Skip to main content

Plasmodium falciparum pre-erythrocytic stage vaccine development

Abstract

Worldwide strategies between 2010 and 2017 aimed at controlling malarial parasites (mainly Plasmodium falciparum) led to a reduction of just 18% regarding disease incidence rates. Many biologically-derived anti-malarial vaccine candidates have been developed to date; this has involved using many experimental animals, an immense amount of work and the investment of millions of dollars. This review provides an overview of the current state and the main results of clinical trials for sporozoite-targeting vaccines (i.e. the parasite stage infecting the liver) carried out by research groups in areas having variable malaria transmission rates. However, none has led to promising results regarding the effective control of the disease, thereby making it necessary to complement such efforts at finding/introducing new vaccine candidates by adopting a multi-epitope, multi-stage approach, based on minimal subunits of the main sporozoite proteins involved in the invasion of the liver.

Background

Human malaria is a transmissible disease having high morbi-mortality worldwide; it is caused by five parasite species from the genus Plasmodium: Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae and Plasmodium knowlesi (P. falciparum having the highest mortality rate) [1, 2].

Following the discovery of the parasite’s life-cycle which begins when the sporozoite (Spz) form is transmitted to humans during the bite of a female Anopheles mosquito [3], efforts at eliminating the disease became aimed at eliminating the vector and its habitats. After the failure of that strategy, the World Health Organization (WHO) efforts were aimed at promoting control programmes, which included long-lasting insecticide-treated mosquito nets and indoor spraying with residual insecticides, anti-malarial drug treatment and early and rapid diagnosis. Government entities in countries having malaria-endemic areas invest around 6.5 billion US dollars annually for controlling the disease worldwide [2].

Despite many efforts and scientific advances, the control and prevention of the disease has still not been achieved, as the WHO estimated 219 million cases of malaria and 435,000 malaria-related deaths for 2017, 93% of which were reported in sub-Saharan Africa, especially in children aged less than 5 years old and in pregnant women. It also estimated that the incidence rate between 2010 and 2017 had only become reduced by 18% [2]. Such statistics increasingly highlight the need for a global attack on malaria, including the development of an integral, multi-epitope, multi-stage, long-lasting vaccine able to induce a cellular and humoral immune response (IR) [4] as a fundamental, complementary and valuable tool for optimizing existing malaria control strategies. Contributing towards eliminating the disease would thereby help save hundreds of thousands of lives every year [2].

The female Anopheles mosquito injects a minimum of Spz (~ 100) during its bite [5, 6]; these remain at the inoculation site, moving in the dermis and seeking a capillary to enable them to migrate towards hepatocytes (having a high heparan sulphate proteoglycan (HSPG) content in their membrane) to invade them. This can last from 10 to 40 min, making Spz highly susceptible to a host IR, involving such a small amount of Spz and infected liver cells. This creates a bottleneck for the parasite during its reproductive cycle, making vaccines targeting Spz proteins and those from the parasite’s hepatic stage attractive vaccine candidates.

As this stage lasts 5.5 to 7 days, prolonging the length of exposure to the IR can detain infection, thereby hampering parasite development in the liver before symptoms appear during the blood stage, gametocyte production and the perpetuation of the parasite’s life-cycle (Fig. 1). Such fundamental approach complements vaccine candidates targeting the asexual erythrocyte stage during which millions of merozoites (Mrz) become exposed to the immune system during the extremely short period of time of around 1–2 min, thereby reducing the chances of success for such an approach [7, 8].

Fig. 1
figure 1

The P. falciparum life-cycle. An infected female Anopheles mosquito inoculates Spz as it bites a host, they then travel in the host’s bloodstream and infect the hepatocytes. Merozoites are released and then invade erythrocytes, where they mature through various stages (ring, trophozoite and schizont stages) and undergo asexual multiplication (~ 10 or lower) every 48 h, releasing new merozoites which perpetuate the asexual cycle. Some of them enter the sexual cycle by becoming female and male gametocytes which are ingested by the mosquito when it bites an infected host, thereby starting the cycle all over again

Based on prolonged IR exposure time, efforts have been focused on developing vaccines targeting Spz proteins. The WHO’s recent update [9] reported that vaccine candidates in clinical phase trials include attenuated Spz vaccines (radiation-attenuated Spz, Spz administered under drug coverage and genetically-attenuated Spz vaccines), recombinant protein vaccines (RTS,S and R21) and recombinant viral vectors vaccines (Chad63 MVA ME-TRAP, CSVAC, ChAd63 METRAP and MVA METRAP with the matrix-M adjuvant) (Table 1).

Table 1 Clinical phases for developing vaccines against sporozoite stage malaria

This review has been aimed at analysing the formulation, dose, safety and immunogenicity of current clinical trials being carried out regarding vaccine candidates’ differing study phases, and including the structure of some protein fragments being studied.

Clinical trials for pre-erythrocyte stage anti-malarial vaccines

The main thrust of research groups developing vaccines against the P. falciparum malaria Spz stage has involved Spz recombinant proteins, DNA or viral vectored protein fragments and attenuated Spz vaccines to induce malaria reactive CD4+ and CD8+ T-lymphocyte counts and high antibody (Abs) titres. Unfortunately, the most advanced candidate formulations to date have had limited efficacy. However, there have been significant developments regarding phase I, II and III trials (Table 1), which should prove useful for further vaccine development.

Attenuated sporozoite vaccines

It has been demonstrated that Abs produced by immunization with whole, attenuated Spz prevent the development of hepatic infection and can immobilize free Spz in the avascular dermis or prevent erythrocyte stage development [10]. Vaccines based on this approach have included radiation-attenuated Spz (RAS), genetically-attenuated parasite (GAP) and Spz administered under drug coverage [11].

Many studies have been aimed at improving attenuated Spz vaccines, focusing on efforts at producing a large repertoire of immunogens, evaluating the impact of a particular regime, dosage and inoculation route, thereby enabling an effective cellular and humoral immune response to be achieved [12].

Radiation-attenuated sporozoites

The P. falciparum Spz (PfSPZ) vaccine is the main candidate containing live, radiation-attenuated, whole, aseptic and metabolically active Spz which have been isolated from the salivary glands of mosquitos infected by P. falciparum [13, 14]. Pioneering studies evaluated the effect of radiation on Plasmodium berghei Spz ability to invade and develop in mouse livers, demonstrating that infection became reduced with higher radiation doses [15] and that mice immunized with X-ray-irradiated P. berghei Spz became protected against homologous challenge and challenge with Plasmodium vinckei [16,17,18,19].

Clinical trials with attenuated Spz were carried out on 11 human volunteers based on the foregoing experimental findings; the volunteers were immunized with more than 1000 bites by irradiated mosquitos infected by Spz from the P. falciparum NF54 strain or 3D7/NF54 clone. All participants were protected against a first homologous challenge [20]; however, only 2/10 volunteers were protected against challenge with the P. falciparum 7G8 strain (heterologous challenge). Such results showed that attenuated Spz immunization could represent a good methodology for developing anti-malarial vaccine candidates, though involving the inconvenience of an impractical administration route despite having demonstrated 90% to 95% effectiveness concerning homologous challenge [20, 21].

It has been demonstrated that immunization by mosquito bite deposits Spz in the dermis and subcutaneous tissue; however, it has not yet been possible to replicate this by innoculation using a standard needle. This has led to many efforts at equalling the efficacy of the classical RAS vaccine, evaluating variables such as the delivery method, the inoculation route and the dose to be administered [10, 13, 22].

Recognizing this limitation, one study has evaluated the safety and immunogenicity of different doses of the PfSPZ vaccine via subcutaneous (SC) vs. intradermal (ID) route. It reported that 2/16 volunteers in the group who had received 4 doses of 3 × 104 PfSPZ became protected and that protected volunteers, one immunized by ID and the other via SC, had T-cell responses to PfSPZ and antibodies (200 and 800 titres) [13].

An open-label trial was performed to evaluate other administration routes in which 64% of volunteers became protected after homologous challenge with the Pf 3D7 strain clone in controlled human malaria infection (CHMI) 19 weeks (~ 4.5 months) later. Subjects who did not have parasitaemia were submitted to a repeat heterologous challenge 33 weeks (~ 8 months) after final immunization with the P. falciparum 7G8 heterologous strain, of these 83% remained without parasitaemia. These results suggested that the PfSPZ vaccine could achieve limited but lasting protection against heterologous strains (~ 8 months or 33 weeks), although CD4+ and CD8+ T-cell responses did not increase, being limited after the second and third immunization [23].

The vaccine was well-tolerated in a clinical trial in Malí [24], having 29% efficacy against heterologous strains during 24-week (~ 6 month) follow-up without incurring in any serious local or systemic adverse events (AE). Effectiveness 3 to 24 weeks (~ 1 to 6 months) after the last immunization was evaluated by homologous intravenous CHMI which showed that 20% of the subjects who received 5 doses of 2.7 × 105 PfSPZ had become fully protected [25].

Promising results were obtained in homologous PfSPZ CHMI prepared with NF54 strain Spz [26]. However, vaccine effectiveness became considerably reduced to 10% after challenge with the heterologous strain (no grade 3 or 4 AE being recorded) [27].

Immunization doses were increased to 9.0 × 105 PfSPZ and 1.8 × 106 PfSPZ in adolescents, children and infants aged 6 months old and older to assess the effects of the PfSPZ dose and the immune response of children and infants who had been less exposed to P. falciparum compared to adults pre-exposed to long-term P. falciparum infection [28]. No significant differences were found in any age group regarding AE amongst vaccinated volunteers. On the other hand, it was found that most vaccinees developed antibodies (Abs) against PfCSP when evaluating the humoral immune response, a higher response being observed in children aged 6 to 10 years old who had received 1.8 × 106 PfSPZ [28].

Higher Abs responses in children and infants who had been less exposed to P. falciparum [28] and subjects living in non-endemic areas [27] suggested that Africans’ reduced immune responses were due to immunoregulation following long-term exposure to P. falciparum infection [24, 25]. All such efforts have shown that PfSPZ efficacy in adults who have not had prior exposure to P. falciparum depends on the administration route (to induce tissue resident T cells in the liver) and the dose (which determines the degree of protection durability against homologous and heterologous challenge). This highlights the need for an improved dosage strategy and/or an alternative vaccine approach in malaria-endemic areas [12].

It is expected that a phase III trial involving around 2100 people aged 2 to 50 years-old will begin in early 2020 on Bioko, an island off the Equatorial Guinea coast. The trial’s objective is to provide data regarding the necessary efficacy and safety for regulatory authorities’ approval. If the trial is successful, Sanaria intends to carry out another clinical trial involving a further 10,000 people on the island (Hoffman S, personal communication).

Sporozoites administered under drug coverage

This approach has highlighted the fact that an anti-malarial vaccine based on immunization with live Spz and chemo-prophylactic cover of chloroquine (CPS-CQ) has achieved protection in 100% of the volunteers 8 weeks after the final immunization, such protection persisting for up to 2 years [29]. Furthermore, it has been reported that inducing high protection depends on the dose in homologous CHMI [29, 30].

Another trial which included live Spz evaluated chemo-prophylactic cover of mefloquine (CPS-MQ), finding similar safety and efficacy profiles (~ 60%) as those for CPS-CQ [31]. Moreover, IV administration of non-irradiated cryopreserved Spz to malaria-naive, healthy adult volunteers taking chloroquine as part of prophylactic anti-malarial treatment (vaccine approach denoted as PfSPZ-CVac) also gave 100% efficacy (9/9 volunteers) against homologous CHMI [32].

Different immunization regimens and pharmacological alternatives such as atovaquone/proguanil, azithromycin and pyrimethamine are currently being studied for developing safer and more effective methodological alternatives [22].

Genetically-attenuated sporozoite vaccines

Another approach concerns genetic manipulation modifying, eliminating or attenuating genes from parasites and altering hepatic stage infection development [33]. Genetically attenuated parasite P36p gene-deficient Spz, have induced protection-inducing immunity against P. berghei in mice, demonstrating the lack of infection during blood stage [34].

The first clinical trial evaluating vaccine safety and immunogenicity in 6 volunteers who received p52 (−)/p36 (−) Spz GAP through the bites of infected Anopheles mosquitoes showed that the vaccine was well tolerated, having mild to moderate local and systemic reactions. Only 1 out of the 6 volunteers developed parasitaemia 12 days after exposure [35].

A phase I clinical trial, involving 10 volunteers tested the p52–/p36–/sap1– (PfGAP3KO) vaccine lacking three genes expressed during P. falciparum pre-erythrocyte stage. This was administered by mosquito bite, mild to moderate AE being reported and the absence of parasitaemia up to day 28 after the last immunization. This demonstrated complete PfGAP3KO attenuation, pre-erythrocyte development becoming arrested. Humoral immune response analysis showed that all subjects developed considerable IgG anti-circumsporozoite protein (CSP) titres [36], thereby supporting the claim that PfGAP3KO is a safe and immunogenic candidate. Efficacy data is expected for this and another candidate involving genetically attenuated P. falciparum Spz (NF54 strain) (PfSPZ-GA1) by eliminating the b9 gene and Spz and liver stage asparagine-rich protein. (SLARP) genes which are important for parasite development during liver stage [22, 37].

Plasmodium falciparum CSP is located on Spz surface and is crucial for parasite morphogenesis and host invasion. It has variable length and 40 to 60 kDa molecular weight. It has an N-terminal domain containing region I, followed by a tandem repeat region consisting of the asparagine-alanine–asparagine-proline (NANP) amino acid (aa) motif repeated 20 to 40 times, interspaced four times with asparagine-valine-aspartate-proline (NVDP) and asparagine-proline-aspartate-proline (NPDP). It has a C-terminal domain (CTD), comprising region II and a glycosylphosphatidylinositol (GPI) anchor sequence [38, 39] (Fig. 2a, c).

Fig. 2
figure 2

a Schematic representation of P. falciparum’s CSP1 (NF54 strain), showing signal peptide (orange), region I (blue), the central repeat regions (yellow, green and red) and region II (blue) with GPI anchor (pink). b Schematic representation of RTS,S vaccine, showing the central repeat regions (orange, grey) and PfCSP1 region II (blue) and hepatitis B virus (purple) surface antigen (S). c Ribbon and surface representation of PfCSP1 region II. (PDB: 3VDK) [177]. d Schematic representation of ChAd63/MVA ME-TRAP vaccine candidate. Left-hand side, above, pSG2 plasmid used to express the ME-TRAP vaccine candidate in either ChAd63 or MVA viruses involving kanamycine resistant (KanR) (in blue) cytomegalovirus, with intron A (pCMV IntA) (in red), bovine growth hormone with terminator polyA (BGH poly-A) (in orange) and Escherichia coli B-galactosidase genes (in yellow). Right-hand side, above, PfTRAP TRS domain in ribbon and surface representation (PDB 2BBX) [178]. Bottom, PfTRAP Von Willebrand factor A domain (vWA), in ribbon and surface, showing the MIDAS domain residues (blue). e pSG2 plasmid used to express the CS vaccine candidate in either ChAd63 or MVA viruses with the same vector as represented in E

An immunological response against NANP repeats has been a crucial point in developing CSP-basad vaccines. An analysis of the interaction between human monoclonal antibodies (mAbs) (RTS,S vaccine-derived 31, 317, Mal1C, Mal2A and Mal3B) and NANP repeats has led to identifying minimal epitope binding and confirming that an increase in the amount of Ab contacts can improve affinity for the repeats in this sequence [38, 40].

Recent studies have described mAbs CIS23, CIS34, CIS42 and CIS43 isolated from P. falciparum CSP-specific memory B-cells from volunteers who had been immunized with the PfSPZ vaccine [41,42,43]. CIS43 and MGG4 mAb had cross-reactivity with NPDP, NVDP and NANP repeat regions and the CTD fragment, thereby enabling them to bind to this protein and alter its cleavage after processing to limit hepatocyte invasion in an animal model [42,43,44]. The next step will involve clinical trials being run by PATH’s Malaria Vaccine Initiative for determining whether mAbs can induce protection against P. falciparum infection.

Recombinant protein vaccines

Recombinant vaccines can use one or multiple specific antigens to induce an immunological response against the parasite. They can be boosted when co-administered with adjuvants, thereby overcoming problems such as reverse virulence and the difficulty of obtaining sufficient amounts of the antigen to facilitate large-scale production [45]. However, using unsuitable antigens having low immunogenicity and a high genetic variation rate has limited the emergence of efficient vaccines against diseases such as malaria [46]. One of the main antigens involved in this approach has been P. falciparum CSP [11]  used as a subunit in the RTS,S vaccine.

RTS,S

The RTS,S vaccine has been the most studied and publicized anti-malarial vaccine candidate in clinical phase trials according to WHO malaria vaccine guidelines [44]. RTS,S consists of a large segment (amino acids 207 to 395) of the P. falciparum NF54 strain CSP protein in which many variable epitopes has been identified [47, 48]. A tetrapeptide from the CSP NANP tandem repeat region (R) and the C-terminal region containing T-cell (T) epitopes (exclusive for the NF54 strain) become fused to hepatitis B surface (S) antigen (HBsAg) expressed in Saccharomyces cerevisiae yeast cells (Fig. 2b). These self-assemble into virus-like particles (VLP) and have a maximum 20% RTS sequence insertion into VLP [49].

The AS01 and AS02 adjuvant systems were well-tolerated and RTS,S/AS01 induced the highest anti-CSP and CD4+ T-cell responses, compared to RTS,S/AS02 when three doses were administered to children and infants instead of two doses [50,51,52,53]; these Abs persisted for at least three and a half years following immunization [54].

Anti-CSP antibody concentrations after a single RTS,S/AS02 booster dose (19 months after initial immunization), persisted for a further 5 years, even though titres became reduced to 4.7 μg/ml from levels preceding the booster dose [55]. Cellular and humoral immunological responses were associated, with protection-inducing responses against asymptomatic and symptomatic parasitaemia states [56, 57]. However, the considerable variation in such results was inexplicable; for example, children might have suffered malaria in spite of having had high anti-CSP titres [58].

Clinical trials have estimated that the vaccine had 30–86% efficacy following the last immunization using a standard three 50 μg dose scheme. However, this became reduced to 0% during the last weeks of follow-up [59,60,61,62].

Clinical evaluation results have suggested that RTS,S can be considered safe in spite of it inducing slight to moderate local reactogenicity, tending to escalate with an increase in dose regardless of age [49, 63, 64]. All doses were highly immunogenic, inducing anti-CSP and anti-HBsAg Abs, this being greater in children aged 1 to 5 years-old [65, 66]. Furthermore, it has been demonstrated that the inductor effect of RTS,S/AS02 protection is not associated with any particular Human Leukocyte Antigen (HLA) allele [60].

Safety and immunogenicity data have provided the basis for expanding the evaluation of new dosing strategies, vaccination schedules and extending the follow-up period, using larger samples of paediatric populations residing in malaria-endemic regions [49, 62].

Clinical trials in different aged paediatric populations have estimated 25.6–53% efficacy for at least 18 months’ follow-up and 0% after 3 years; this can be attributed to the intensity of transmission, the choice of adjuvant and the age of the population when being immunized [67,68,69,70,71]. However, significantly higher Abs responses have been reported after the third dose, even though these have not been long-lasting [72].

As the target population for immunization with RTS,S was infants, its safety and efficacy profile has been investigated due to being administered with other vaccines included in the Expanded Programme on Immunization (EPI) [73]. It was found that RTS,S did not interfere with the immunological responses of EPI antigens co-administered with it in infants [74] and that it had 52.5% efficacy against a first or single episode of malaria and 59.1% efficacy against all episodes during a 19-month period [73]. RTS,S/AS01E’s favourable safety profile suggested that the vaccine could be administered using a 0, 1 and 2 month scheme, which is why this scheme was chosen for a clinical evaluation in a multicentre phase III trial, delivering the vaccine via EPI. It was demonstrated that a scheme involving a complete dose of RTS,S at 0 and 1 months, together with a third fractioned dose at 7 months, increased protection against CHMI (86%) and improved immunogenicity by increasing specific antibody avidity and somatic hyper-mutation frequency in B-cells. The effect of changes in the vaccination scheme and the dose on protection-inducing immunity and vaccine efficacy must thus be studied in depth [75, 76].

A double blind, randomized controlled trial was carried out between 2009 and 2014 for evaluating RTS,S efficacy. It involved 15,460 participants divided into two age groups (6 to 12 week-olds and 5 to 17 month-olds) in 7 sub-Saharan Africa countries having different malaria transmission rates 14 months after the first vaccination, finding 34% efficacy against severe malaria in the combined age categories and 55.8% against clinical malaria in the 5 to 17 month-old group [77]. After 4 years follow-up, efficacy against episodes of clinical malaria was greater in the 5 to 17 month-old group (36.3%) compared to the 6 to 12 week-old group (25.9%) and against severe malaria (32.2% and 17.3%, respectively) [78].

It was found during a 7-year follow-up of a group of infants aged 5 to 17 months old who had received RTS,S that the efficacy of the vaccine against all episodes of malaria became reduced to − 3.6% in the fifth year and that average efficacy was 4.4% during the follow-up period [79]. Protection became reduced as time elapsed, becoming undetectable or exhibiting − 48% to − 56% negative efficacy during the last study period in the group which received three doses. This led to including a booster dose in the vaccination scheme after infants became 5 months old, considering that efficacy was lower in infants [78, 80].

RTS,S safety profile has been confirmed according to the data from phase I–III trials where local and systemic grade 3 AE incidence was low, study groups having similar frequency [78, 80, 81]. The fourth dose of RTS,S/AS01 was more reactogenic, having more systemic and local AE during the 7 days following vaccination compared to the group which received just three doses [58, 78]. Severe malaria incidence became reduced following vaccination with 50 µg RTS,S/AS01 in 3-year-old children in Tanzania, Kenya and Burkina Faso during 7-year follow-up, regardless of immunization scheme [82].

The European Medicines Agency (EMA) evaluated RTS,S’s clinical development in 2015, issuing a cautious scientific opinion regarding its quality [83], even though pre-clinical studies’ results are only being published 20 years after its clinical evaluation began. In a recently publicized trial, the WHO has recommended carrying out pilot introduction (with 360,000 participants) in three sub-Saharan countries (Kenya, Malawi and Ghana) having moderate to high levels of malarial transmission and only administering the four-dose scheme in the 5 to 17 month-old age group. It also suggested an initial scheme as being 3-dose, with a minimum 4-week interval between doses, followed by a 4th dose 15–18 months after the 3rd dose [84].

Several points regarding RTS,S have raised concern, such as high parasitaemia levels in individuals considered “protected” (> 5000 parasites/µl or 0.1% parasitaemia) [77, 78, 80] and the selected CSP region’s high genetic variability [85,86,87,88]. A not fully-defined adjuvant system has been used, mainly consisting of QS-21 (a saponin inducing cell activation through poorly understood mechanisms) [89,90,91], some RTS,S components have induced proapoptotic signals [92, 93] and it has had short-term efficacy [75, 78].

R21

The R21 subunit-based vaccine is based on a single fusion protein; it consists of the P. falciparum NF54 strain CSP C-terminus bound to the HBsAg N-terminus. It has been developed as an improved version of RTS,S, containing a larger amount of CSP compared to HBsAg, promoting potent humoral immune responses to CSP and minimum Ab for the HBsAg portion. Efficacy against exposure to a transgenic Spz improved when BALB/c mice were given low doses of R21 [94].

A clinical trial carried out between 2015 and 2017 evaluated R21 safety and immunogenicity when administered with the ASO1 adjuvant; 20 healthy English participants received three doses of the vaccine on days 0, 28 and 56 of the trial. Good anti-CSP Ab responses were observed after a 6-month follow-up when using 10 μg and 50 μg doses, this being comparable with RTS,S levels induced against malaria. Both doses were well-tolerated, however there were safety-related AE. This study is registered in (ClinicalTrials.gov: NCT02600975), although no further information has been published.

Recombinant viral vectors vaccines

Viral vectors represent promising tools for vaccine development, because they enable intracellular antigens to be expressed by increasing the ability to generate robust cytotoxic T-lymphocyte responses and proinflammatory interferon and cytokine production without the need for an adjuvant [95]. However, there is great concern regarding their genotoxicity due to possible viral genome integration; this has led to many efforts aimed at finding a high level of safety and efficacy.

Several viral [96,97,98,99,100], bacterial [101,102,103,104] and parasite [105,106,107] vectors have been used in anti-malarial vaccine candidates; currently, many clinical trials are exploring their advantages to increase their potential and accelerate their use in vaccines [11, 108].

Chad63 MVA ME-TRAP

This anti-malarial vaccine was developed using chimpanzee adenovirus 63 (Chad63) and modified Vaccinia virus Ankara (MVA) into which were inserted genes encoding the thrombospondin-related adhesion protein (TRAP) multiple epitope (ME) chain [109, 110].

The ME-TRAP hybrid is thus a 2398 base pair (bp) insert encoding a single 789 aa-long peptide, covering the complete P. falciparum TRAP sequence, fused to a chain of 20 malaria T- and B-cell epitopes (14 targeting MHC class I, 3 MHC class II and 1 murine) (Fig. 2d) [111].

The MVA virus is highly attenuated and has been used efficiently as a non-replicating viral vector for developing new vaccines [112]. Chad63 serotypes do not circulate in human populations and thus neutralizing antibodies targeting them have seldom been demonstrated [113].

TRAP belongs to a family of proteins found in the micronemes during the invasion stages of parasites from the phylum Apicomplexa and in apical complex secretor vesicles. It is a 63 kDa, ~ 550 aa-long, conserved type I microneme protein, having two binding regions: the von Willebrand type A1 (VWA) region I, which includes the metal-ion-dependent-adhesion-site (MIDAS) and the TSR domain (region II), known for its role in protein–protein interactions. It also has a proline-rich region (region III), a transmembrane domain (region IV) and acidic C-terminal cytoplasmic tail (Fig. 2d) [114].

Sequential administration of MVA and Chad63 vectors, spaced by an interval of time (primary heterologous booster dose), is aimed at inducing CD4+ and CD8+ T-cells producing interferon gamma (IFN-ɣ) due to their main role in mediating protection during the hepatic stage [115].

A study with 54 participants, reported 184 local AE 28 days after initial vaccination (pain, erythema, oedema, pruritus and heat). All participants who received ID route vaccination reported local AE, lower incidence being reported by those who had received ChAd63 ME-TRAP by intramuscular (IM) route [116], thereby concluding that the ID route was associated with greater local reactogenicity compared to the IM route [111].

Systemic AE reported in a phase I study included fatigue (87%), general discomfort (69%) and fever (54%); 69% of them occurred and were resolved during the first 48 h after vaccination, increasing with vaccine dose regardless of administration route [116]. Such data is contrary to that described in another study where greater reactogenicity associated with vaccination route occurred (IM compared to ID) (i.e. no significant difference between doses) [110]. This study concluded that MVA ME-TRAP was more reactogenic than ChAd63 as it had greater AE incidence; however, both were well-tolerated [110].

Regarding the alterations reflected in the laboratory tests, there were increased transaminase levels following vaccination with ChAd63 ME-TRAP at the expense of alanine aminotransferase (ALT), eosinophilia and thrombocytopenia; this became resolved in 4 out of 54 participants [115]. This was contrary to that described in a study involving west-African children where no alterations in the participants’ haematological and biochemical tests were reported following vaccination [110].

A trial involving adults in Senegal [117] to assess vaccine efficacy using a polymerase chain reaction (PCR) assay was able to detect > 10 parasites/μl blood. PCR was positive for 12 out of 57 participants vaccinated with ChAd63 ME-TRAP with a booster dose of MVA ME-TRAP and 13 out of 58 control patients who received an anti-rabies vaccine were positive by PCR, giving 8% efficacy (which was not statistically significant). They thus grouped the results with the 67% efficacy obtained in a study in Kenya and, using Cox regression, showed 50% overall vaccine efficacy in both populations [117, 118].

CSVAC

CSVAC, a vaccine from Chad63 and MVA to encode the P. falciparum CS protein, continued such line of research into plasmid DNA anti-malarial vaccines; the CS insert was a codon-optimized cDNA encoding the CS protein truncated at the C-terminal extreme thereby lacking 14 C-terminal aa and thus omitting the GPI anchor (Fig. 2f) [119].

No serious AE were found when evaluating this vaccine’s safety profile; 91% were slight and 80% were resolved within 48 h. It was found that 58% of the 24 volunteers had suffered one or more local AE following vaccination with ChAd63 CS compared to 83% of the volunteers suffering one or more systemic AE following vaccination, mostly affecting participants who had received 5 × 1010 vp ChAd56CS doses; it was concluded that MVA CS was more reactogenic in 87% of the volunteers [120].

The antigen-specific T-cell responses of two doses of ChAd63 CS were compared between group I (5 × 109 vp) and group II (5 × 1010 vp) for evaluating immunogenicity. Reduced levels was reported up to day 56 (not statistically significant); responses in all volunteers increased significantly 7 days after administrating MVA CS, followed by a gradual decrease until follow-up day 140 [120].

CD4+ and CD8+ T-cell polyfunctionality was also evaluated, concluding that CD4+ produced greater TNF and IL2 levels, unlike IFNɣ values produced in similar amounts by CD4+ and CD8+ (no significant difference) [120].

All volunteers had IgG titres below the detection limit on day zero. The MVA CS booster dose produced a significant increase in Ab concentration on day 84 in group 1B compared to group 1A without booster dose; likewise, average Ab response was greater in group 2B compared to group 1B on day 140 (no statistically significant difference) (Table 1 gives detailed information about the groups) [120].

A CHMI study with P. falciparum Spz, involving a challenge which consisted of the infectious bites of 5 mosquitos evaluated vaccination efficacy by combining ChAd63/MVA CS with ChAd63/MVA ME-TRAP [121]. They reported that all infectivity controls (100%) and 27/30 (90%) of vaccinated participants were diagnosed with malaria and that 85% experienced at least one severe AE after challenge. They concluded that ME-TRAP had greater clinical efficacy by inducing sterile protection in 2 out of 15 participants (13%), unlike ChAd63/MVA CS which induced sterile protection in 1 out of 15 vaccinated participants (7%).

ChAd63 METRAP and MVA METRAP with Matrix-M adjuvant

Vaccine candidates ChAd63 METRAP and MVA METRAP safety and immunogenicity have been evaluated when they have been administered with Matrix-M, a saponin-based adjuvant which stimulates the immune response and antigen presentation to local lymph nodes [122].

No increase in local reactogenicity was revealed in a phase I study involving 23 participants vaccinated with this adjuvant, pain in the inoculation area being the most commonly occurring local AE. More systemic AE were reported in the group which received the vaccine with the adjuvant, fever having greater prevalence in 8 volunteers (3 in the control group, 2 in the 25 µg Matrix-M group and 3 in the 50 µg Matrix-M group). Regarding cellular and humoral immunogenicity, there were no differences between control group and the group which received the vaccine with the adjuvant [123].

Considering the objective of using an adjuvant to boost an antigen-induced IR, the authors concluded that using the Matrix-M adjuvant had not lead to significant changes in vaccine immunogenicity [123].

Future directions

Recent scientific advances have given rise to the need for safer formulations increasing antigen efficacy. “Nanovaccinology” has emerged during the last few years, which will surely come to play an important role in malaria vaccine development [124].

Using nanoparticles has enabled antigen stability, immunogenicity, selective administration and slow release to become improved [124]. Such characteristics have facilitated developing different vaccines from nanoparticles which have been approved for human use, varying in composition, form, surface properties and size (1–1000 nm) similar to cell components, enabling them to enter cells via mechanisms such as pinocytosis [125,126,127].

Nanoparticles have been used as delivery systems for vaccine candidates aimed at preventing disease caused by viral and bacterial, parasite and fungal pathogens [128,129,130,131], as well as non-infectious disease like cancer [132,133,134], Alzheimer’s [135], hypertension [136] and nicotine addiction [137]. Regarding parasitic diseases, CSP protein of P. falciparum has been encapsulated thereby enabling better Abs responses inhibiting the invasion of hepatocytes, inducing an immunological response which could contribute towards developing long-lasting protection-inducing immunity [138,139,140,141].

A promising alternative delivery system for subunit-based vaccines has been developed recently [134] and used with vaccine candidates against several infectious diseases such as HIV [142], toxoplasma [143,144,145], SARS [146], influenza [147] and/or malaria [148,149,150]. The technique is known as Self-Assembling Protein Nanoparticles (SAPNs) and involves the expression of a peptide/protein containing a target antigen covalently linked to an adjuvant sequence (flagellin-derived) and, in some cases, a universal epitope such as the Pan-DR T-helper epitope (PADRE) sequence. This peptide/protein can self-assemble in specific conditions, thus forming ~ 20–50 nm nanoparticles and, when formulated or emulsified with an adjuvant such as GLA-SE or Army Liposome Formulation (ALF), has managed to produce a protection-inducing response against several diseases [151, 152].

However, further studies are required to expedite understanding of how changes in nanoparticle properties might affect an immunological response against malaria and thus contribute towards effective vaccine design [153].

On the other hand, advances have been made in the fields of bioinformatics, genetic engineering and molecular biology, contributing towards using alternative methodological approaches. One such approach is reverse vaccinology for the relatively rapid identification of vaccine candidate molecules based on in silico analysis of complete sequences from the genomes of various pathogens for studying and evaluating their microbial biology and host–pathogen interactions [154,155,156]. Such methodology can be used with culturable and non-culturable microorganisms and, together with computational analysis, enables DNA sequences encoding proteins playing important roles in parasite biology to be identified and therefore become possible vaccine candidates [107, 108].

Conclusions

The great scientific progress made regarding research into anti-malarial vaccine candidates over the last four decades has resulted from strategies promoted by scientific, academic and government institutions worldwide and extensive and generous support by official entities and philanthropic organizations clearly and deeply committed to resolving the malaria conundrum.

Current anti-malarial vaccine candidates have had limited efficacy due to the intrinsically complex problem and the multiple factors governing an appropriate immune response and the amount of external factors. The choice of antigen to be used is quite complicated due to factors such as the parasite’s complex life-cycle involving two reproduction cycles (sexual and asexual), different development stages and two hosts (the Anopheles mosquito and human beings). All this can be added to the multiple invasion routes described so far for each of its target cells (hepatocytes and/or erythrocytes), the parasite’s ability to modify its gene expression and the genetic variability between P. falciparum circulating strains [157,158,159,160,161].

Likewise, results to date have led to the conclusion that whole organism- or subunit-based vaccines involving a single parasite variant are insufficient to cover its wide genetic diversity.

Developing an anti-malarial vaccine based on sub-units derived from the proteins involved in parasite invasion and infection (multi-epitope) covering the parasite’s different forms (multistage) for overcoming such complications has been suggested for several decades now. Such subunits must consist of sequences which are conserved amongst P. falciparum circulating strains to induce a strain-transcending vaccine and overcome the parasite’s genetic variability [4, 114, 162, 163].

The next major challenge concerns the host’s genetic variability, particularly major histocompatibility class II (MHCII) complex molecules exerting their mechanism by synthesizing proteins encoded by the HLA-DR regions β1*, β3*, β4* and β5* where the HLA-DR β1* region encodes more than 1500 genetic variants grouped into 16 allele families called HLA-DRβ1*01, *03, *04, *07, etc. [164, 165]. Parasite proteins’ interaction with the human immune system should be analysed by predicting B and T epitopes (using NetMHCIIpan 3.2 or other predictors) and/or in vivo evaluation in models such as the Aotus monkeys (highly susceptible to developing human malaria and having a ~ 90% identical immune system with that of humans) [166,167,168,169,170,171,172].

Various adjuvants and delivery systems have been developed for improving vaccine efficacy. Clinical trials for Spz-stage anti-malarial vaccines have involved using adjuvants consisting of a combination of immunostimulants and viral vectors. The AS01 adjuvant has been used in RTS/S, consisting of a combination of immunostimulants, monophosphoryl lipid A (MPL) in a liposome formulation and Quillaja saponaria fraction 21 (QS21) in water-in-oil emulsion [91, 173].

Chimpanzee adenovirus (ChAd) has been developed as a vector due to concern at human adenoviruses’ pre-existing immunity and immunological potency [121, 174]. The vaccine involving a viral vector derived from serotype 63 ChAd (ChAd63) and modified vaccinia virus Ankara (MVA) has been widely evaluated in humans; it has been seen to be safe and a potent CD8+ T-cell and Ab inducer [116, 175, 176].

This review has thus described the great amount of knowledge accumulated to date whilst awaiting clinical phase results for the candidates described here, together with researchers’ other alternatives still being developed, as well as the difficulties and challenges still to be overcome as part of this long but fruitful way of developing vaccines. The target disease has been malaria, having a high global impact but, ideally, any approach demonstrating favourable results could be used regarding many other infectious diseases afflicting humanity.

Availability of data and materials

All data mentioned in this study are available in the referenced papers.

Abbreviations

aa:

Amino acid

Abs:

Antibodies

AE:

Adverse events

ALF:

Army Liposome Formulation

ALT:

Alanine aminotransferase

AS:

Adjuvant system

Chad63:

Chimpanzee adenovirus 63

CHMI:

Controlled human malaria infection

CPS-CQ:

Chemo-prophylactic cover of cloroquine

CPS-MQ:

Chemo-prophylactic cover of mefloquine

CSP:

Circumsporozoite protein

CTD:

C-terminal domain

EMA:

European Medicines Agency

EPI:

Expanded Programme on Immunization

GAP:

Genetically-attenuated parasite

GPI:

Glycosylphosphatidylinositol

HBsAg:

Hepatitis B surface antigen

HLA:

Human leukocyte antigen

HSPG:

High heparan sulphate proteoglycan

ID:

Intradermal

IFN-ɣ:

Interferon gamma

IgG:

Immunoglobulin G

IR:

Immune response

IV:

Intravenous

mAbs:

Monoclonal antibodies

ME:

Multiple epitope

MHC:

Major histocompatibility complex

MIDAS:

Metal-ion-dependent-adhesion-site

MPL:

Monophosphoryl lipid A

Mrz:

Merozoites

MVA:

Modified Vaccinia virus Ankara

NANP:

Asparagine-alanine–asparagine-proline

NMRC:

Naval Medical Research Center

NPDP:

Asparagine-proline-aspartate-proline

NVDP:

Asparagine-valine-aspartate-proline

PADRE:

Pan-DR T-helper epitope

PCR:

Polymerase chain reaction

PfSPZ:

P. falciparum Spz

RAS:

Radiation-attenuated Spz

SC:

Subcutaneous

SFC:

Spot-forming cell

SLARP:

Liver stage asparagine-rich protein

Spz:

Sporozoite

TRAP:

Thrombospondin-related adhesion protein

VLP:

Virus-like particle

vp:

Viral particle

WHO:

World Health Organization

References

  1. Dayananda K, Achur R, Gowda DC. Epidemiology, drug resistance, and pathophysiology of Plasmodium vivax malaria. J Vector Borne Dis. 2018;55:1–8.

    Article  PubMed  Google Scholar 

  2. WHO. World malaria report 2018. Geneva: World Health Organization; 2018.

    Google Scholar 

  3. Cox FE. History of the discovery of the malaria parasites and their vectors. Parasit Vectors. 2010;3:5.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Patarroyo ME, Bermúdez A, Alba MP, Vanegas M, Moreno-Vranich A, Poloche LA, et al. IMPIPS: the immune protection-inducing protein structure concept in the search for steric-electron and topochemical principles for complete fully-protective chemically synthesised vaccine development. PLoS ONE. 2015;10:e0123249.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Hopp CS, Kanatani S, Archer NK, Miller RJ, Liu H, Chiou K, et al. Quantitative intravital imaging of Plasmodium falciparum sporozoites: A novel platform to test malaria intervention strategies. Microbiology. 2019. https://doi.org/10.1101/716878.

    Article  Google Scholar 

  6. Rosenberg R, Wirtz RA, Schneider I, Burge R. An estimation of the number of malaria sporozoites ejected by a feeding mosquito. Trans R Soc Trop Med Hyg. 1990;84:209–12.

    Article  CAS  PubMed  Google Scholar 

  7. Gaur D, Mayer DCG, Miller LH. Parasite ligand-host receptor interactions during invasion of erythrocytes by Plasmodium merozoites. Int J Parasitol. 2004;34:1413–29.

    Article  CAS  PubMed  Google Scholar 

  8. Beeson JG, Drew DR, Boyle MJ, Feng G, Fowkes FJI, Richards JS. Merozoite surface proteins in red blood cell invasion, immunity and vaccines against malaria. FEMS Microbiol Rev. 2016;40:343–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. WHO. The Rainbow Tables. Geneva: World Health Organization; 2017.

    Google Scholar 

  10. Vanderberg JP, Frevert U. Intravital microscopy demonstrating antibody-mediated immobilisation of Plasmodium berghei sporozoites injected into skin by mosquitoes. Int J Parasitol. 2004;34:991–6.

    Article  PubMed  Google Scholar 

  11. Goh YS, McGuire D, Rénia L. Vaccination with sporozoites: models and correlates of protection. Front Immunol. 2019;10:1227.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Coelho CH, Doritchamou JYA, Zaidi I, Duffy PE. Advances in malaria vaccine development: report from the 2017 malaria vaccine symposium. Npj Vaccines. 2017;2:34.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Epstein JE, Tewari K, Lyke KE, Sim BKL, Billingsley PF, Laurens MB, et al. Live attenuated malaria vaccine designed to protect through hepatic CD8+ T cell immunity. Science. 2011;334:475–80.

    Article  CAS  PubMed  Google Scholar 

  14. Delemarre BJ, van der Kaay HJ. Tropical malaria contracted the natural way in the Netherlands. Ned Tijdschr Geneeskd. 1979;123:1981–2.

    CAS  PubMed  Google Scholar 

  15. Vanderberg JP, Nussenzweig RS, Most H, Orton CG. Protective immunity produced by the injection of x-irradiated sporozoites of Plasmodium berghei. II. Effects of radiation on sporozoites. J Parasitol. 1968;54:1175–80.

    Article  CAS  PubMed  Google Scholar 

  16. Nussenzweig RS, Vanderberg JP, Most H, Orton C. Specificity of protective immunity produced by X-irradiated Plasmodium berghei sporozoites. Nature. 1969;222:488–9.

    Article  CAS  PubMed  Google Scholar 

  17. Vanderberg J, Nussenzweig R, Most H. Protective immunity produced by the injection of X-irradiated sporozoites of Plasmodium berghei. Mil Med. 1969;134:1183–90.

    Article  CAS  PubMed  Google Scholar 

  18. Nussenzweig RS, Zavala F. A malaria vaccine based on a sporozoite antigen. N Engl J Med. 1997;336:128–30.

    Article  CAS  PubMed  Google Scholar 

  19. Nussenzweig V, Nussenzweig RS. Rationale for the development of an engineered sporozoite malaria vaccine. Adv Immunol. 1989;45:283–334.

    Article  CAS  PubMed  Google Scholar 

  20. Hoffman SL, Goh LML, 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;185:1155–64.

    Article  PubMed  Google Scholar 

  21. Walgate R. Quest for malaria vaccine revs up, but much work remains. Bull World Health Organ. 2001;79:1002–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Richie TL, Billingsley PF, Sim BKL, James ER, Chakravarty S, Epstein JE, et al. Progress with Plasmodium falciparum sporozoite (PfSPZ)-based malaria vaccines. Vaccine. 2015;33:7452–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lyke KE, Ishizuka AS, Berry AA, Chakravarty S, DeZure A, Enama ME, et al. Attenuated PfSPZ Vaccine induces strain-transcending T cells and durable protection against heterologous controlled human malaria infection. Proc Natl Acad Sci USA. 2017;114:2711–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sissoko MS, Healy SA, Katile A, Omaswa F, Zaidi I, Gabriel EE, et al. Safety and efficacy of PfSPZ vaccine against Plasmodium falciparum via direct venous inoculation in healthy malaria-exposed adults in Mali: a randomised, double-blind phase 1 trial. Lancet Infect Dis. 2017;17:498–509.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jongo SA, Shekalage SA, Church LWP, Ruben AJ, Schindler T, Zenklusen I, et al. Safety, immunogenicity, and protective efficacy against controlled human malaria infection of Plasmodium falciparum sporozoites vaccine in tanzanian adults. Am J Trop Med Hyg. 2018;99:338–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mordmüller B, Supan C, Sim KL, Gómez-Pérez GP, Ospina Salazar CL, Held J, et al. Direct venous inoculation of Plasmodium falciparum sporozoites for controlled human malaria infection: a dose-finding trial in two centres. Malar J. 2015;14:117.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Epstein JE, Paolino KM, Richie TL, Sedegah M, Singer A, Ruben AJ, et al. Protection against Plasmodium falciparum malaria by PfSPZ vaccine. JCI Insight. 2017;2:e89154.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Jongo SA, Church LWP, Mtoro AT, Chakravarty S, Ruben AJ, Swanson PA, et al. Safety and differential antibody and T-cell responses to the Plasmodium falciparum sporozoite malaria vaccine, PfSPZ vaccine, by age in tanzanian adults, adolescents, children, and infants. Am J Trop Med Hyg. 2019;100:1433–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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.

    Article  CAS  PubMed  Google Scholar 

  30. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJF, van Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361:468–77.

    Article  CAS  PubMed  Google Scholar 

  31. Bijker EM, Schats R, Obiero JM, Behet MC, van Gemert G-J, van de Vegte-Bolmer M, et al. Sporozoite immunization of human volunteers under mefloquine prophylaxis is safe, immunogenic and protective: a double-blind randomized controlled clinical trial. PLoS ONE. 2014;9:e112910.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Mordmüller B, Surat G, Lagler H, Chakravarty S, Ishizuka AS, Lalremruata A, et al. Sterile protection against human malaria by chemoattenuated PfSPZ vaccine. Nature. 2017;542:445–9.

    Article  PubMed  CAS  Google Scholar 

  33. Vaughan A, Wang R, Kappe SHI. Genetically engineered, attenuated whole-cell vaccine approaches for malaria. Hum Vaccin. 2010;6:107–13.

    Article  CAS  PubMed  Google Scholar 

  34. van Dijk MR, Douradinha B, Franke-Fayard B, Heussler V, van Dooren MW, van Schaijk B, et al. Genetically attenuated, P36p-deficient malarial sporozoites induce protective immunity and apoptosis of infected liver cells. Proc Natl Acad Sci USA. 2005;102:12194–9.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  35. Spring M, Murphy J, Nielsen R, Dowler M, Bennett JW, Zarling S, et al. First-in-human evaluation of genetically attenuated Plasmodium falciparum sporozoites administered by bite of Anopheles mosquitoes to adult volunteers. Vaccine. 2013;31:4975–83.

    Article  PubMed  Google Scholar 

  36. Kublin JG, Mikolajczak SA, Sack BK, Fishbaugher ME, Seilie A, Shelton L, et al. Complete attenuation of genetically engineered Plasmodium falciparum sporozoites in human subjects. Sci Transl Med. 2017;9:eaad9099.

    Article  PubMed  CAS  Google Scholar 

  37. van Schaijk BCL, Ploemen IHJ, Annoura T, Vos MW, Foquet L, van Gemert G-J, et al. A genetically attenuated malaria vaccine candidate based on P. falciparum b9/slarp gene-deficient sporozoites. eLife. 2014;3:e03582.

    Article  PubMed Central  Google Scholar 

  38. Oyen D, Torres JL, Wille-Reece U, Ockenhouse CF, Emerling D, Glanville J, et al. Structural basis for antibody recognition of the NANP repeats in Plasmodium falciparum circumsporozoite protein. Proc Natl Acad Sci USA. 2017;114:E10438–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Fisher CR, Sutton HJ, Kaczmarski JA, McNamara HA, Clifton B, Mitchell J, et al. T-dependent B cell responses to Plasmodium induce antibodies that form a high-avidity multivalent complex with the circumsporozoite protein. PLoS Pathog. 2017;13:e1006469.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Foquet L, Hermsen CC, van Gemert G-J, Van Braeckel E, Weening KE, Sauerwein R, et al. Vaccine-induced monoclonal antibodies targeting circumsporozoite protein prevent Plasmodium falciparum infection. J Clin Invest. 2014;124:140–4.

    Article  CAS  PubMed  Google Scholar 

  41. Scally SW, Julien J-P. Peek-peak-pique: repeating motifs of subtle variance are targets for potent malaria antibodies. Immunity. 2018;48:851–4.

    Article  CAS  PubMed  Google Scholar 

  42. Kisalu NK, Idris AH, Weidle C, Flores-Garcia Y, Flynn BJ, Sack BK, et al. A human monoclonal antibody prevents malaria infection by targeting a new site of vulnerability on the parasite. Nat Med. 2018;24:408–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tan J, Sack BK, Oyen D, Zenklusen I, Piccoli L, Barbieri S, et al. A public antibody lineage that potently inhibits malaria infection through dual binding to the circumsporozoite protein. Nat Med. 2018;24:401–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. WHO Expert Committee on Biological Standardization. Annex 3: guidelines on the quality, safety and efficacy of recombinant malaria vaccines targeting the preerythrocytic and blood stages of Plasmodium falciparum. Geneva: World Health Organization; 2014.

    Google Scholar 

  45. Nascimento IP, Leite LC. Recombinant vaccines and the development of new vaccine strategies. Braz J Med Biol Res. 2012;45:1102–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lemaire D, Barbosa T, Rihet P. Coping with genetic diversity: the contribution of pathogen and human genomics to modern vaccinology. Braz J Med Biol Res. 2012;45:376–85.

    Article  CAS  PubMed  Google Scholar 

  47. Ouattara A, Barry AE, Dutta S, Remarque EJ, Beeson JG, Plowe CV. Designing malaria vaccines to circumvent antigen variability. Vaccine. 2015;33:7506–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Cohen J, Nussenzweig V, Vekemans J, Leach A. From the circumsporozoite protein to the RTS,S/AS candidate vaccine. Hum Vaccin. 2010;6:90–6.

    Article  CAS  PubMed  Google Scholar 

  49. Stoute J, Heppner DGJ, Manson C, Siangla J, Opollo M, Kester KE, et al. Phase 1 safety and immunogenicity trial of malaria vaccine RTS,S/ASO2A in adults in a hyperendemic region of western Kenya. Am J Trop Med Hyg. 2006;75:166–70.

    Article  PubMed  Google Scholar 

  50. Ansong D, Asante KP, Vekemans J, Owusu SK, Owusu R, Brobby NAW, et al. T-cell responses to the RTS,S/AS01E and RTS,S/AS02D malaria candidate vaccines administered according to different schedules to Ghanaian children. PLoS ONE. 2011;6:e18891.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Owusu-Agyei S, Ansong D, Asante K, Kwarteng Owusu S, Owusu R, Wireko Brobby NA, et al. Randomized controlled trial of RTS,S/AS02D and RTS,S/AS01E malaria candidate vaccines given according to different schedules in Ghanaian children. PLoS ONE. 2009;4:e7302.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Agnandji ST, Fendel R, Mestré M, Janssens M, Vekemans J, Held J, et al. Induction of Plasmodium falciparum-specific CD4+ T cells and memory B cells in Gabonese children vaccinated with RTS,S/AS01E and RTS,S/AS02D. PLoS ONE. 2011;6:e18559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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.

    Article  CAS  PubMed  Google Scholar 

  54. Aide P, Dobaño C, Sacarlal J, Aponte JJ, Mandomando I, Guinovart C, et al. Four year immunogenicity of the RTS,S/AS02A malaria vaccine in Mozambican children during a phase IIb trial. Vaccine. 2011;29:6059–67.

    Article  PubMed  Google Scholar 

  55. Bojang K, Milligan P, Pinder M, Doherty T, Leach A, Ofori-Anyinam O, et al. Five year safety and immunogenicity of GlaxoSmithKline’s candidate malaria vaccine RTS,S/AS02 following administration to semi-immune adult men living in a malaria-endemic region of The Gambia. Hum Vaccin. 2009;5:242–7.

    Article  CAS  PubMed  Google Scholar 

  56. Kester KE, Cummings JF, Ofori-Anyinam O, Ockenhouse CF, Krzych U, Moris P, et al. Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. J Infect Dis. 2009;200:337–46.

    Article  CAS  PubMed  Google Scholar 

  57. Olotu A, Moris P, Mwacharo J, Vekemans J, Kimani D, Janssens M, et al. Circumsporozoite-specific T cell responses in children vaccinated with RTS,S/AS01E and protection against P falciparum clinical malaria. PLoS ONE. 2011;6:e25786.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Olotu A, Clement F, Jongert E, Vekemans J, Njuguna P, Ndungu FM, et al. Avidity of anti-circumsporozoite antibodies following vaccination with RTS,S/AS01E in young children. PLoS ONE. 2014;9:e115126.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Agnandji ST, Fernandes JF, Bache EB, Ramharter M. Clinical development of RTS,S/AS malaria vaccine: a systematic review of clinical Phase I-III trials. Future Microbiol. 2015;10:1553–78.

    Article  CAS  PubMed  Google Scholar 

  60. Alloueche A, Milligan P, Conway D, Pinder M, Bojang K, Doherty T, et al. Protective efficacy of the RTS,S/ASO2 Plasmodium falciparum malaria vaccines is not strain specific. Am J Trop Med Hyg. 2003;68:97–101.

    Article  PubMed  Google Scholar 

  61. Bojang KA, Milligan PJM, 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;358:1927–34.

    Article  CAS  PubMed  Google Scholar 

  62. Kester KE, McKinney DA, Tornieporth N, Ockenhouse CF, Heppner DG, Hall T, et al. Efficacy of recombinant circumsporozoite protein vaccine regimens against experimental Plasmodium falciparum malaria. J Infect Dis. 2001;183:640–7.

    Article  CAS  PubMed  Google Scholar 

  63. Greenwood BM, Holland CA, Cohen J, Momin P, Pinder M, Ballou WR, et al. A phase I safety and immunogenicity trial with the candidate malaria vaccine RTS,S/SBAS2 in semi-immune adults in The Gambia. Am J Trop Med Hyg. 1999;61:865–8.

    Article  PubMed  Google Scholar 

  64. Garçon N, Heppner DG, Cohen J. Development of RTS,S/AS02: a purified subunit-based malaria vaccine candidate formulated with a novel adjuvant. Expert Rev Vaccines. 2003;2:231–8.

    Article  PubMed  Google Scholar 

  65. Aponte JJ, Aide P, Renom M, Mandomando I, Bassat Q, Sacarlal J, et al. Safety of the RTS,S/AS02D candidate malaria vaccine in infants living in a highly endemic area of Mozambique: a double blind randomised controlled phase I/IIb trial. Lancet. 2007;370:1543–51.

    Article  CAS  PubMed  Google Scholar 

  66. 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;23:4148–57.

    Article  CAS  PubMed  Google Scholar 

  67. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. 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 falciparum disease in Mozambican children: single-blind extended follow-up of a randomised controlled trial. Lancet. 2005;366:2012–8.

    Article  CAS  PubMed  Google Scholar 

  69. Sacarlal J, Aide P, Aponte JJ, Renom M, Leach A, Mandomando I, et al. Long-term safety and efficacy of the RTS,S/AS02A malaria vaccine in Mozambican children. J Infect Dis. 2009;200:329–36.

    Article  PubMed  Google Scholar 

  70. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Bejon P, White MT, Olotu A, Bojang K, Lusingu JP, Salim N, et al. Efficacy of RTS,S malaria vaccines: individual-participant pooled analysis of phase 2 data. Lancet Infect Dis. 2013;13:319–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lell B, Agnandji S, von Glasenapp I, Haertle S, Oyakhiromen S, Issifou S, et al. A Randomized trial assessing the safety and immunogenicity of AS01 and AS02 adjuvanted RTS,S malaria vaccine candidates in children in Gabon. PLoS ONE. 2009;4:e7611.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. 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.

    Article  CAS  PubMed  Google Scholar 

  74. 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;359:2533–44.

    Article  CAS  PubMed  Google Scholar 

  75. Regules JA, Cicatelli SB, Bennett JW, Paolino KM, Twomey PS, Moon JE, et al. Fractional third and fourth dose of RTS,S/AS01 malaria candidate vaccine: a phase 2a controlled human malaria parasite infection and immunogenicity study. J Infect Dis. 2016;214:762–71.

    Article  CAS  PubMed  Google Scholar 

  76. Chaudhury S, Regules JA, Darko CA, Dutta S, Wallqvist A, Waters NC, et al. Delayed fractional dose regimen of the RTS,S/AS01 malaria vaccine candidate enhances an IgG4 response that inhibits serum opsonophagocytosis. Sci Rep. 2017;7:7998.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. The RTS,S Clinical Trials Partnership. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N Engl J Med. 2011;365:1863–75.

    Article  Google Scholar 

  78. The RTS,S Clinical Trials Partnership. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet. 2015;386:31–45.

    Article  CAS  Google Scholar 

  79. Olotu A, Fegan G, Wambua J, Nyangweso G, Leach A, Lievens M, et al. Seven-year efficacy of RTS,S/AS01 malaria vaccine among young African children. N Engl J Med. 2016;374:2519–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. The RTS,S Clinical Trials Partnership. Efficacy and safety of the RTS,S/AS01 malaria vaccine during 18 months after vaccination: a phase 3 randomized, controlled trial in children and young infants at 11 African sites. PLoS Med. 2014;11:e1001685.

    Article  Google Scholar 

  81. The RTS,S Clinical Trials Partnership. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med. 2012;367:2284–95.

    Article  CAS  Google Scholar 

  82. Tinto H, Otieno W, Gesase S, Sorgho H, Otieno L, Liheluka E, et al. Long-term incidence of severe malaria following RTS,S/AS01 vaccination in children and infants in Africa: an open-label 3-year extension study of a phase 3 randomised controlled trial. Lancet Infect Dis. 2019;19:821–32.

    Article  PubMed  Google Scholar 

  83. WHO. Malaria vaccine: WHO position paper 2016. Geneva: World Health Organization; 2016.

    Google Scholar 

  84. WHO. Malaria vaccines: position paper. SAGE/MPAC evidence to recommendations table on the use of malaria vaccines 2016. Geneva: World Health Organization; 2016.

    Google Scholar 

  85. Weedall GD, Preston BMJ, Thomas AW, Sutherland CJ, Conway DJ. Differential evidence of natural selection on two leading sporozoite stage malaria vaccine candidate antigens. Int J Parasitol. 2007;37:77–85.

    Article  CAS  PubMed  Google Scholar 

  86. Chenet SM, Branch OH, Escalante AA, Lucas CM, Bacon DJ. Genetic diversity of vaccine candidate antigens in Plasmodium falciparum isolates from the Amazon basin of Peru. Malar J. 2008;7:93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Gandhi K, Thera MA, Coulibaly D, Traoré K, Guindo AB, Ouattara A, et al. Variation in the circumsporozoite protein of Plasmodium falciparum: vaccine development implications. PLoS ONE. 2014;9:e101783.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Zeeshan M, Alam MT, Vinayak S, Bora H, Tyagi RK, Alam MS, et al. Genetic variation in the Plasmodium falciparum circumsporozoite protein in India and its relevance to RTS,S malaria vaccine. PLoS ONE. 2012;7:e43430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Marty-Roix R, Vladimer GI, Pouliot K, Weng D, Buglione-Corbett R, West K, et al. Identification of QS-21 as an inflammasome-activating molecular component of saponin adjuvants. J Biol Chem. 2016;291:1123–36.

    Article  CAS  PubMed  Google Scholar 

  90. Fernández-Tejada A, Chea EK, George C, Pillarsetty N, Gardner JR, Livingston PO, et al. Development of a minimal saponin vaccine adjuvant based on QS-21. Nat Chem. 2014;6:635–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Kaslow DC, Biernaux S. RTS,S: toward a first landmark on the Malaria Vaccine Technology Roadmap. Vaccine. 2015;33:7425–32.

    Article  PubMed  Google Scholar 

  92. Chaudhury S, Ockenhouse CF, Regules JA, Dutta S, Wallqvist A, Jongert E, et al. The biological function of antibodies induced by the RTS,S/AS01 malaria vaccine candidate is determined by their fine specificity. Malar J. 2016;15:301.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Kurup SP, Butler NS, Harty JT. T cell-mediated immunity to malaria. Nat Rev Immunol. 2019;19:457–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Collins KA, Snaith R, Cottingham MG, Gilbert SC, Hill AVS. Enhancing protective immunity to malaria with a highly immunogenic virus-like particle vaccine. Sci Rep. 2017;7:46621.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801.

    Article  CAS  PubMed  Google Scholar 

  96. Ewer KJ, Sierra-Davidson K, Salman AM, Illingworth JJ, Draper SJ, Biswas S, et al. Progress with viral vectored malaria vaccines: a multi-stage approach involving “unnatural immunity”. Vaccine. 2015;33:7444–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Rodrigues M, Li S, Murata K, Rodriguez D, Rodriguez JR, Bacik I, et al. Influenza and vaccinia viruses expressing malaria CD8+ T and B cell epitopes Comparison of their immunogenicity and capacity to induce protective immunity. J Immunol Baltim Md. 1950;1994(153):4636–48.

    Google Scholar 

  98. Tine JA, Lanar DE, Smith DM, Wellde BT, Schultheiss P, Ware LA, et al. NYVAC-Pf7: a poxvirus-vectored, multiantigen, multistage vaccine candidate for Plasmodium falciparum malaria. Infect Immun. 1996;64:3833–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Tsuji M, Bergmann CC, Takita-Sonoda Y, Murata K, Rodrigues EG, Nussenzweig RS, et al. Recombinant Sindbis viruses expressing a cytotoxic T-lymphocyte epitope of a malaria parasite or of influenza virus elicit protection against the corresponding pathogen in mice. J Virol. 1998;72:6907–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Hansen SG, Womack J, Scholz I, Renner A, Edgel KA, Xu G, et al. Cytomegalovirus vectors expressing Plasmodium knowlesi antigens induce immune responses that delay parasitemia upon sporozoite challenge. PLoS ONE. 2019;14:e0210252.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Sadoff J, Ballou W, Baron L, Majarian W, Brey R, Hockmeyer W, et al. Oral Salmonella typhimurium vaccine expressing circumsporozoite protein protects against malaria. Science. 1988;240:336–8.

    Article  CAS  PubMed  Google Scholar 

  102. Aggarwal A, Kumar S, Jaffe R, Hone D, Gross M, Sadoff J. Oral Salmonella: malaria circumsporozoite recombinants induce specific CD8+ cytotoxic T cells. J Exp Med. 1990;172:1083–90.

    Article  CAS  PubMed  Google Scholar 

  103. Arama C, Assefaw-Redda Y, Rodriguez A, Fernández C, Corradin G, Kaufmann SHE, et al. Heterologous prime-boost regimen adenovector 35-circumsporozoite protein vaccine/recombinant Bacillus Calmette-Guérin expressing the Plasmodium falciparum circumsporozoite induces enhanced long-term memory immunity in BALB/c mice. Vaccine. 2012;30:4040–5.

    Article  CAS  PubMed  Google Scholar 

  104. Bergmann-Leitner ES, Hosie H, Trichilo J, DeRiso E, Ranallo RT, Alefantis T, et al. Self-adjuvanting bacterial vectors expressing pre-erythrocytic antigens induce sterile protection against malaria. Front Immunol. 2013;4:176.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Wang H, Rogers W, Kang Y, Sedegah M, Hoffman S. Partial protection against malaria by immunization with Leishmania enriettii expressing the Plasmodium yoelii circumsporozoite protein. Mol Biochem Parasitol. 1995;69:139–48.

    Article  CAS  PubMed  Google Scholar 

  106. Di Cristina M, Ghouze F, Kocken CH, Naitza S, Cellini P, Soldati D, et al. Transformed Toxoplasma gondii tachyzoites expressing the circumsporozoite protein of Plasmodium knowlesi elicit a specific immune response in rhesus monkeys. Infect Immun. 1999;67:1677–82.

    PubMed  PubMed Central  Google Scholar 

  107. Charest H, Sedegah M, Yap GS, Gazzinelli RT, Caspar P, Hoffman SL, et al. Recombinant attenuated Toxoplasma gondii expressing the Plasmodium yoelii circumsporozoite protein provides highly effective priming for CD8+ T cell-dependent protective immunity against malaria. J Immunol. 2000;165:2084–92.

    Article  CAS  PubMed  Google Scholar 

  108. Ura T, Okuda K, Shimada M. Developments in viral vector-based vaccines. Vaccines. 2014;2:624–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Gilbert SC, Plebanski M, Harris SJ, Allsopp CEM, Thomas R, Layton GT, et al. A protein particle vaccine containing multiple malaria epitopes. Nat Biotechnol. 1997;15:1280–4.

    Article  CAS  PubMed  Google Scholar 

  110. Afolabi MO, Tiono AB, Adetifa UJ, Yaro JB, Drammeh A, Nébié I, et al. Safety and immunogenicity of ChAd63 and MVA ME-TRAP in West African children and infants. Mol Ther. 2016;24:1470–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Ogwang C, Afolabi M, Kimani D, Jagne YJ, Sheehy SH, Bliss CM, et al. Safety and immunogenicity of heterologous prime-boost Immunisation with Plasmodium falciparum malaria candidate vaccines, ChAd63 ME-TRAP and MVA ME-TRAP, in healthy Gambian and Kenyan adults. PLoS ONE. 2013;8:e57726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Swadling L, Capone S, Antrobus RD, Brown A, Richardson R, Newell EW, et al. A human vaccine strategy based on chimpanzee adenoviral and MVA vectors that primes, boosts, and sustains functional HCV-specific T cell memory. Sci Transl Med. 2014;6:261ra153.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Xiang Z, Li Y, Cun A, Yang W, Ellenberg S, Switzer WM, et al. Chimpanzee Adenovirus antibodies in humans, sub-Saharan Africa. Emerg Infect Dis. 2006;12:1596–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Patarroyo ME, Arévalo-Pinzón G, Reyes C, Moreno-Vranich A, Patarroyo MA. Malaria parasite survival depends on conserved binding peptides’ critical biological functions. Curr Issues Mol Biol. 2016;18:57–8.

    PubMed  Google Scholar 

  115. Spencer AJ, Cottingham MG, Jenks JA, Longley RJ, Capone S, Colloca S, et al. Enhanced vaccine-induced CD8+ T cell responses to malaria antigen ME-TRAP by fusion to MHC Class II invariant chain. PLoS ONE. 2014;9:e100538.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. O’Hara GA, Duncan CJA, Ewer KJ, Collins KA, Elias SC, Halstead FD, et al. Clinical assessment of a recombinant simian adenovirus ChAd63: a potent new vaccine vector. J Infect Dis. 2012;205:772–81.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Mensah VA, Gueye A, Ndiaye M, Edwards NJ, Wright D, Anagnostou NA, et al. Safety, immunogenicity and efficacy of prime-boost vaccination with ChAd63 and MVA encoding ME-TRAP against Plasmodium falciparum infection in adults in Senegal. PLoS ONE. 2016;11:e0167951.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Ogwang C, Kimani D, Edwards NJ, Roberts R, Mwacharo J, Bowyer G, et al. Prime-boost vaccination with chimpanzee adenovirus and modified vaccinia Ankara encoding TRAP provides partial protection against Plasmodium falciparum infection in Kenyan adults. Sci Transl Med. 2015;7:286re5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Webster DP, Dunachie S, McConkey S, Poulton I, Moore AC, Walther M, et al. Safety of recombinant fowlpox strain FP9 and modified vaccinia virus Ankara vaccines against liver-stage P. falciparum malaria in non-immune volunteers. Vaccine. 2006;24:3026–34.

    Article  CAS  PubMed  Google Scholar 

  120. de Barra E, Hodgson SH, Ewer KJ, Bliss CM, Hennigan K, Collins A, et al. A Phase Ia study to assess the safety and immunogenicity of new malaria vaccine candidates ChAd63 CS administered alone and with MVA CS. PLoS ONE. 2014;9:e115161.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Hodgson SH, Ewer KJ, Bliss CM, Edwards NJ, Rampling T, Anagnostou NA, et al. Evaluation of the efficacy of ChAd63-MVA vectored vaccines expressing circumsporozoite protein and ME-TRAP against controlled human malaria infection in malaria-naive individuals. J Infect Dis. 2015;211:1076–86.

    Article  CAS  PubMed  Google Scholar 

  122. Lövgren Bengtsson K, Morein B, Osterhaus AD. ISCOM technology-based Matrix M™ adjuvant: success in future vaccines relies on formulation. Expert Rev Vaccines. 2011;10:401–3.

    Article  PubMed  Google Scholar 

  123. Venkatraman N, Anagnostou N, Bliss C, Bowyer G, Wright D, Lövgren-Bengtsson K, et al. Safety and immunogenicity of heterologous prime-boost immunization with viral-vectored malaria vaccines adjuvanted with Matrix-M™. Vaccine. 2017;35:6208–17.

    Article  CAS  PubMed  Google Scholar 

  124. Mamo T, Poland GA. Nanovaccinology: the next generation of vaccines meets 21st century materials science and engineering. Vaccine. 2012;30:6609–11.

    Article  CAS  PubMed  Google Scholar 

  125. Couvreur P, Vauthier C. Nanotechnology: intelligent design to treat complex disease. Pharm Res. 2006;23:1417–50.

    Article  CAS  PubMed  Google Scholar 

  126. Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. FASEB J. 2005;19:311–30.

    Article  CAS  PubMed  Google Scholar 

  127. Treuel L, Jiang X, Nienhaus GU. New views on cellular uptake and trafficking of manufactured nanoparticles. J R Soc Interface. 2013;10:20120939.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Roldão A, Mellado MC, Castilho LR, Carrondo MJ, Alves PM. Virus-like particles in vaccine development. Expert Rev Vaccines. 2010;9:1149–76.

    Article  PubMed  Google Scholar 

  129. Correia-Pinto JF, Csaba N, Alonso MJ. Vaccine delivery carriers: insights and future perspectives. Int J Pharm. 2013;440:27–38.

    Article  CAS  PubMed  Google Scholar 

  130. Kushnir N, Streatfield SJ, Yusibov V. Virus-like particles as a highly efficient vaccine platform: diversity of targets and production systems and advances in clinical development. Vaccine. 2012;31:58–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Plummer EM, Manchester M. Viral nanoparticles and virus-like particles: platforms for contemporary vaccine design: platforms for contemporary vaccine design. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2011;3:174–96.

    Article  CAS  PubMed  Google Scholar 

  132. Bolhassani A, Safaiyan S, Rafati S. Improvement of different vaccine delivery systems for cancer therapy. Mol Cancer. 2011;10:3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Krishnamachari Y, Geary SM, Lemke CD, Salem AK. Nanoparticle delivery systems in cancer vaccines. Pharm Res. 2011;28:215–36.

    Article  CAS  PubMed  Google Scholar 

  134. Hamdy S, Haddadi A, Hung RW, Lavasanifar A. Targeting dendritic cells with nano-particulate PLGA cancer vaccine formulations. Adv Drug Deliv Rev. 2011;63:943–55.

    Article  CAS  PubMed  Google Scholar 

  135. Chackerian B. Virus-like particle based vaccines for Alzheimer disease. Hum Vaccin. 2010;6:926–30.

    Article  CAS  PubMed  Google Scholar 

  136. Tissot AC, Maurer P, Nussberger J, Sabat R, Pfister T, Ignatenko S, et al. Effect of immunisation against angiotensin II with CYT006-AngQb on ambulatory blood pressure: a double-blind, randomised, placebo-controlled phase IIa study. Lancet. 2008;371:821–7.

    Article  CAS  PubMed  Google Scholar 

  137. Maurer P, Jennings GT, Willers J, Rohner F, Lindman Y, Roubicek K, et al. A therapeutic vaccine for nicotine dependence: preclinical efficacy, and phase I safety and immunogenicity. Eur J Immunol. 2005;35:2031–40.

    Article  CAS  PubMed  Google Scholar 

  138. Richards RL, Hayre MD, Hockmeyer WT, Alving CR. Liposomes, lipid A, and aluminum hydroxide enhance the immune response to a synthetic malaria sporozoite antigen. Infect Immun. 1988;56:682–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. van Rooijen N, van Nieuwmegen R. Liposomes in immunology: multilamellar phosphatidylcholine liposomes as a simple, biodegradable and harmless adjuvant without any immunogenic activity of its own. Immunol Commun. 1980;9:243–56.

    Article  PubMed  Google Scholar 

  140. Raman S, Machaidze G, Lustig A, Aebi U, Burkhard P. Structure-based design of peptides that self-assemble into regular polyhedral nanoparticles. Nanomedicine. 2006;2:95–102.

    Article  CAS  PubMed  Google Scholar 

  141. Schroeder U, Graff A, Buchmeier S, Rigler P, Silvan U, Tropel D, et al. Peptide nanoparticles serve as a powerful platform for the immunogenic display of poorly antigenic actin determinants. J Mol Biol. 2009;386:1368–81.

    Article  CAS  PubMed  Google Scholar 

  142. Wahome N, Pfeiffer T, Ambiel I, Yang Y, Keppler OT, Bosch V, et al. Conformation-specific display of 4E10 and 2F5 epitopes on self-assembling protein nanoparticles as a potential HIV vaccine. Chem Biol Drug Des. 2012;80:349–57.

    Article  CAS  PubMed  Google Scholar 

  143. El Bissati K, Zhou Y, Dasgupta D, Cobb D, Dubey JP, Burkhard P, et al. Effectiveness of a novel immunogenic nanoparticle platform for Toxoplasma peptide vaccine in HLA transgenic mice. Vaccine. 2014;32:3243–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. El Bissati K, Chentoufi AA, Krishack PA, Zhou Y, Woods S, Dubey JP, et al. Adjuvanted multi-epitope vaccines protect HLA-A*11:01 transgenic mice against Toxoplasma gondii. JCI Insight. 2016;1:e85955.

    Article  PubMed  PubMed Central  Google Scholar 

  145. El Bissati K, Zhou Y, Paulillo SM, Raman SK, Karch CP, Roberts CW, et al. Protein nanovaccine confers robust immunity against Toxoplasma. Npj Vaccines. 2017;2:24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Pimentel TAPF, Yan Z, Jeffers SA, Holmes KV, Hodges RS, Burkhard P. Peptide nanoparticles as novel immunogens: design and analysis of a prototypic severe acute respiratory syndrome vaccine. Chem Biol Drug Des. 2009;73:53–61.

    Article  CAS  PubMed  Google Scholar 

  147. Babapoor S, Neef T, Mittelholzer C, Girshick T, Garmendia A, Shang H, et al. A novel vaccine using nanoparticle platform to present immunogenic M2e against avian influenza infection. Influenza Res Treat. 2011;2011:126794.

    PubMed  Google Scholar 

  148. Kaba SA, Brando C, Guo Q, Mittelholzer C, Raman S, Tropel D, et al. A Nonadjuvanted polypeptide nanoparticle vaccine confers long-lasting protection against rodent malaria. J Immunol. 2009;183:7268–77.

    Article  CAS  PubMed  Google Scholar 

  149. Kaba SA, McCoy ME, Doll TAPF, Brando C, Guo Q, Dasgupta D, et al. Protective antibody and CD8+ T-cell responses to the Plasmodium falciparum circumsporozoite protein induced by a nanoparticle vaccine. PLoS ONE. 2012;7:e48304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Guo Q, Dasgupta D, Doll TAPF, Burkhard P, Lanar DE. Expression, purification and refolding of a self-assembling protein nanoparticle (SAPN) malaria vaccine. Methods. 2013;60:242–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Kaba SA, Karch CP, Seth L, Ferlez KMB, Storme CK, Pesavento DM, et al. Self-assembling protein nanoparticles with built-in flagellin domains increases protective efficacy of a Plasmodium falciparum based vaccine. Vaccine. 2018;36:906–14.

    Article  CAS  PubMed  Google Scholar 

  152. Seth L, Bingham Ferlez KM, Kaba SA, Musser DM, Emadi S, Matyas GR, et al. Development of a self-assembling protein nanoparticle vaccine targeting Plasmodium falciparum circumsporozoite protein delivered in three army liposome formulation adjuvants. Vaccine. 2017;35:5448–54.

    Article  CAS  PubMed  Google Scholar 

  153. Powles L, Xiang S, Selomulya C, Plebanski M. The use of synthetic carriers in malaria vaccine design. Vaccines. 2015;3:894–929.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Rappuoli R. Reverse Vaccinology and genomics. Science. 2003;302:602.

    Article  CAS  PubMed  Google Scholar 

  155. Rappuoli R. Reverse vaccinology, a genome-based approach to vaccine development. Vaccine. 2001;19:2688–91.

    Article  CAS  PubMed  Google Scholar 

  156. Tuju J, Kamuyu G, Murungi LM, Osier FHA. Vaccine candidate discovery for the next generation of malaria vaccines. Immunology. 2017;152:195–206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Weiss GE, Crabb BS, Gilson PR. Overlaying molecular and temporal aspects of malaria parasite invasion. Trends Parasitol. 2016;32:284–95.

    Article  CAS  PubMed  Google Scholar 

  158. Weiss GE, Gilson PR, Taechalertpaisarn T, Tham W-H, de Jong NWM, Harvey KL, et al. Revealing the sequence and resulting cellular morphology of receptor-ligand interactions during Plasmodium falciparum invasion of erythrocytes. PLoS Pathog. 2015;11:e1004670.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Talapko J, Škrlec I, Alebić T, Jukić M, Včev A. Malaria: the past and the present. Microorganisms. 2019;7:179.

    Article  PubMed Central  Google Scholar 

  160. Zhang L, Wang W, Wang S. Effect of vaccine administration modality on immunogenicity and efficacy. Expert Rev Vaccines. 2015;14:1509–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Moser KA, Drábek EF, Dwivedi A, Crabtree J, Stucke EM, Dara A, et al. Strains used in whole organism Plasmodium falciparum vaccine trials differ in genome structure, sequence, and immunogenic potential. Genome Med. 2019;12:6.

    Article  CAS  Google Scholar 

  162. Patarroyo ME, Patarroyo MA. Emerging rules for subunit-based, multiantigenic, multistage chemically synthesized vaccines. Acc Chem Res. 2008;41:377–86.

    Article  CAS  PubMed  Google Scholar 

  163. Patarroyo ME, Bermúdez A, Patarroyo MA. Structural and immunological principles leading to chemically synthesized, multiantigenic, multistage, minimal subunit-based vaccine development. Chem Rev. 2011;111:3459–507.

    Article  CAS  PubMed  Google Scholar 

  164. Lyke KE, Fernández-Viňa MA, Cao K, Hollenbach J, Coulibaly D, Kone AK, et al. Association of HLA alleles with Plasmodium falciparum severity in Malian children. Tissue Antigens. 2011;77:562–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Matern BM, Olieslagers TI, Voorter CEM, Groeneweg M, Tilanus MGJ. Insights into the polymorphism in HLA-DRA and its evolutionary relationship with HLA Haplotypes. HLA. 2019;95:117–27.

    Article  PubMed  CAS  Google Scholar 

  166. Suárez CF, Pabón L, Barrera A, Aza-Conde J, Patarroyo MA, Patarroyo ME. Structural analysis of owl monkey MHC-DR shows that fully-protective malaria vaccine components can be readily used in humans. Biochem Biophys Res Commun. 2017;491:1062–9.

    Article  PubMed  CAS  Google Scholar 

  167. Diaz D, Naegeli M, Rodriguez R, Nino-Vasquez JJ, Moreno A, Patarroyo ME, et al. Sequence and diversity of MHC DQA and DQB genes of the owl monkey Aotus nancymaae. Immunogenetics. 2000;51:528–37.

    Article  CAS  PubMed  Google Scholar 

  168. Suárez MCF, Patarroyo MA, Patarroyo ME. Characterisation and comparative analysis of MHC-DPA1 exon 2 in the owl monkey (Aotus nancymaae). Gene. 2011;470:37–45.

    Article  CAS  Google Scholar 

  169. Cardenas PP, Suarez CF, Martinez P, Patarroyo ME, Patarroyo MA. MHC class I genes in the owl monkey: mosaic organisation, convergence and loci diversity. Immunogenetics. 2005;56:818–32.

    Article  CAS  PubMed  Google Scholar 

  170. Baquero JE, Miranda S, Murillo O, Mateus H, Trujillo E, Suarez C, et al. Reference strand conformational analysis (RSCA) is a valuable tool in identifying MHC-DRB sequences in three species of Aotus monkeys. Immunogenetics. 2006;58:590–7.

    Article  CAS  PubMed  Google Scholar 

  171. Suárez CF, Patarroyo ME, Trujillo E, Estupiñán M, Baquero JE, Parra C, et al. Owl monkey MHC-DRB exon 2 reveals high similarity with several HLA-DRB lineages. Immunogenetics. 2006;58:542–58.

    Article  PubMed  CAS  Google Scholar 

  172. Guerrero JE, Pacheco DP, Suárez CF, Martínez P, Aristizabal F, Moncada CA, et al. Characterizing T-cell receptor gamma-variable gene in Aotus nancymaae owl monkey peripheral blood. Tissue Antigens. 2003;62:472–82.

    Article  CAS  PubMed  Google Scholar 

  173. Stassijns J, Bollaerts K, Baay M, Verstraeten T. A systematic review and meta-analysis on the safety of newly adjuvanted vaccines among children. Vaccine. 2016;34:714–22.

    Article  PubMed  Google Scholar 

  174. Colloca S, Barnes E, Folgori A, Ammendola V, Capone S, Cirillo A, et al. Vaccine vectors derived from a large collection of simian adenoviruses induce potent cellular immunity across multiple species. Sci Transl Med. 2012;4:115ra2.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  175. Barnes E, Folgori A, Capone S, Swadling L, Aston S, Kurioka A, et al. Novel adenovirus-based vaccines induce broad and sustained T cell responses to HCV in Man. Sci Transl Med. 2012;4:115ra1.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Sheehy SH, Duncan CJ, Elias SC, Choudhary P, Biswas S, Halstead FD, et al. ChAd63-MVA-vectored blood-stage malaria vaccines targeting MSP1 and AMA1: assessment of efficacy against mosquito bite challenge in humans. Mol Ther. 2012;20:2355–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Doud MB, Koksal AC, Mi L-Z, Song G, Lu C, Springer TA. Unexpected fold in the circumsporozoite protein target of malaria vaccines. Proc Natl Acad Sci USA. 2012;109:7817–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Tossavainen H, Pihlajamaa T, Huttunen TK, Raulo E, Rauvala H, Permi P, et al. The layered fold of the TSR domain of P falciparum TRAP contains a heparin binding site. Protein Sci. 2006;15:1760–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank Universidad de Boyacá for its academic support and Jason Garry for translating this manuscript.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

JMF, LCC, AC, CR, MG and DRS analysed the studies and wrote the manuscript. MAP and MEP revised and wrote the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Manuel Alfonso Patarroyo or Manuel Elkin Patarroyo.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Molina-Franky, J., Cuy-Chaparro, L., Camargo, A. et al. Plasmodium falciparum pre-erythrocytic stage vaccine development. Malar J 19, 56 (2020). https://doi.org/10.1186/s12936-020-3141-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12936-020-3141-z

Keywords