blood-stage vaccine controlled human malaria infection (CHMI) studies

If GIA is an important mechanism through which antibodies mediate BS protection in vivo, one might expect an association between in vitro GIA and in vivo PMR. This hypothesis has only been tested in a single CHMI study,Citation62 in which a significant positive correlation was observed in a small group of AMA-1 vaccinated malaria-naïve volunteers despite no effects on pre-patent period or overall PMR in the vaccine group. This interesting finding needs to be replicated in larger cohorts, but does indicate that in vitro GIA may be a useful surrogate for in vivo PMR in vaccinated malaria-naïve individuals. However the association does not imply causality. Additional immune effector mechanisms have been associated with PMR in CHMI including cytokines and regulatory T cells.Citation73 Unfortunately no other BS vaccine CHMI studies have directly examined the relationship between GIA and PMR. Spring and colleagues modeled PMR by a group method that does not yield values for individual volunteers, meaning correlations could not be assessed, although there was a trend to reduced PMR in vaccine groups with GIA activity.Citation27 By contrast, in another CHMI study of an AMA-1-containing multistage virosomal vaccine, there was no measurable activity by GIA, despite indications of blood-stage efficacy in one immunised individual,Citation26 suggesting that either the blood-stage efficacy observed was mediated by a separate immune mechanisms than antibody inhibitory activity, or that the GIA failed to predict potential efficacy. In a separate BS vaccine study, GIA was not performed, and the vaccine had no observed impact on PMR,Citation71 despite later indications of possible limited strain-specific effects in a phase IIb study.Citation28 Overall, there is currently insufficient data from CHMI to conclusively determine the relationship between in vitro GIA and in vivo PMR, or the predictive power of GIA, although a promising indication warrants further investigation.

Going forward, CHMI may have significant potential in proof of concept studies to validate GIA and other functional assays, in contrast to field studies where problems of unexposed individuals and interfering antibodies (discussed above) confound the assessment of protection and its relationship to immune markers. Moreover, CHMIs in well-defined semi-immune adult populationsCitation97 may also shed light on the various contributions of functional antibodies (and other immune effector mechanisms) to BS immunity.Citation96

Field efficacy studies

In part due to prohibitive blood-draw limits during phase IIb studies in young children and infants, GIA has not generally been performed in field efficacy studies of BS vaccine candidates to date,Citation25,Citation28,Citation102,Citation103 meaning that prospective correlations with efficacy cannot be assessed. In one pediatric phase IIb trial of an AMA-1 vaccine with Alhydrogel,Citation103 a modest vaccine-induced increase in GIA activity was observed in children without pre-existing GIA activity (< 20%),Citation46 but GIA did not correlate with parasitemia,Citation46 and there was no field efficacy with this vaccine.Citation103 In order to understand more about the value of functional antibody assays in predicting efficacy (or a lack thereof), approaches to facilitate GIA in field efficacy studies should be explored.Citation51,Citation54 Feedback of such data will inform future vaccine design.Citation97

Closing remarks

Although BS vaccine development has been largely disappointing to date, there are several reasons for cautious optimism. Immunogenicity of some vaccine platforms, particularly viral-vectored vaccines and prime-boost regimens, is improving significantly,Citation12,Citation104 and many potential candidate antigens remain to be tested.Citation9 For example, a recent promising study of vectored vaccines against the conserved candidate antigen Rh5Citation105 suggests that many untested antigens could be effective vaccine targets. Indications of strain-specific efficacy with some monovalent AMA-1 vaccines are also promising,Citation25 but more work is required to understand how best to down-select the many potential BS vaccine candidates in the pipeline. In vitro functional antibody assays may be important to this effort due to their scalable nature, but it is critical to establish whether the widely used assays such as GIA are of value in predicting vaccine efficacy. To this end, GIA should be prospectively validated in standardized CHMI models to follow up on promising indications of a correlation with in vivo parasite multiplication rate.Citation62 The kinetics of GIA activity could be better defined by prospective studies in endemic settings by controlling for exposure, and by studying kinetics in CHMI. Finally, field vaccine efficacy studies should endeavor to report GIA data to improve our understanding of the important relationship between efficacy and in vitro GIA.

Abbreviations:
GIA=

growth inhibition assay

IIA=

invasion inhibition assay

PMR=

parasite multiplication rate

BS=

blood-stage

CHMI=

controlled human malaria infection

ITN=

insecticide treated bed nets

ACT=

artemesinin-combination therapy

AMA-1=

apical membrane antigen 1

MSP-1=

merozoite surface protein 1

HIV=

human immunodeficiency virus

WHO=

World Health Organization

EBA=

erythrocyte binding antigen

glutamate-rich protein=

GLURP

green fluorescent protein=

GFP

enzyme-linked immunosorbent assay=

ELISA

NYVAC=

Attenuated Copenhagen strain Vaccinia virus

PEV3A=

virosome formulation of apical membrane antigen-1 and circumsporozoite protein

FP9=

Fowlpox-9

MVA=

Modified Vaccinia virus Ankara strain

AS=

Adjuvant System

AMA-1/C1=

Apical membrane antigen-1/combination 1

WRAIR=

Walter Read Army Institute for Research, USA

QIMR=

Queensland Institute for Medical Research, Australia

CCVTM=

Centre for Clinical Vaccinology and Tropical Medicine, UK

CHMI=

Controlled human malaria infection

Spz=

Sporozoite challenge

BSP=

Blood-stage parasite challenge

PMR=

Parasite multiplication rate (in vivo)

ND=

Not done

NP=

Not presented

qPCR=

Quantitative polymerase chain reaction

PcMSP-119/PyMSP-119=

chimeric P. falciparum expressing P. chabaudi or P. yoelii merozoite surface protein-119

pLDH=

parasite lactate dehydrogenase activity

3H=

Tritium radiometric viability assay

Acknowledgments

We thank Dr Simon J. Draper for helpful comments on the manuscript. This research was supported in part by the Intramural Research Program of the NIH, NIAID (RDE), the Wellcome Trust (CJAD and AVSH) and the UK NIHR (AVSH).

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • O’Meara WP, Bejon P, Mwangi TW, Okiro EA, Peshu N, Snow RW, et al. Effect of a fall in malaria transmission on morbidity and mortality in Kilifi, Kenya. Lancet 2008; 372:1555 - 62; http://dx.doi.org/10.1016/S0140-6736(08)61655-4; PMID: 18984188
  • Roberts L, Enserink M. Malaria. Did they really say ... eradication?. Science 2007; 318:1544 - 5; http://dx.doi.org/10.1126/science.318.5856.1544; PMID: 18063766
  • Dondorp AM, Nosten F, Yi P, Das D, Phyo AP, Tarning J, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 2009; 361:455 - 67; http://dx.doi.org/10.1056/NEJMoa0808859; PMID: 19641202
  • Trape JF, Tall A, Diagne N, Ndiath O, Ly AB, Faye J, et al. Malaria morbidity and pyrethroid resistance after the introduction of insecticide-treated bednets and artemisinin-based combination therapies: a longitudinal study. Lancet Infect Dis 2011; 11:925 - 32; http://dx.doi.org/10.1016/S1473-3099(11)70194-3; PMID: 21856232
  • Sauerwein RW, Roestenberg M, Moorthy VS. Experimental human challenge infections can accelerate clinical malaria vaccine development. Nat Rev Immunol 2011; 11:57 - 64; http://dx.doi.org/10.1038/nri2902; PMID: 21179119
  • Plowe CV, Alonso P, Hoffman SL. The potential role of vaccines in the elimination of falciparum malaria and the eventual eradication of malaria. J Infect Dis 2009; 200:1646 - 9; http://dx.doi.org/10.1086/646613; PMID: 19877844
  • First Results of Phase 3 Trial of RTS,S/AS01 Malaria Vaccine in African Children. N Engl J Med 2011;
  • Hill AV. Vaccines against malaria. Philos Trans R Soc Lond B Biol Sci 2011; 366:2806 - 14; http://dx.doi.org/10.1098/rstb.2011.0091; PMID: 21893544
  • Crompton PD, Pierce SK, Miller LH. Advances and challenges in malaria vaccine development. J Clin Invest 2010; 120:4168 - 78; http://dx.doi.org/10.1172/JCI44423; PMID: 21123952
  • Langhorne J, Ndungu FM, Sponaas AM, Marsh K. Immunity to malaria: more questions than answers. Nat Immunol 2008; 9:725 - 32; http://dx.doi.org/10.1038/ni.f.205; PMID: 18563083
  • Anders RF, Adda CG, Foley M, Norton RS. Recombinant protein vaccines against the asexual blood stages of Plasmodium falciparum. Hum Vaccin 2010; 6:39 - 53; http://dx.doi.org/10.4161/hv.6.1.10712; PMID: 20061790
  • Hill AV, Reyes-Sandoval A, O’Hara G, Ewer K, Lawrie A, Goodman A, et al. Prime-boost vectored malaria vaccines: progress and prospects. Hum Vaccin 2010; 6:78 - 83; http://dx.doi.org/10.4161/hv.6.1.10116; PMID: 20061802
  • Hoffman SL, Billingsley PF, James E, Richman A, Loyevsky M, Li T, et al. Development of a metabolically active, non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria. Hum Vaccin 2010; 6:97 - 106; http://dx.doi.org/10.4161/hv.6.1.10396; PMID: 19946222
  • Clyde DF, Most H, McCarthy VC, Vanderberg JP. Immunization of man against sporozite-induced falciparum malaria. Am J Med Sci 1973; 266:169 - 77; http://dx.doi.org/10.1097/00000441-197309000-00002; PMID: 4583408
  • Ellis RD, Sagara I, Doumbo O, Wu Y. Blood stage vaccines for Plasmodium falciparum: current status and the way forward. Hum Vaccin 2010; 6:627 - 34; http://dx.doi.org/10.4161/hv.6.8.11446; PMID: 20519960
  • Goodman AL, Draper SJ. Blood-stage malaria vaccines - recent progress and future challenges. Ann Trop Med Parasitol 2010; 104:189 - 211; http://dx.doi.org/10.1179/136485910X12647085215534; PMID: 20507694
  • Dutta S, Sullivan JS, Grady KK, Haynes JD, Komisar J, Batchelor AH, et al. High antibody titer against apical membrane antigen-1 is required to protect against malaria in the Aotus model. PLoS One 2009; 4:e8138; http://dx.doi.org/10.1371/journal.pone.0008138; PMID: 19997632
  • Singh S, Miura K, Zhou H, Muratova O, Keegan B, Miles A, et al. Immunity to recombinant plasmodium falciparum merozoite surface protein 1 (MSP1): protection in Aotus nancymai monkeys strongly correlates with anti-MSP1 antibody titer and in vitro parasite-inhibitory activity. Infect Immun 2006; 74:4573 - 80; http://dx.doi.org/10.1128/IAI.01679-05; PMID: 16861644
  • Boyle MJ, Wilson DW, Richards JS, Riglar DT, Tetteh KK, Conway DJ, et al. Isolation of viable Plasmodium falciparum merozoites to define erythrocyte invasion events and advance vaccine and drug development. Proc Natl Acad Sci U S A 2010; 107:14378 - 83; http://dx.doi.org/10.1073/pnas.1009198107; PMID: 20660744
  • 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; 39:91 - 6; http://dx.doi.org/10.1016/j.ijpara.2008.09.007; PMID: 18952091
  • Persson KE. Erythrocyte invasion and functionally inhibitory antibodies in Plasmodium falciparum malaria. Acta Trop 2010; 114:138 - 43; http://dx.doi.org/10.1016/j.actatropica.2009.05.017; PMID: 19481996
  • Garnham PC. Malarial immunity in Africans; effects in infancy and early childhood. Ann Trop Med Parasitol 1949; 43:47 - 61; PMID: 18121270
  • Collins WE, Jeffery GM. A retrospective examination of sporozoite- and trophozoite-induced infections with Plasmodium falciparum: development of parasitologic and clinical immunity during primary infection. Am J Trop Med Hyg 1999; 61:Suppl 4 - 19; PMID: 10432041
  • Peters J, Fowler E, Gatton M, Chen N, Saul A, Cheng Q. High diversity and rapid changeover of expressed var genes during the acute phase of Plasmodium falciparum infections in human volunteers. Proc Natl Acad Sci U S A 2002; 99:10689 - 94; http://dx.doi.org/10.1073/pnas.162349899; PMID: 12142467
  • Thera MA, Doumbo OK, Coulibaly D, Laurens MB, Ouattara A, Kone AK, et al. A field trial to assess a blood-stage malaria vaccine. N Engl J Med 2011; 365:1004 - 13; http://dx.doi.org/10.1056/NEJMoa1008115; PMID: 21916638
  • Thompson FM, Porter DW, Okitsu SL, Westerfeld N, Vogel D, Todryk S, et al. Evidence of blood stage efficacy with a virosomal malaria vaccine in a phase IIa clinical trial. PLoS One 2008; 3:e1493; http://dx.doi.org/10.1371/journal.pone.0001493; PMID: 18231580
  • Spring MD, Cummings JF, Ockenhouse CF, Dutta S, Reidler R, Angov E, et al. Phase 1/2a study of the malaria vaccine candidate apical membrane antigen-1 (AMA-1) administered in adjuvant system AS01B or AS02A. PLoS One 2009; 4:e5254; http://dx.doi.org/10.1371/journal.pone.0005254; PMID: 19390585
  • 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; 185:820 - 7; http://dx.doi.org/10.1086/339342; PMID: 11920300
  • Olotu A, Lusingu J, Leach A, Lievens M, Vekemans J, Msham S, et al. Efficacy of RTS,S/AS01E malaria vaccine and exploratory analysis on anti-circumsporozoite antibody titres and protection in children aged 5-17 months in Kenya and Tanzania: a randomised controlled trial. Lancet Infect Dis 2011; 11:102 - 9; http://dx.doi.org/10.1016/S1473-3099(10)70262-0; PMID: 21237715
  • Bejon P, Lusingu J, Olotu A, Leach A, Lievens M, Vekemans J, et al. Efficacy of RTS,S/AS01E vaccine against malaria in children 5 to 17 months of age. N Engl J Med 2008; 359:2521 - 32; http://dx.doi.org/10.1056/NEJMoa0807381; PMID: 19064627
  • Asante KP, Abdulla S, Agnandji S, Lyimo J, Vekemans J, Soulanoudjingar S, et al. Safety and efficacy of the RTS,S/AS01E candidate malaria vaccine given with expanded-programme-on-immunisation vaccines: 19 month follow-up of a randomised, open-label, phase 2 trial. Lancet Infect Dis 2011; 11:741 - 9; http://dx.doi.org/10.1016/S1473-3099(11)70100-1; PMID: 21782519
  • Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, Milman J, et al. Efficacy of the RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in young African children: randomised controlled trial. Lancet 2004; 364:1411 - 20; http://dx.doi.org/10.1016/S0140-6736(04)17223-1; PMID: 15488216
  • Bejon P, Cook J, Bergmann-Leitner E, Olotu A, Lusingu J, Mwacharo J, et al. Effect of the pre-erythrocytic candidate malaria vaccine RTS,S/AS01E on blood stage immunity in young children. J Infect Dis 2011; 204:9 - 18; http://dx.doi.org/10.1093/infdis/jir222; PMID: 21628653
  • Okiro EA, Al-Taiar A, Reyburn H, Idro R, Berkley JA, Snow RW. Age patterns of severe paediatric malaria and their relationship to Plasmodium falciparum transmission intensity. Malar J 2009; 8:4; http://dx.doi.org/10.1186/1475-2875-8-4; PMID: 19128453
  • Coggeshall LT, Kumm HW. Demonstration of Passive Immunity in Experimental Monkey Malaria. J Exp Med 1937; 66:177 - 90; http://dx.doi.org/10.1084/jem.66.2.177; PMID: 19870655
  • Cohen S, McGREGOR IA, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature 1961; 192:733 - 7; http://dx.doi.org/10.1038/192733a0; PMID: 13880318
  • Roestenberg M, Teirlinck AC, McCall MB, Teelen K, Makamdop KN, Wiersma J, et al. Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet 2011; 377:1770 - 6; http://dx.doi.org/10.1016/S0140-6736(11)60360-7; PMID: 21514658
  • Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, van Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med 2009; 361:468 - 77; http://dx.doi.org/10.1056/NEJMoa0805832; PMID: 19641203
  • 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; 360:610 - 7; http://dx.doi.org/10.1016/S0140-6736(02)09784-2; PMID: 12241933
  • Edstein MD, Kotecka BM, Anderson KL, Pombo DJ, Kyle DE, Rieckmann KH, et al. Lengthy antimalarial activity of atovaquone in human plasma following atovaquone-proguanil administration. Antimicrob Agents Chemother 2005; 49:4421 - 2; http://dx.doi.org/10.1128/AAC.49.10.4421-4422.2005; PMID: 16189139
  • Cowen NL, Clancy RL, Tulloch JL, Cripps AW, Alpers MP. Analysis of patterns of growth inhibition of P. falciparum in synchronised cultures induced by serum from children and adults from Madang, Papua New Guinea. Aust J Exp Biol Med Sci 1985; 63:513 - 20; http://dx.doi.org/10.1038/icb.1985.55; PMID: 3911931
  • Brown GV, Anders RF, Knowles G. Differential effect of immunoglobulin on the in vitro growth of several isolates of Plasmodium falciparum. Infect Immun 1983; 39:1228 - 35; PMID: 6188695
  • Brown GV, Anders RF, Mitchell GF, Heywood PF. Target antigens of purified human immunoglobulins which inhibit growth of Plasmodium falciparum in vitro. Nature 1982; 297:591 - 3; http://dx.doi.org/10.1038/297591a0; PMID: 7045680
  • Mitchell GH, Butcher GA, Voller A, Cohen S. The effect of human immune IgG on the in vitro development of Plasmodium falciparum. Parasitology 1976; 72:149 - 62; http://dx.doi.org/10.1017/S0031182000048459; PMID: 817249
  • Cohen S, Butcher GA, Crandall RB. Action of malarial antibody in vitro. Nature 1969; 223:368 - 71; http://dx.doi.org/10.1038/223368a0; PMID: 4980851
  • Miura K, Zhou H, Diouf A, Tullo G, Moretz SE, Aebig JA, et al. Immunological responses against Plasmodium falciparum Apical Membrane Antigen 1 vaccines vary depending on the population immunized. Vaccine 2011; 29:2255 - 61; http://dx.doi.org/10.1016/j.vaccine.2011.01.043; PMID: 21277408
  • Miura K, Zhou H, Diouf A, Moretz SE, Fay MP, Miller LH, et al. Anti-apical-membrane-antigen-1 antibody is more effective than anti-42-kilodalton-merozoite-surface-protein-1 antibody in inhibiting plasmodium falciparum growth, as determined by the in vitro growth inhibition assay. Clin Vaccine Immunol 2009; 16:963 - 8; http://dx.doi.org/10.1128/CVI.00042-09; PMID: 19439523
  • Malkin EM, Diemert DJ, McArthur JH, Perreault JR, Miles AP, Giersing BK, et al. Phase 1 clinical trial of apical membrane antigen 1: an asexual blood-stage vaccine for Plasmodium falciparum malaria. Infect Immun 2005; 73:3677 - 85; http://dx.doi.org/10.1128/IAI.73.6.3677-3685.2005; PMID: 15908397
  • Trager W, Jensen JB. Human malaria parasites in continuous culture. Science 1976; 193:673 - 5; http://dx.doi.org/10.1126/science.781840; PMID: 781840
  • Vande Waa JA, Jensen JB, Akood MA, Bayoumi R. Longitudinal study on the in vitro immune response to Plasmodium falciparum in Sudan. Infect Immun 1984; 45:505 - 10; PMID: 6378799
  • Bergmann-Leitner ES, Duncan EH, Burge JR, Spring M, Angov E. Miniaturization of a high-throughput pLDH-based Plasmodium falciparum growth inhibition assay for small volume samples from preclinical and clinical vaccine trials. Am J Trop Med Hyg 2008; 78:468 - 71; PMID: 18337345
  • 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; 75:437 - 42; PMID: 16968918
  • O’Donnell 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; 193:1403 - 12; http://dx.doi.org/10.1084/jem.193.12.1403; PMID: 11413195
  • Persson KE, Lee CT, Marsh K, Beeson JG. Development and optimization of high-throughput methods to measure Plasmodium falciparum-specific growth inhibitory antibodies. J Clin Microbiol 2006; 44:1665 - 73; http://dx.doi.org/10.1128/JCM.44.5.1665-1673.2006; PMID: 16672391
  • Wilson DW, Crabb BS, Beeson JG. Development of fluorescent Plasmodium falciparum for in vitro growth inhibition assays. Malar J 2010; 9:152; http://dx.doi.org/10.1186/1475-2875-9-152; PMID: 20525251
  • Perraut R, Marrama L, Diouf B, Sokhna C, Tall A, Nabeth P, et al. Antibodies to the conserved C-terminal domain of the Plasmodium falciparum merozoite surface protein 1 and to the merozoite extract and their relationship with in vitro inhibitory antibodies and protection against clinical malaria in a Senegalese village. J Infect Dis 2005; 191:264 - 71; http://dx.doi.org/10.1086/426398; PMID: 15609237
  • Bergmann-Leitner ES, Mease RM, Duncan EH, Khan F, Waitumbi J, Angov E. Evaluation of immunoglobulin purification methods and their impact on quality and yield of antigen-specific antibodies. Malar J 2008; 7:129; http://dx.doi.org/10.1186/1475-2875-7-129; PMID: 18625058
  • Miura K, Zhou H, Moretz SE, Diouf A, Thera MA, Dolo A, et al. Comparison of biological activity of human anti-apical membrane antigen-1 antibodies induced by natural infection and vaccination. J Immunol 2008; 181:8776 - 83; PMID: 19050299
  • Mlambo G, Kumar N. A modified Plasmodium falciparum growth inhibition assay (GIA) to assess activity of plasma from malaria endemic areas. Exp Parasitol 2007; 115:211 - 4; http://dx.doi.org/10.1016/j.exppara.2006.08.003; PMID: 16987515
  • Patarapotikul J, Tharavanij S, Poonthong C. Multiple strains of Plasmodium falciparum are necessary for the growth inhibition assay. Southeast Asian J Trop Med Public Health 1983; 14:149 - 53; PMID: 6356378
  • Shi YP, Udhayakumar V, Oloo AJ, Nahlen BL, Lal AA. Differential effect and interaction of monocytes, hyperimmune sera, and immunoglobulin G on the growth of asexual stage Plasmodium falciparum parasites. Am J Trop Med Hyg 1999; 60:135 - 41; PMID: 9988337
  • Duncan CJ, Sheehy SH, Ewer KJ, Douglas AD, Collins KA, Halstead FD, et al. Impact on malaria parasite multiplication rates in infected volunteers of the protein-in-adjuvant vaccine AMA1-C1/Alhydrogel+CPG 7909. PLoS One 2011; 6:e22271; http://dx.doi.org/10.1371/journal.pone.0022271; PMID: 21799809
  • Mahdi Abdel Hamid M, Remarque EJ, van Duivenvoorde LM, van der Werff N, Walraven V, Faber BW, et al. Vaccination with Plasmodium knowlesi AMA1 formulated in the novel adjuvant co-vaccine HT™ protects against blood-stage challenge in rhesus macaques. PLoS One 2011; 6:e20547; http://dx.doi.org/10.1371/journal.pone.0020547; PMID: 21655233
  • Bouharoun-Tayoun H, Attanath P, Sabchareon A, Chongsuphajaisiddhi T, Druilhe P. Antibodies that protect humans against Plasmodium falciparum blood stages do not on their own inhibit parasite growth and invasion in vitro, but act in cooperation with monocytes. J Exp Med 1990; 172:1633 - 41; http://dx.doi.org/10.1084/jem.172.6.1633; PMID: 2258697
  • Chimma P, Roussilhon C, Sratongno P, Ruangveerayuth R, Pattanapanyasat K, Pérignon JL, et al. A distinct peripheral blood monocyte phenotype is associated with parasite inhibitory activity in acute uncomplicated Plasmodium falciparum malaria. PLoS Pathog 2009; 5:e1000631; http://dx.doi.org/10.1371/journal.ppat.1000631; PMID: 19851453
  • Joos C, Marrama L, Polson HE, Corre S, Diatta AM, Diouf B, et al. Clinical protection from falciparum malaria correlates with neutrophil respiratory bursts induced by merozoites opsonized with human serum antibodies. PLoS One 2010; 5:e9871; http://dx.doi.org/10.1371/journal.pone.0009871; PMID: 20360847
  • Crabb BS, Beeson JG. Promising functional readouts of immunity in a blood-stage malaria vaccine trial. PLoS Med 2005; 2:e380; http://dx.doi.org/10.1371/journal.pmed.0020380; PMID: 16266219
  • Dent AE, Bergmann-Leitner ES, Wilson DW, Tisch DJ, Kimmel R, Vulule J, et al. Antibody-mediated growth inhibition of Plasmodium falciparum: relationship to age and protection from parasitemia in Kenyan children and adults. PLoS One 2008; 3:e3557; http://dx.doi.org/10.1371/journal.pone.0003557; PMID: 18958285
  • McIntosh RS, Shi J, Jennings RM, Chappel JC, de Koning-Ward TF, Smith T, et al. The importance of human FcgammaRI in mediating protection to malaria. PLoS Pathog 2007; 3:e72; http://dx.doi.org/10.1371/journal.ppat.0030072; PMID: 17511516
  • Cheng Q, Lawrence G, Reed C, Stowers A, Ranford-Cartwright L, Creasey A, et al. Measurement of Plasmodium falciparum growth rates in vivo: a test of malaria vaccines. Am J Trop Med Hyg 1997; 57:495 - 500; PMID: 9347970
  • Lawrence 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; 18:1925 - 31; http://dx.doi.org/10.1016/S0264-410X(99)00444-2; PMID: 10699342
  • Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, Desmons P, et al. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J Med 1997; 336:86 - 91; http://dx.doi.org/10.1056/NEJM199701093360202; PMID: 8988885
  • Walther M, Tongren JE, Andrews L, Korbel D, King E, Fletcher H, et al. Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity 2005; 23:287 - 96; http://dx.doi.org/10.1016/j.immuni.2005.08.006; PMID: 16169501
  • Webster DP, Dunachie S, Vuola JM, Berthoud T, Keating S, Laidlaw SM, et al. Enhanced T cell-mediated protection against malaria in human challenges by using the recombinant poxviruses FP9 and modified vaccinia virus Ankara. Proc Natl Acad Sci U S A 2005; 102:4836 - 41; http://dx.doi.org/10.1073/pnas.0406381102; PMID: 15781866
  • Hamid MM, Remarque EJ, El Hassan IM, Hussain AA, Narum DL, Thomas AW, et al. Malaria infection by sporozoite challenge induces high functional antibody titres against blood stage antigens after a DNA prime, poxvirus boost vaccination strategy in Rhesus macaques. Malar J 2011; 10:29; http://dx.doi.org/10.1186/1475-2875-10-29; PMID: 21303498
  • Eisen DP, Wang L, Jouin H, Murhandarwati EE, Black CG, Mercereau-Puijalon O, et al. Antibodies elicited in adults by a primary Plasmodium falciparum blood-stage infection recognize different epitopes compared with immune individuals. Malar J 2007; 6:86; http://dx.doi.org/10.1186/1475-2875-6-86; PMID: 17605823
  • Murhandarwati EE, Black CG, Wang L, Weisman S, Koning-Ward TF, Baird JK, et al. Acquisition of invasion-inhibitory antibodies specific for the 19-kDa fragment of merozoite surface protein 1 in a transmigrant population requires multiple infections. J Infect Dis 2008; 198:1212 - 8; http://dx.doi.org/10.1086/591943; PMID: 18717639
  • Weiss GE, Traore B, Kayentao K, Ongoiba A, Doumbo S, Doumtabe D, et al. The Plasmodium falciparum-specific human memory B cell compartment expands gradually with repeated malaria infections. PLoS Pathog 2010; 6:e1000912; http://dx.doi.org/10.1371/journal.ppat.1000912; PMID: 20502681
  • McCallum FJ, Persson KE, Mugyenyi CK, Fowkes FJ, Simpson JA, Richards JS, et al. Acquisition of growth-inhibitory antibodies against blood-stage Plasmodium falciparum. PLoS One 2008; 3:e3571; http://dx.doi.org/10.1371/journal.pone.0003571; PMID: 18958278
  • Bolad A, Nebié I, Cuzin-Ouattara N, Traore A, Esposito F, Berzins K. Antibody-mediated in vitro growth inhibition of field isolates of Plasmodium falciparum from asymptomatic children in Burkina Faso. Am J Trop Med Hyg 2003; 68:728 - 33; PMID: 12887035
  • Murhandarwati EE, Wang L, Black CG, Nhan DH, Richie TL, Coppel RL. Inhibitory antibodies specific for the 19-kilodalton fragment of merozoite surface protein 1 do not correlate with delayed appearance of infection with Plasmodium falciparum in semi-immune individuals in Vietnam. Infect Immun 2009; 77:4510 - 7; http://dx.doi.org/10.1128/IAI.00360-09; PMID: 19620342
  • Fowkes FJ, Richards JS, Simpson JA, Beeson JG. The relationship between anti-merozoite antibodies and incidence of Plasmodium falciparum malaria: A systematic review and meta-analysis. PLoS Med 2010; 7:e1000218; http://dx.doi.org/10.1371/journal.pmed.1000218; PMID: 20098724
  • Jensen JB, Boland MT, Allan JS, Carlin JM, Vande Waa JA, Divo AA, et al. Association between human serum-induced crisis forms in cultured Plasmodium falciparum and clinical immunity to malaria in Sudan. Infect Immun 1983; 41:1302 - 11; PMID: 6350183
  • Marsh K, Otoo L, Hayes RJ, Carson DC, Greenwood BM. Antibodies to blood stage antigens of Plasmodium falciparum in rural Gambians and their relation to protection against infection. Trans R Soc Trop Med Hyg 1989; 83:293 - 303; http://dx.doi.org/10.1016/0035-9203(89)90478-1; PMID: 2694458
  • Corran PH, O’Donnell RA, Todd J, Uthaipibull C, Holder AA, Crabb BS, et al. The fine specificity, but not the invasion inhibitory activity, of 19-kilodalton merozoite surface protein 1-specific antibodies is associated with resistance to malarial parasitemia in a cross-sectional survey in The Gambia. Infect Immun 2004; 72:6185 - 9; http://dx.doi.org/10.1128/IAI.72.10.6185-6189.2004; PMID: 15385530
  • John CC, O’Donnell RA, Sumba PO, Moormann AM, de Koning-Ward TF, King CL, et al. Evidence that invasion-inhibitory antibodies specific for the 19-kDa fragment of merozoite surface protein-1 (MSP-1 19) can play a protective role against blood-stage Plasmodium falciparum infection in individuals in a malaria endemic area of Africa. J Immunol 2004; 173:666 - 72; PMID: 15210830
  • Courtin D, Oesterholt M, Huismans H, Kusi K, Milet J, Badaut C, et al. The quantity and quality of African children’s IgG responses to merozoite surface antigens reflect protection against Plasmodium falciparum malaria. PLoS One 2009; 4:e7590; http://dx.doi.org/10.1371/journal.pone.0007590; PMID: 19859562
  • Crompton PD, Miura K, Traore B, Kayentao K, Ongoiba A, Weiss G, et al. In vitro growth-inhibitory activity and malaria risk in a cohort study in mali. Infect Immun 2010; 78:737 - 45; http://dx.doi.org/10.1128/IAI.00960-09; PMID: 19917712
  • Bejon P, Warimwe G, Mackintosh CL, Mackinnon MJ, Kinyanjui SM, Musyoki JN, et al. Analysis of immunity to febrile malaria in children that distinguishes immunity from lack of exposure. Infect Immun 2009; 77:1917 - 23; http://dx.doi.org/10.1128/IAI.01358-08; PMID: 19223480
  • Bejon P, Ogada E, Peshu N, Marsh K. Interactions between age and ITN use determine the risk of febrile malaria in children. PLoS One 2009; 4:e8321; http://dx.doi.org/10.1371/journal.pone.0008321; PMID: 20037643
  • Aponte JJ, Menendez C, Schellenberg D, Kahigwa E, Mshinda H, Vountasou P, et al. Age interactions in the development of naturally acquired immunity to Plasmodium falciparum and its clinical presentation. PLoS Med 2007; 4:e242; http://dx.doi.org/10.1371/journal.pmed.0040242; PMID: 17676985
  • Miura K, Perera S, Brockley S, Zhou H, Aebig JA, Moretz SE, et al. Non-apical membrane antigen 1 (AMA1) IgGs from Malian children interfere with functional activity of AMA1 IgGs as judged by growth inhibition assay. PLoS One 2011; 6:e20947; http://dx.doi.org/10.1371/journal.pone.0020947; PMID: 21695140
  • Holder AA. The carboxy-terminus of merozoite surface protein 1: structure, specific antibodies and immunity to malaria. Parasitology 2009; 136:1445 - 56; http://dx.doi.org/10.1017/S0031182009990515; PMID: 19627632
  • Okech BA, Corran PH, Todd J, Joynson-Hicks A, Uthaipibull C, Egwang TG, et al. Fine specificity of serum antibodies to Plasmodium falciparum merozoite surface protein, PfMSP-1(19), predicts protection from malaria infection and high-density parasitemia. Infect Immun 2004; 72:1557 - 67; http://dx.doi.org/10.1128/IAI.72.3.1557-1567.2004; PMID: 14977962
  • Miura K, Zhou H, Muratova OV, Orcutt AC, Giersing B, Miller LH, et al. In immunization with Plasmodium falciparum apical membrane antigen 1, the specificity of antibodies depends on the species immunized. Infect Immun 2007; 75:5827 - 36; http://dx.doi.org/10.1128/IAI.00593-07; PMID: 17923516
  • Duncan CJ, Draper SJ. Controlled Human Blood-Stage Malaria Infection: Current Status and Potential Applications. Am J Trop Med Hyg 2012; In press
  • Moorthy VS, Diggs C, Ferro S, Good MF, Herrera S, Hill AV, et al. Report of a consultation on the optimization of clinical challenge trials for evaluation of candidate blood stage malaria vaccines, 18-19 March 2009, Bethesda, MD, USA. Vaccine 2009; 27:5719 - 25; http://dx.doi.org/10.1016/j.vaccine.2009.07.049; PMID: 19654061
  • Bejon P, Andrews L, Andersen RF, Dunachie S, Webster D, Walther M, et al. Calculation of liver-to-blood inocula, parasite growth rates, and preerythrocytic vaccine efficacy, from serial quantitative polymerase chain reaction studies of volunteers challenged with malaria sporozoites. J Infect Dis 2005; 191:619 - 26; http://dx.doi.org/10.1086/427243; PMID: 15655787
  • Hermsen CC, de Vlas SJ, van Gemert GJ, Telgt DS, Verhage DF, Sauerwein RW. Testing vaccines in human experimental malaria: statistical analysis of parasitemia measured by a quantitative real-time polymerase chain reaction. Am J Trop Med Hyg 2004; 71:196 - 201; PMID: 15306710
  • Sanderson F, Andrews L, Douglas AD, Hunt-Cooke A, Bejon P, Hill AV. Blood-stage challenge for malaria vaccine efficacy trials: a pilot study with discussion of safety and potential value. Am J Trop Med Hyg 2008; 78:878 - 83; PMID: 18541763
  • Douglas AD, Andrews L, Draper SJ, Bojang K, Milligan P, Gilbert SC, et al. Substantially reduced pre-patent parasite multiplication rates are associated with naturally acquired immunity to Plasmodium falciparum. J Infect Dis 2011; 203:1337 - 40; http://dx.doi.org/10.1093/infdis/jir033; PMID: 21459819
  • Ogutu BR, Apollo OJ, McKinney D, Okoth W, Siangla J, Dubovsky F, et al, MSP-1 Malaria Vaccine Working Group. Blood stage malaria vaccine eliciting high antigen-specific antibody concentrations confers no protection to young children in Western Kenya. PLoS One 2009; 4:e4708; http://dx.doi.org/10.1371/journal.pone.0004708; PMID: 19262754
  • Sagara I, Dicko A, Ellis RD, Fay MP, Diawara SI, Assadou MH, et al. A randomized controlled phase 2 trial of the blood stage AMA1-C1/Alhydrogel malaria vaccine in children in Mali. Vaccine 2009; 27:3090 - 8; http://dx.doi.org/10.1016/j.vaccine.2009.03.014; PMID: 19428923
  • Sheehy SH, Duncan CJ, Elias SC, Collins KA, Ewer KJ, Spencer AJ, et al. Phase Ia clinical evaluation of the Plasmodium falciparum blood-stage antigen MSP1 in ChAd63 and MVA vaccine vectors. Mol Ther 2011; 19:2269 - 76; http://dx.doi.org/10.1038/mt.2011.176; PMID: 21862998
  • Douglas AD, Williams AR, Illingworth JJ, Kamuyu G, Biswas S, Goodman AL, et al. The blood-stage malaria antigen PfRH5 is susceptible to vaccine-inducible cross-strain neutralizing antibody. Nat Commun 2011; 2:601; http://dx.doi.org/10.1038/ncomms1615; PMID: 22186897
  • Wilson DW, Fowkes FJ, Gilson PR, Elliott SR, Tavul L, Michon P, et al. Quantifying the importance of MSP1-19 as a target of growth-inhibitory and protective antibodies against Plasmodium falciparum in humans. PLoS One 2011; 6:e27705; http://dx.doi.org/10.1371/journal.pone.0027705; PMID: 22110733
  • Ockenhouse CF, Sun PF, Lanar DE, Wellde BT, Hall BT, Kester K, et al. Phase I/IIa safety, immunogenicity, and efficacy trial of NYVAC-Pf7, a pox-vectored, multiantigen, multistage vaccine candidate for Plasmodium falciparum malaria. J Infect Dis 1998; 177:1664 - 73; http://dx.doi.org/10.1086/515331; PMID: 9607847

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.