Next Article in Journal
Febrile Seizures and Measles-Containing Vaccines in China: A Self-Controlled Case Series Study
Next Article in Special Issue
The Rise of Vectored Vaccines: A Legacy of the COVID-19 Global Crisis
Previous Article in Journal
Worldwide Vaccination Willingness for COVID-19: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plasmodium falciparum Malaria Vaccines and Vaccine Adjuvants

by
Srinivasa Reddy Bonam
1,*,†,
Laurent Rénia
2,3,4,*,†,
Ganesh Tadepalli
5,
Jagadeesh Bayry
1,6 and
Halmuthur Mahabalarao Sampath Kumar
5,*
1
Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, F-75006 Paris, France
2
A*STAR Infectious Diseases Labs, 8A Biomedical Grove, Singapore 138648, Singapore
3
Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
4
School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
5
Vaccine Immunology Laboratory, Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
6
Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678623, India
*
Authors to whom correspondence should be addressed.
Equally contributed.
Vaccines 2021, 9(10), 1072; https://doi.org/10.3390/vaccines9101072
Submission received: 14 August 2021 / Revised: 10 September 2021 / Accepted: 22 September 2021 / Published: 24 September 2021

Abstract

:
Malaria—a parasite vector-borne disease—is a global health problem, and Plasmodium falciparum has proven to be the deadliest among Plasmodium spp., which causes malaria in humans. Symptoms of the disease range from mild fever and shivering to hemolytic anemia and neurological dysfunctions. The spread of drug resistance and the absence of effective vaccines has made malaria disease an ever-emerging problem. Although progress has been made in understanding the host response to the parasite, various aspects of its biology in its mammalian host are still unclear. In this context, there is a pressing demand for the development of effective preventive and therapeutic strategies, including new drugs and novel adjuvanted vaccines that elicit protective immunity. The present article provides an overview of the current knowledge of anti-malarial immunity against P. falciparum and different options of vaccine candidates in development. A special emphasis has been made on the mechanism of action of clinically used vaccine adjuvants.

1. Introduction

Malaria, caused by apicomplexan Plasmodium spp., remains one of the world’s most threatening diseases of humans and other animals with high morbidity and mortality rates [1]. The recent findings of the World Health Organization (WHO) recorded 228 million cases and 405,000 deaths globally in 2018 [2]. Although eight species of Plasmodium can infect humans, most malarial cases are due to P. falciparum or P. vivax, but deaths are mostly due to falciparum malaria. The African countries carry the highest share of the global malaria burden (~90% as per 2017 WHO reports) [3]. In Asian countries, e.g., India, which accounts for 4% of the global burden, malaria is still a serious health threat [4]. As an answer, the Special Programme for Research and Training in Tropical Diseases (TDR; co-sponsored by the WHO), US National Institutes of Health, UK department for international development, Bill & Melinda Gates Foundation, and other organizations have increased funding for research and development and other control measures (including vaccination) to minimize the malaria cases [2,5]. WHO has also put more effort into implementing essential malaria commodities, such as rapid diagnostic tests, insecticide-treated mosquito nets, vector control, artemisinin-based combination therapy, and in defeating insecticide resistance of malaria vectors. These programs were highly successful and did reduce significantly the numbers of malaria-related deaths [4,6,7].
The malaria parasite has a complex life cycle (Figure 1). The Ronald Ross and Giovanni Battista Grassi legacy laid the cornerstone for understanding the malaria parasite life cycle (Figure 1) [8]. There are pre-erythrocytic and erythrocytic stages in humans (host) and the sexual life cycle in the mosquito vector. (a) Pre-erythrocytic stage. When the infected mosquito bites the host for blood meal, it injects sporozoite mostly into the dermis or sometimes directly into the blood vessels [9]. Once sporozoite reaches the liver, it invades and infects the hepatocytes. Inside the hepatocytes, sporozoites multiply extensively, generating 10–30,000 merozoites [10]. (b) Erythrocytic stage. Merozoites enter the red blood cells (RBCs) and mature into trophozoite and schizont. Each schizont generates 6–12 merozoites, which are released into the circulation (remain one minute to the next infection) and infect more RBCs [11,12]. During the process, a small number of parasites develop into male and female gametocytes. The formed gametocytes mature first in the bone marrow (I–IV stages) and then spleen (V stage) [13]. (c) Mosquito/sexual stage. In every progressing cycle, a small number of parasites deviates from asexual reproduction and develops into male and female gametocytes, which will enter the stomach of the Anopheles mosquito during the blood meal. The male and female gametocytes develop into flagellated microgametes (eight) and a single macrogamete respectively within the mosquito’s midgut. A zygote resulting from the fusion of a macrogamete and microgamete develops into ookinete via meiosis and penetrates the mosquito’s gut wall to form oocysts and produce sporozoites. These sporozoites travel to salivary glands after the rupture of oocysts and will infect a new host during a mosquito bite (Figure 1) [12,14].
P. falciparum transmission can be managed through vector control approaches, such as spraying insecticides and/or using chemically-treated mosquito nets, and through the use of antimalarial drugs for prophylaxis and radical cure [15]. Early diagnosis, use of bed nets, and timely treatment with anti-malarial compounds are the global strategies for controlling P. falciparum. However, the parasite has developed resistance to all anti-malarial drugs which are in present use [16]. In the last century, chloroquine was an essential tool to eradicate malaria in many countries [17]. However, in the 1970s resistance to chloroquine monotherapy emerged, and nowadays, chloroquine-resistant P. falciparum parasites are present worldwide. Other drugs such as pyrimethamine, mefloquine, or artemisinin derivatives were also developed and replaced chloroquine progressively. Because resistance to these drugs also developed, combination therapies were set to treat P. falciparum infections [18]. Artemisinin-based combination therapies have been essential to decrease the death toll. However, resistance to artemisinin and its partner drugs has become a severe concern in the last decade [19,20]. Thus, there is a need for novel drugs and vaccines, particularly for controlling and possibly eliminating P. falciparum malaria. The focus of this article is to review the current status of malaria vaccines research, with an emphasis of the use of adjuvants and their mechanisms of action.

2. Immunity against P. falciparum

Knowledge on the interaction between the parasite and host immune system and how clinical immunity is acquired against malarial infection is critical for the development of effective vaccines, anti-malarial drugs, and immunomodulators.

2.1. Dendritic Cells and the Initiation of the Immune Response

Among the innate immune cells, professional antigen-presenting cells (APCs), such as dendritic cells (DCs), have a prominent role in antigen presentation [21]. Although each innate immune cell has a role in homeostasis and immunity, DCs’ role in vaccine and vaccine adjuvant development has been extensively studied [22,23]. Considering the central role of DCs in the activation and polarization of naïve T cells [24] and shaping the immune response to vaccine adjuvants (Figure 2), we focused our discussion on DCs [13] in the innate immune system.
DCs appear to interact with the parasite at various anatomical locations of the host such as skin, liver, blood, and spleen. DCs recognize pathogens through specific receptors called pattern-recognition receptors (PRRs), such as toll-like receptors (TLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), and others. PRRs recognize highly conserved structures of microorganisms, including parasites, via pathogen-associated molecular patterns (PAMPs). PAMPs are processed by the DCs and present the unique sequences of PAMPs to the T helper (Th) cells, which seeds the development of immunological memory [22]. Although a few PAMPs (e.g., Plasmodium multidomain scavenger receptor-like protein [PxSR], glycosylphosphatidylinositol [GPI], hemozoin, RNA, plasmodial DNA, CpG motifs, and others) (Figure 2) [25,29,30,31] have been identified in Plasmodium spp., their interaction with DCs and the related immune defense are still not fully elucidated [32]. Interestingly, PRRs (particularly TLRs) have an essential role in both immune defense and pathogenicity (Figure 2) [33]. In addition, DCs have differential functional capabilities (tolerogenic versus inflammatory/activated) based on the tissues they reside in and signals they receive from the pathogen and microenvironment [34,35].
The malaria parasite in the mammalian host interacts with different tissues (the skin dermis, the liver, blood capillaries, and vessels) and can be either extra- or intracellular. This dictates which types of DCs it will interact with (for an in depth-review, see [35,36]).
In the dermis, the extracellular sporozoites are mostly phagocytosed by monocyte-derived macrophages (MDM) rather than by dermal DCs and monocyte-derived DCs. After sporozoites uptake, MDM display a regulatory phenotype, which negatively affects the antigen-presenting capacity of dermal APCs [37]. Nevertheless, some dermal DCs, after their interaction with sporozoites, could reach draining lymph nodes to prime CD4+ or CD8+ T cells [38,39]. Of all the sporozoites entering the blood circulation in their mammalian hosts, only a fraction can invade liver cells since most of them are retained in the spleen [40]. They can be phagocytosed by splenic DCs and presented to T and B cells. For the intracellular liver parasites, it is not clear how DCs acquire newly synthetized liver-stage antigens. It was shown that monocyte-derived DCs are responsible for CD8+ T cell priming [41]. However, the way they acquire antigen remains unknown. It has been proposed that DCs can either phagocyte apoptotic hepatocytes resulting from an aborted infection [38] or they can acquire antigens from infected hepatocytes [42]. It has long been known that intradermal immunization is less effective than the intravenous route when animals and humans have been injected with whole sporozoites [43,44]. This suggests that the priming of DCs in the spleen or liver is more potent than the skin’s priming. Since it is the natural route of infection, malaria parasites have evolved immune escape mechanisms at the skin. Identifying the adjuvants that can antagonize these mechanisms may help in the development of whole sporozoite skin immunization protocols. Of note, liver-resident DCs, which produce high amounts of IL-10, are more tolerogenic than other DC subsets. Cracking this tolerance, possibly through the use of proper adjuvants, represents one of the rational approaches to target liver-stage malaria infection [13,45,46].
During the erythrocytic stage, the more likely site of priming of the immune response is the spleen [47]. During its blood-stage development, the parasites remodel the infected erythrocytes, leading to changes in deformability and rheological properties of the cells [48]. The infected red blood cells (iRBC), which cannot deform, go through small capillaries and accumulate in the spleen. These iRBCs are then recognized and destroyed by phagocytes [49]. P. falciparum parasites have evolved to avoid splenic clearance by expressing neoantigens at the surface of iRBC. These antigens, such as P. falciparum erythrocyte membrane protein 1 (PfEMP1), allow parasites to sequester in deep tissues by binding to a variety of receptors expressed on the surface of capillary endothelial cells [50]. Splenic DCs or blood DCs can capture iRBC through CD36 and present to naïve T cells [51]. However, iRBC binding to CD36 and CD54 on immature DCs limits the DC maturation [52]. In vitro experiments in fact have shown that infected erythrocytes reduce the expression of co-stimulatory molecules on DCs [52] and affect the antigen processing capacity of these professional APCs [52]. It was also found that malaria infection alters the expression of TLRs on DC subsets [53]. In one cross-sectional study, circulating DCs from acutely infected Papuan adults had DCs, which did not mature well in vitro after stimulation and were more prone to apoptosis. This immune dysregulation of DCs is associated with an increased level of IL-10. Antagonizing the IL-10 actions restored DC functions [54]. By decreasing DC maturation efficiency, the parasite prevents the establishment of a potent immune response and facilitates immune evasion. Nevertheless, blood and splenic DCs are effective in inducing inflammatory signals against Plasmodium infection. New vaccine platforms are harnessing DCs for the development of novel malaria vaccines [55]. In addition, any immunopotentiating substances such as adjuvants, which can enhance the antigen presentation capacity of circulating or tissue-specific DCs to induce a protective immune response, are needed.
Majority of the myeloid cells, such as monocytes, macrophages, NK cells, eosinophils, neutrophils, and DCs, participate in the antibody-dependent cell-mediated cytotoxicity (ADCC) functions. Previous studies have given a clue that passive transfer of immune sera has the capacity to activate ADCC to protect the host against malaria [56]. In vitro studies on RBCs, from infected and non-infected malaria, have confirmed that NK cell-mediated ADCC is essential for acquired immunity [57]. In addition, upon parasite exposure, NK cells are accompanied with an early capacity to produce copious amounts of cytokines (e.g., IFN-γ). Data from an RBC-supplemented, immune cell-optimized humanized mouse model have revealed that NK cells interact with P. falciparum-infected erythrocytes through lymphocyte-associated antigen 1 and kill the infected cells [58,59].
A specialized type of NK cells called adaptive NK cells, which express CD56dim without the expression of promyelocytic leukemia zinc finger (PLZF) transcription factor and Fc receptor γ-chain (FcRγ) have been recently described. Studies conducted by Geoffrey et al., in children and young adults, have predicted that presence of adaptive NK cells is associated with protection against P. falciparum-induced malaria infection [60]. As NK cells are heterogeneous, further work is necessary to dissect the role of various subsets of NK cells in the protection against malaria. Nevertheless, based on the above discussion, ADCC function might play an essential role in blood-stage malaria.

2.2. Adaptive Immunity

2.2.1. T Cell-Mediated Immunity

T cells, such as CD4+ (T helper, T follicular helper (Tfh) cells, cytotoxic CD4+ T cells), cytotoxic CD8+, and non-classical T cells such as mucosal-associated invariant T (MAIT) cells and γδ T cells have been shown to have a role in antimalarial immunity (for a comprehensive review, see [61]).

2.2.2. T Cell-Mediated Immunity against Liver-Stage Malaria Parasites

Most of the knowledge on T cell immunity against the malaria liver stage has been obtained in rodent malaria models of protection using whole parasite immunization either with irradiated sporozoites or live sporozoite under drug prophylaxis [62,63,64,65] (Table 1). In these models, it has been shown that CD8+ and CD4+ T cells recognize peptide-derived antigens, which are presented by MHC class I and Class II on the surface of infected hepatocytes [66,67,68,69]. T cells can eliminate infected parasites via different mechanisms [70]. They can produce IFN-γ, which induces nitric oxide synthase to produce nitric oxide, a toxic molecule, in response to liver parasites [71,72,73]. They can also eliminate infected hepatocytes through contact-dependent perforin-dependent mechanisms [74]. IL-4-secreting CD4+ T cells have been shown to be critical for the induction of CD8+ T-cell response to liver-stage malaria [41,75,76]. Liver-resident CD8+ T cells (CXCR6+CD69+) induced by vaccination with whole parasites or a subunit formulation of the Plasmodium ribosomal protein RPL6 are essential to confer protection in mice [77,78]. Experimental vaccination experiments in models using irradiated sporozoites have also suggested that protection is dependent on the presence of liver-resident CD8+ T cells [79]. In human vaccine experiments, CD8+ and CD4+ T cell responses were induced after P. falciparum sporozoite immunization [80]. Vaccine-induced protection was correlated with CD4+ T cell responses [80]. The route of infection is critical for optimum T cell-mediated immunity against liver parasite. In fact, intravenous but not the intradermal injection of irradiated sporozoites induced a full sterilizing immune response [79], possibly due to the induction of resident CD8+ T cells [44,79]. Therefore, vaccines, delivery systems, or adjuvants which that can induce liver-resident T cells and increase antigen processing and presentation by MHC molecules at the surface of infected hepatocytes are highly desirable. This has led to strategies using viral vectors and heterologous prime-boost approaches [80,81,82]. Recently, a novel and promising approach called ‘prime and target’ has been shown to increase the frequency of tissue-resident memory CD8+ T cells in the liver in a mouse model using intravenous injections of viral vectors or nanoparticles [81]. Other cell types, such as γδ or MAIT cells, may also have a significant role in mediating the P. falciparum sporozoite vaccine-induced protection as suggested by the studies done with adults in the United States and Mali [80,83,84]. However, it is yet to be determined how these cells are induced during vaccination and what the best adjuvant will be to activate them to act synergistically with classical T cells for an effective protective immunity.

2.2.3. T Cell-Mediated Immunity against Asexual Blood-Stage Malaria Parasites

T cells are essential for the control and resolution of blood-stage infections. Experiments in rodent malaria models have shown that a CD4+ Th1 response is needed to control the first phase of infection by limiting the parasitemia [85,86,87]. Resolution of the infection is mediated by Th2 and Tfh CD4+ cells, which provide help for efficient protective antibody production [85,86,87]. γδ T and NK cells have been shown to be able to kill the intraerythrocytic parasites in vitro, but their exact role in protection remains a debated question [83,84,88,89].
Until recently, CD8+ T cells were thought to have no role in the protection against blood-stage malaria. However, experiments in rodent models using mice deficient in programme death (PD)-1, a marker of T cell activation and exhaustion, have demonstrated an effector role for this T cell subset [90,91]. Because normocytes, the mature erythrocytes, lack MHC molecules, T cells cannot directly eliminate the parasite inside RBC. Malaria-specific T cells secrete IFN-γ, which activates monocytes/macrophages for phagocytosis and elimination of iRBC [90,92]. A handful of studies in humans have shown that CD8+ T cell activation occurs during falciparum infection. In a recent study in Ghanaian children, expansion of a subset of CD8+ T cells expressing granzyme B correlated with an increase in parasitemia and was more pronounced in patients with severe disease [93]. Thus, it is likely that CD8+ T cells may have a more pathogenic effect than protective role during falciparum infection. This is supported by immunohistopathology investigations on the brain vasculature of 31 children who died from human cerebral malaria. A substantial number of CD3+ CD8+ T cells was found adjacent to endothelial cells [94]. In a mouse model of cerebral malaria, pathogenic CD8+ T cells are responsible for the neurological symptoms and ensuing lethality induced by infection with P. berghei ANKA [95,96]. These cells recognize malaria antigens presented by endothelial cells and release granzyme B after engagement with the MHC molecules. Thus, for blood-stage vaccine development, induction of a strong immune response may not be desirable since it may induce unwanted pathology. Nevertheless, there is a need for well-calibrated T cell and well-targeted B cell responses.
As already mentioned above, malaria infection frequently induces immunosuppression through diverse mechanisms [97]. One of the active T cell suppression pathways involves the induction of regulatory T cells (Tregs). Tregs expressing specific markers such as cytotoxic-T-lymphocyte-associated protein-4 (CTLA-4), lymphocyte-activation gene 3 (LAG-3), programmed cell death-1 (PD-1), and others function as immunosuppressive cells. Both in vitro studies in peripheral blood mononuclear cells (PBMCs) and longitudinal studies in humans have shown that the frequency of Tregs in the patients is negatively associated with the parasite load and/or disease severity [96,98] (extensively reviewed elsewhere [61]). Studies in humans and mice have shown that Tregs via CTLA-4 inhibit the development of a protective anti-blood-stage immunity [99]. Furthermore, clinical studies on malaria-infection/vaccination showed an increased Treg population in the acute and uncomplicated infection and during the convalescence phase [100,101,102]. In contrast, a decrease in the Treg population was also observed in children under chronic malaria exposure [103,104,105]. Thus, knowledge of the factors influencing Treg functions, such as age, level of exposure (high or low transmission intensity), and others, is needed to precisely define their role in the pathogenesis of malaria. Approaches using adjuvants to prevent induction or counteract Treg activity in individuals residing in endemic regions might help in the development of an effective anti-blood-stage vaccine.

2.2.4. Antibody-Mediated Immunity

In malaria infection, antibodies mediate protective immune responses via different mechanisms, such as inhibition of parasite motility, invasion, egress, adhesion and hepatocyte traversal ability, promotion of antibody-dependent complement-mediated sporozoite/merozoite lysis, phagocytosis, antibody-dependent cellular cytotoxicity, transmission-blocking activity, and others [80,106,107] (reviewed in [108]). Interestingly, the generation of natural immunity against P. falciparum is ethnicity dependent. The majority of non-immune Western travelers, if not all, require a single infection to induce invasion inhibitory antibodies against P. falciparum rather than endemic residents, who require two infections [109,110]. Similarly, protection against malaria in infants born in the endemic regions correlate with the pre-existing antibodies (i.e., maternal antibodies) [110]. In addition, children (5 years) who are repeatedly exposed to the natural infection are more resistant to the severe clinical symptoms of malaria [110,111]. Development of P. falciparum antigen-specific B cells to secrete monoclonal antibodies and passive transfer of immunoglobulins have been attractive strategies in malaria research [80,112].
Many malaria candidate vaccines have been designed to induce an effective antibody response. Antibodies against sporozoites or against neo-antigens expressed on the surface of infected hepatocytes mediate protection by preventing or limiting pre-erythrocytic-stage infection and development. Anti-sporozoite antibodies have been shown to inhibit sporozoite motility in the dermis and liver [113], destroy sporozoites in the skin [114], facilitate opsonization and phagocytosis by monocytes or macrophages in the spleen or the liver [114,115], inhibit sporozoite invasion into hepatocytes [116], and inhibit sporozoite development inside the hepatocytes [116]. The type of functional antibodies produced during the infection illuminates the type of protection they offer. For example, IgG1 and IgG3 exhibit potent capacity to activate complement, FcɣR signaling and opsonization compared to IgG2 and IgG4. The former function discriminates the IgG subclasses into cytophilic (IgG1, IgG3) and non-cytophilic (IgG2, IgG4) antibodies [117,118,119]. The above-mentioned antibody functional properties have been confirmed against the majority of Plasmodium spp., including P. falciparum in different life-cycle stages [108]. Although the role of IgG and its subclasses is significant in host to symptomatic malaria infection, the role of IgM must not be ignored. Particularly, the protective role of IgM against the blood-stage parasite of P. falciparum has been established [120,121,122].
Antibodies against parasite neo-antigens, such as heat shock protein, expressed on the surface of infected hepatocytes induce liver parasite killing through an antibody-dependent cell-mediated mechanism, which is likely to involve Kupffer cells or NK cells [123]. Different in vitro assays have been developed to measure antibody functionality, but it is not yet clear which of the immune mechanisms described above are essential or associated with the protection. It might be that, in addition to the high level of neutralizing antibodies, the quality (i.e., avidity, affinity, isotype) of the antibodies is also important. This knowledge is of paramount importance to design better immunogen and use adequate adjuvant.
Antibody-mediated immunity against the blood stage is more complex than in the pre-erythrocytic stage. Anti-merozoite antibodies can (i) prevent merozoites from invading RBC [124,125,126] alone or in conjunction with complement factors [118], (ii) prevent merozoite egress from RBC, (iii) agglutinate free merozoites, (iv) facilitate phagocytosis of merozoites, and (v) promote clearance of iRBC by phagocytic cells through a mechanism called antibody-cell-dependent inhibition (ACDI) [127]. In ACDI, anti-merozoite IgG1 or IgG3 antibodies bind to merozoites, and the immune complexes promote phagocytes, such as monocytes/macrophages or neutrophils, to release cytokines (e.g., TNF-α). This cytokine then stimulates the phagocytes to produce mediators, which leads to the killing of intra-erythrocytic parasites [128,129]. As mentioned above P. falciparum parasites express antigens on the surface of iRBC. These antigens are mainly encoded by multigene families, such as the var [130], stevor [131], and rifin gene families [132]. These antigens are involved in cytoadherence to endothelial cells and in other adhesive phenomena, such as rosetting (the binding of an iRBC to non-infected RBC) and agglutination (the binding to iRBC through bridging by platelets) (for a review, see [50]). The cytoadherence ability of the malaria parasites is key to many of the pathologies induced by P. falciparum infection. Antibodies targeting the surface antigens may prevent cytoadherence and promote iRBC phagocytosis or iRBC agglutination [133,134].
Antibodies targeting parasite toxins could also protect from P. falciparum-induced disease. During the blood stage of the infection, diverse parasite toxins are released at the time of iRBC rupture. These toxins include hemozoin, a by-product of heme degradation by the parasite [135], GPI moieties, which are present in many merozoite proteins [136], a TatD-like DNase [137], and a tyrosine-t RNA synthase [138]. Protection from disease by anti-toxin antibodies has been achieved experimentally using synthetic glycans mimicking GPI [139].
Gametocytes, the sexual forms of P. falciparum parasites, are also targets for antibody-mediated immunity [140]. Gametocytes express a range of antigens, which are targeted by antibodies [141]. The latter facilitates the complement-mediated killing of the gametocytes [142]. In the mosquito after feeding, anti-sexual form antibodies can prevent fusion of gametes [143], induce complement-killing of gametes or ookinetes [144], and prevent ookinete motility, penetration of the midgut wall, and formation of oocyst [145].
Vaccination/infection determines the dominant IgG subtypes induced, which in turn are useful to predict the type of immune responses acquired. Among the IgG subtypes, opsonizing cytophilic antibodies have gained much attention for malaria vaccine development [146,147,148]. These antibodies recruit FcɣR-containing immune cells (particularly macrophages and basophils) via their Fc domain and activate the parasite elimination procedures. Cytophilic antibodies both naturally acquired [149,150,151,152] and vaccine-induced (IgG1 and IgG3 responses against blood-stage antigens) [107] have shown more protective responses in diverse studies.
During the vaccine design, it important to consider both naturally acquired and vaccine-induced immunity. The development of naturally acquired immunity depends on many factors such as region, age group, number of exposures (symptomatic or asymptomatic), number of targeted antigens, and others [149,150]. The vaccine-induced immunity depends on the type of antigens targeted and the quality (type) and quantity of antibodies produced [153]. Although both have similar specificities, the duration and diversity of the immune response is more for naturally acquired immunity over vaccine-induced immunity [154]. Evidence from different studies suggests that naturally acquired immunity, unlike vaccine-induced immunity, reduces the risk of infection irrespective of age [155,156]. Moreover, children living in perennial transmission areas induce more protective natural antibodies (e.g., anti-erythrocyte binding antigen 175RIII–V) than in seasonal transmission areas [149]. However, development of naturally acquired immunity is the best bet in the host. Circumstantially, newborns and infants (<6 months of age) are conferred protection against malaria infection due to the existence of maternal antibodies (extensively reviewed elsewhere [157]).
Antibodies’ breadth (antibodies raised against number of antigens), magnitude (quality), and quantity (affinity and avidity) decide the protection efficiency against clinical malaria. However, it is difficult to estimate the quantity of antibodies that are required for protection, and it purely relies on the antigen specificity. A study conducted in Kenyan children (n = 119), for six months, confirmed that children who possess a breadth of naturally acquired antibodies against blood-stage antigens are inversely correlated with the risk of malaria [158]. With regard to the vaccine development, whole-sporozoite vaccines are ideal to obtain a breadth of antibodies against the parasite. Like natural exposure, whole-sporozoite vaccines increase the magnitude of antibodies due to the varying antigenic content [80].
Interestingly, it has been observed that antibody breadth and magnitude increase with age (4 years < 14 years < adults) [159]. Though not always, increased risk of infection has also been observed in the presence of antigen-specific antibodies (e.g., anti-AMA1) [160,161]. However, additional studies are required to confirm the breadth, magnitude, affinity, and avidity of the protective antibodies in both asymptomatic and symptomatic infections [162,163].
As mentioned above, passive transfer of neutralizing monoclonal antibodies is an alternative therapeutic strategy, which gives short-term protection. In addition, epitope-specific monoclonal antibodies have been generated against different stages of P. falciparum and evaluated in several models (reviewed, recently) [112]. At present, subunit, DNA, and viral vector vaccines are capable of inducing high antibody titers, though the potency and breadth are not sufficient to give complete protection [164]. To achieve more germinal center reaction and memory B cell generation, the addition of vaccine adjuvants and adjuvant delivery systems are essential (discussed in the later section). Finally, natural or vaccine-induced high antibody titers are needed to prevent the hepatic entry of the parasite and erythrocyte infection.

3. Vaccines

Vaccines are one of the most efficient prophylactic treatments for many diseases, ranging from smallpox to the recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, which causes coronavirus disease 2019 (COVID-19)). With the ever-increasing knowledge of immunity, vaccine strategies have focused on identifying the target antigens and inducing a protective immune response with low to no side effects. Due to the complex antigenicity and immune attacking mechanisms of P. falciparum, the development of vaccine for this parasite has been and is still challenging (Appendix A) [165]. However, many traditional vaccine strategies do not answer the question of life-cycle complexity and polymorphism of parasite proteins, which are just two of the many reasons we do not yet have a highly effective malaria vaccine [80].
A variety of malaria vaccines have been developed and evaluated from the past decades, ranging from classic approaches such as whole inactivated parasites to subunit vaccines and new delivery systems. Although adjuvants enhance vaccines’ immune response, they are occasionally not efficacious in formulations with malarial antigens. Different recombinant malarial antigens have been produced (Table 1 and Table 2) in various expression systems (e.g., E. coli, L. lactis, N. benthamiana, P. pastoris, S. cerevisiae, and others) with a variety of delivery platforms (e.g., conjugates, fusion proteins, virus-like particles, virosomes, and others). Over the years, researchers have used various antigens for vaccine development against the different stages (sporozoite, merozoite, and sexual) of the parasite life cycle [166]. However, it has been challenging to identify the ideal target antigen, among the 5000 proteins encoded in the P. falciparum genome [167]. Genomic, transcriptomics, and proteomics approaches have helped to define protein expression patterns during the P. falciparum life cycle [168]. Among the proteins identified, the uniqueness of sporozoite, trophozoite, merozoite, and gametocyte proteins are categorized [168]. The different protein targets (mentioned in Table 1 and Table 2) are exploited in distinct ways for developing malaria vaccines: (i) pre-erythrocytic vaccines, (ii) erythrocytic vaccines, (iii) transmission-blocking vaccines (extensively reviewed elsewhere [80,110,169]). Several candidates have been tested; several immunogens are at preclinical stage, among which 30 immunogens are in clinical evaluation though only 2 have crossed Phase IIb trials or beyond (without adjuvants; https://clinicaltrials.gov/ accessed on 1 June 2020).
In the past 100 years, many adjuvants have been evaluated preclinically against diverse antigens of malaria (Table 2). Notably, RTS’S/AS01 (Mosquirix™) (Appendix B) and PAMAVAC vaccines have successfully entered advanced clinical trials.

4. Vaccine Adjuvants

In general, most vaccine-based antigens are inefficient at mounting protective immune responses due to the dearth of immunogenicity, even though they proved to be perfect target antigens. Nevertheless, an additional component, named adjuvant (adjuvare (Latin): To help) has proven to be critical in boosting the antigens’ immunogenicity. Several reviews have covered the qualities of most vaccine adjuvants and their mechanism of action [170,171,172,173,174,175]. Like immune response in infections, adjuvants induce an immune response by triggering innate and adaptive immune responses. The type of immune response induced by the adjuvant is critical for the choice of vaccines. For example, adjuvants inducing Th1 response are preferable in vaccines against intracellular pathogens, and adjuvants inducing Th2 responses are preferable in vaccines against extracellular pathogens [174,176]. In both cases, Tfh cell responses are critical in inducing prolonged antibody responses [177]. Tfh cells are distinguished from the other T helper cells via expression of a CXCR5 receptor and a transcription factor B cell lymphoma 6 (Bcl-6). During the infection or vaccination (with or without adjuvants) [178], Tfh cells migrate to the secondary lymphoid organs and provide help to the germinal center B cells to induce high-affinity memory B cells and long-lived plasma cells [177,179]. The role of Tfh cells in humoral immunity has become more motivating in the design of subunit vaccines [180].
Protective immunity against P. falciparum requires neutralizing antibodies [181,182] and optimal Th1-mediated immunity [183]. For enhancing, modulating, and prolonging the specified immune response against any P. falciparum vaccine candidate, vaccine adjuvants that facilitate these responses are needed. These adjuvants will reduce the antigen concentration and frequency of immunization to attain protective efficacy, thereby making vaccines more cost-effective. Though adjuvants face many challenges (physicochemical interactions, stability, pairing with partner antigen, and others), recent technologies in vaccine adjutants have come to the stage of increasing immune responses via immune synergy (use of multiple adjuvants) [172]. Reasonably, the immune synergy strategy has not been used extensively in the vaccines composed of parasitic antigens [184].
Different types of adjuvants have been categorized, such as TLRs-, nucleotide-binding oligomerization domain-like receptors (NLRs)-, C-type lectin receptors (CLRs)-based, and some other non-specific PRRs agonists, NLR family pyrin domain containing 3 (NLRP3) activators, formulations including liposomes, Adjuvant System (AS), nano/micro particles-based adjuvants, immune-complexes, and others. Recent years have seen the approval of several new-generation adjuvants containing vaccines for human use [185]. Inflammation (either specific receptor activation or non-specific activation) is the primary response to a majority of the adjuvants leading to antigen-specific cellular immunity [185]. Among the long list, formulation vaccines have gained much attention with respect to their efficacy, such as AS01, AS02, AS03, AS04, MF59, and others. The majority of these systems have more than one immune-potentiator in them. The use of these formulations has induced a protective immune response against antigens and has also increased the immune responsiveness in an elderly population, e.g., influenza vaccine (MF59 or AS03) and herpes zoster vaccine (AS01) [185]. Although many adjuvants, including alum (a century-old approved adjuvant), are used in clinical trials, the lack of a clear understanding of mechanisms of action against malaria has impaired the development of efficient immunogen/adjuvant formulation [186]. Viral vector-based vaccines have shown better immunogenicity than adjuvanted protein vaccines [187]. Herein, we discuss the importance of current adjuvants, which are being employed in developing malaria vaccines at the clinical stage, focusing on their outputs like immunological profile and mechanism of action of successful adjuvants.

5. Adjuvants under Clinical Evaluation

5.1. Alum

Alum (common name for aluminum potassium sulfate) is a 90-year-old gold standard vaccine adjuvant that produces primarily humoral immune responses [188,189]. A recent review detailed the physico-chemical and biological properties of alum adjuvants [188]. Mostly, two types of aluminum adjuvants (aluminum hydroxide (Al(OH)3) and aluminum phosphate (AlPO4)) are used in the clinically approved vaccines and these are the most widely applied adjuvants for evaluating the malaria vaccines [190]. The merits of aluminum adjuvants include safety, amplification of antibody responses, and a comparatively easy process to produce on a large scale. It also has certain drawbacks like the inability to induce cell-mediated immune responses (Th1 and cytotoxic T lymphocytes (CTL)), which are essential for P. falciparum [190]. Since alum induces Th2 biased cellular responses, it was administered concomitantly with other adjuvants (alum+CpG motifs (ODN) [191] alum+glucopyranosyl lipid adjuvant (GLA) [192]) or adjuvant formulations in the development of malaria vaccines. In preclinical studies, often, the antigen is mixed with alum before injection in mice [193,194]. Unlike other adjuvants, alum is the most successful single vial- or prefilled syringe-based vaccine adjuvant. Though intense research has been done on the alum, its precise mechanism of action, however, is still elusive. Interestingly, studies in mice showed that alum forms a depot at the injection site and enters the innate cells via phagocytosis. It activates the NLRP3 inflammasome via phagolysosomes followed by the activation of caspase-1 and release of IL-1β. The known mechanism of action of alum was reviewed elsewhere [189]. Nevertheless, the proposed mechanism is not observed either in humans or in monkeys [195,196]. Thus, there should be another mechanism of action of alum in humans, which is yet to be identified [185]. The vaccination of multistage antigen SpF66 combined with alum resulted in the development of short-lived antibodies and minimal cellular response with no or little protection. Further, the evaluation of pre-erythrocytic protein Pfcs102 with alum was not successful in enhancing an antigen-specific antibody response [190,197]. Blood-stage vaccines (AMA1[PfAMA-1-FVO25–545], AMA1-C1, GLURP85–213, and MSP1-C142) adjuvanted with alum induced modest antibody levels, a poor cellular response, and no protection in field clinical trials [190]. In a randomized, double-blind, placebo-controlled study, one vaccine (PRIMVAC) adjuvanted either with alhydrogel (aluminum hydroxide wet gel suspension; regularly 2% Al(OH)3 is used) or GLA-SE had a significant immunogenic response and VAR2CSA-specific antibodies with the desired inhibitory properties [198].
The transmission-blocking Pf25, a sexual-stage vaccine, has been clinically evaluated in combination with alum. It induced local reactogenicity and limited immunogenicity [199,200]. Altogether, it is clear that alum alone is not sufficient to induce effective responses to malaria vaccines [201].

5.2. Vaccine Delivery Systems/Formulations

Although effective or moderately effective protein vaccine candidates have been developed for malaria, they have poor immunogenicity. Targeted delivery of subunit vaccines via systems possessing adjuvant properties is of paramount importance and a very classical approach. It ensures their effective delivery as well as the ability to increase protective immunity [166]. However, like alum, the mechanism of adjuvanticity of emulsion-type delivery systems remains to be identified [185].

5.2.1. Liposomes

They are synthetic phospholipid models (nm to µm size), which carry antigens encapsulated into the aqueous core (hydrophilic molecules), adsorbed to the surface (lipophilic molecules), or integrated into the lipid layers (amphiphilic molecules) [202]. The adjuvanticity of liposome is mainly reliant on size, charge, preparation method, and number of lipid layers [203]. Cationic adjuvant formulations made of dimethyl dioctadecyl ammonium combined with the stabilizer glycolipid trehalosedibehenate (TDB) were found to be immunogenic liposome forms [204]. The disadvantages of liposomes are stability, manufacturing process, high costs, pain at the injection site, and the addition of immunostimulatory molecules like AS01 [190]. Because of the instability of liposomal vaccine delivery systems in a single vial liquid format, the formulation should be admixed with the vaccine before administration, e.g., Mosquirix® vaccine [201].

5.2.2. AS01

AS01 is a liposome-based vaccine adjuvant system containing two immunostimulants: 3-O-desacyl-4′-monophosphoryl lipid A (MPL) and the saponin QS-21. It has been extensively studied for vaccines against various infectious diseases, including catarrhalis, influenza, tuberculosis, HIV, and others [205]. Interestingly, unlike other vaccine adjuvant systems, AS01 has a recognized mechanism of action. It activates the resident macrophages, followed by draining to the lymph nodes, where it activates the NK cells to release IFN-γ. The released IFN-γ activates DCs, which induce an antigen-specific Th1 response (i.e., IL-2, IFN-γ, and TNF-α) [206,207,208]. The above-mentioned mechanism of action is due to the synergistic effect of two immunostimulants, i.e., MPL (monophosphoryl lipid A) and QS-21, present in AS01 (Figure 3). Both the molecules induce Th1-mediated immune responses [209]. In addition, in delayed fractional doses AS01/RTS, S has been shown to induce IL-21-secreting antigen-specific peripheral Tfh cells with protective B cell responses [210]. However, individual components have their specific effects (see the relevant text). QS-21 delivered via liposomes protects the cells from QS-21-induced cell death [211]. This might be the reason that QS-21-containing AS01 has exhibited more improved efficacy than other formulations. In addition, malaria vaccine clinical trials have used different forms of AS01, i.e., AS01B full dose of immunostimulants for adults and AS01E half dose of immunostimulants for children [185,212] (Table 2).
Note: Glucopyranosyl Lipid Adjuvant (GLA) is a synthetic hexaacylated lipid A derivative. Monophosphoryl lipid A (MPL®) is primarily produced from Salmonella minnesota R595 by eliminating phosphate from the reducing-end glucosamine, core carbohydrate group, and the acyl chain from the 39-position of the disaccharide backbone [213].

5.2.3. Emulsions

Emulsions are biphasic liquid dosage forms composed of three main components, oil phase, aqueous phase, and emulsifying agents (surfactants), and classified under conventional, widely used vaccine delivery systems. They are water-in-oil (W/O), oil-in-water (O/W), and multiple emulsions (water-in-oil-in-water (W/O/W) and oil-in-water-in-oil (O/W/O)). Among the emulsion types, O/W type emulsions are highly preferred because of less reactogenicity and sustained release. For example, oleic acid O/W emulsion with monophosphoryl lipid A (MPLA) and squalene-based O/W emulsion with murabutide induce a significant cellular as well as high avidity humoral immune response in mouse models [214,215]. A potent O/W emulsion-based immunological adjuvant, MF59, which is composed of squalene droplets stabilized with Tween 80 and Span 85, elicited higher antibody titers with balanced antibody subclasses than those observed in alum studies. In contrast to conventional emulsions, MF59 is composed of metabolizable oils (squalene; component in cholesterol synthesis pathway), making MF59 much safer. Strikingly, although MF59 is more highly efficacious, stable, and potent than other vaccine adjuvants (alum and CpG) with ease of scale-up competence [185,216], it did not offer reasonable results in malaria vaccines; hence, there was no further progress with MF59 in developing malaria vaccine [217,218,219,220]. Irrespective of antigens, O/W emulsions elicit Th1/Th2 responses rather than Th1/Th17 responses [221]. Other emulsion-based adjuvants have been developed and have shown to be more efficient.

5.2.4. AS02

It is composed of two immunostimulants: QS-21 and MPL (3-deacylated monophosphoryl lipid A) in squalene O/W emulsion. Preliminary results in rodents and non-human primates have shown that AS02 is efficacious [222,223]. Further, early clinical trials of AS02 with different malarial antigens, such as PfCS102 (a P. falciparum circumsporozoite protein immunogen) and LSA-1 (liver-stage antigen) have shown an increased Th1 response [224]. Initial trials with RTS, S/AS02 formulation (AS02A and AS02D are varying doses of formulation for adults and infants, respectively) have found that the vaccine is safe, well-tolerated, and immunogenic in adults, infants, children, and semi-immune adults [225,226,227,228,229]. Interestingly, RTS, S/AS02 formulation exhibited effective cellular immunity and significant protection against sporozoite-challenge malaria infection when compared to other AS formulations [186,212,230]. However, further trials, with distinct dosage regimens have reported no or minimal protection [186,212,231]. In view of these outcomes, among the AS formulations for malaria vaccines, AS01 holds a superior efficacy and has been selected for further clinical trials [186,212,225].

5.2.5. Montanides (ISA 51, ISA 720)

Montanide ISA 51 and ISA 720 are W/O type emulsions, which are composed of mineral oil and non-mineral oil, respectively, with mannide monooleate as an emulsifier. ISA 51 was approved for lung cancer due to its high CTL responses. Clinical studies on ISA 51 adjuvant with mosquito-stage antigen (Table 2) yielded unexpected systemic adverse events (erythema nodosum), which stopped further exploratory studies in malarial vaccines [232]. ISA 720 has been extensively studied in malaria vaccines because of its ability to induce high antibody production rather than T cell responses. In most of the malaria vaccines, ISA 720 has been shown to induce a strong immune response, but with associated problems such as pain and unacceptable reactogenicity at the site of injection and formulation instability [190,233]. All these findings further support that O/W emulsions are preferred in human vaccines.

5.3. Immune Potentiators or Immunomodulators

Substances, such as particulate materials, complexes, delivery systems, and others, which modulate the immune responses either by targeting the innate immune system or skewing the Th1- and Th2-dependent antibody production, are referred to as immune potentiators. Many molecules derived from the classes of structure-guided vaccine adjuvants or immune potentiators, e.g., PamCysk4 analogues (TLR2 agonists), α-Glactosylceramide analogues (CD1d-iNKT linkers), saponins/glycolipids, and others, show immune-modulating potential [193,194,234,235,236,237,238,239]. In this section, we describe different potent immunomodulators, which are potent inducers of Th1 responses and have been used in malaria vaccines, either alone or in combinations.

5.3.1. QS-21

QS-21 saponin is derived from a natural source, the bark of Quillaja saponaria Molina. Very recently, the mode of action of QS-21 was documented. After injection, it is drained to the lymph nodes and activates subcapsular macrophages (CD11b+CD169+), where activated macrophages recruit neutrophils and DCs. DCs undergo maturation and induce antigen-specific T cell (CD4+ and CD8+) and antibody responses. Interestingly, further studies confirmed that QS-21, like ISCOMATRIX and alum, induces the activation of inflammasome and the release of IL-1β. Inflammasome activation is mediated via the MyD88 pathway with the partial activation of high-mobility group protein B1-TLR4 (Figure 4) [240]. QS-21 (in AS01)-dependent DC activation is mediated through cathepsin B. Moreover, QS-21-mediated Syk kinase activation and lysosomal destabilization are essential for monocyte-derived DC activation and antigen-specific immune response (Figure 4) [241]. Nevertheless, a structural-activity relationship clearly suggests that triterpene aldehyde and fatty acyl side chains are the vital moieties for QS-21 action. Strikingly, deacylation reduces the induction of Th1-type responses [211].
Though it is effective in inducing immunity and widely used in many vaccines, including malaria, HIV-1, and cancer, QS-21 also possesses certain limitations, such as complex steps of manufacture/synthesis, hemolytic effects, biodiversity problems (due to the cutting down of trees), the dose to efficacy ratio, and others [174]. Considering all these limitations, novel structural mimics and alternative or simplified structures are under progress [239]. Due to the virtue of its immune response, many formulations are use QS-21 as a discrete component in the composition [185]. Interestingly, QS-21 containing nanopatch–skin delivery systems have been shown to be effective for influenza protein antigen. In this study, a nanopatch, which contains a 30-fold lower dose of QS-21, has exhibited an equal antigen-specific humoral response to QS-21 alone, given intramuscularly [242]. This clearly suggests that in the future, QS-21 with malarial antigen must be tested in nanopatches or a similar delivery system to spare the doses of both antigen and adjuvant.

5.3.2. Other Saponin Based Adjuvants (Quil-A and ISCOMs)

Quillaja saponaria is the base for Quil-A and is the most widely used adjuvant from the saponin group. It exhibits lower toxic effects but enhanced adjuvanticity. Further, complex saponin adjuvant, immunostimulating complexes (ISCOMs; Matrix-M™) (spherical, open, and cage-like structures) formed by the cholesterol and phospholipids with the adjuvant (Quil-A or QS-21), trigger both cell-mediated and humoral-mediated immune responses. On the other hand, Quil-A is complicated on account of its chemical-related problems plus local reactogenicity. Although malaria vaccine candidates combined with ISCOMs have shown good results in preclinical investigations [243], very few clinical trials have been conducted with malarial vaccines composed of ISCOMs (NCT02905019), probably due to the pain at the injection site and other drawbacks such as flu-like symptoms, fever, and malaise with less reactogenicity [190,244,245].

5.3.3. CpG ODN

Oligodeoxynucleotides (ODNs) adjuvants are single standard DNA molecules with varying degrees of unmethylated CpG motifs. Like most vaccine adjuvants, CpG ODN also enhances the innate immune cell (pDCs, monocytes, NK cells)-dependent adaptive immune responses (Th1 and B cells) against the co-administered antigens [246]. Considering the strong immunomodulatory properties, especially Th1 responses, CpG ODNs are widely used in various diseases such as infections, cancer, and allergies [246]. Based on their structural-activity relationship, CpG ODNs are divided into different types: class A (Type D), class B (Type K), and class C [174,246] (reviewed in [174]). Among the ODNs, ODN 2006 (7909) is widely used because of its immunostimulatory properties. It comes under the class B category, and it stimulates pDCs to secrete IFN-α via binding to the TLR9 receptor. The detailed mechanism of action of CpG ODNs has been reviewed in the literature [174,246]. Many preclinical studies, either alone or in combination, have documented the efficacy of CpG in P. falciparum infection [247]. This ODN has successfully entered the malaria vaccine clinical trials and has been used as an adjuvant for malarial antigens, such as AMA1, MSP1(42), and their different combinations (Table 2). In one of the clinical studies, naïve human volunteers who received AMA1-based blood-stage malaria vaccine along with CpG ODN, either alone or in combination with alhydrogel, exhibited enhanced and long-lasting antigen-specific antibody responses. In addition, antigen-specific antibody (IgG) showed enhanced growth inhibition of homologous P. falciparum, in vitro [248]. However, subsequent studies with semi-immune adults living in Mali did not give encouraging results [249]. The above results perhaps depend on the refractory/chemistry of CpG ODNs and should be taken into consideration for the future use of CpG ODN-containing vaccines in malaria endemic areas. One more blood-stage vaccine candidate, serine repeat antigen 5 (SERA5), has shown protective immunity in Aotus and squirrel monkeys. A recombinant antigen, SE36 (which lacks serine repeats of SERA5), was absorbed into alhydrogel and evaluated in clinical trials. Surprisingly, it did not induce high antigen-specific antibody titers in clinical trials [250]. Further, SE36/alhydrogel was tested in rodents and non-human primates (monkeys) with or without CpG ODNs. Among the CpG ODNs tested, K3 CpG ODN (K3 CpG ODN is a subclass of ODNs under class B (Type K)) has exhibited safe, efficacious, and immunogenic properties [251]. However, with CpG ODN, these positive preclinical data are still not translated into clinics [252].

6. Concluding Remarks

P. falciparum is an infectious agent of malaria, which kills millions of people around the globe. A vigorous effort has been made for the global elimination of P. falciparum malaria, but asymptomatic individuals are regarded as reservoirs for parasite transmission due to the lack of effective diagnostic methods for detecting the parasite in endemic areas.
For the treatment of malaria, the spread of anti-malarial drug resistance has become a major problem. If it is not handled appropriately, it could reverse the malaria control program and worldwide containment achieved so far. Therefore, more research is necessary to find new anti-malarial drugs for combating multidrug-resistant P. falciparum [253].
The factors like complex life cycle, genetic diversity, and the various immune escape mechanisms developed by these parasites, i.e., antigenic variations in P. falciparum [97], are the major obstacles responsible for the delay in the development of a suitable vaccine. Because of the high number of polymorphism or allele-specific variations in the proteins, single protein-based vaccines have limited success [254]. Vaccines composed of a chimeric molecule of the pre-erythrocytic target have shown protective responses in mice models [255]. The current efforts towards malaria parasite and vector control should be complemented with an effective P. falciparum vaccine for the successful fight against the malaria burden. Presently, vaccines from different stages of P. falciparum are in development and in the coming days, they might be combined (multivalent vaccines) to overcome the parasitic immune control mechanism attributed to antigenic polymorphism. A better understanding of the P. falciparum life cycle and the parasites’ interaction with the host should provide guidelines for the future vaccine development programs. The ideal malaria vaccine should be safe, highly effective, stable, easy to administer, and must provide long-term immunity. Such vaccines should also be cost-effective and affordable in poor malaria endemic areas.
Nowadays, researchers are fully aware of using suitable adjuvants for the development of novel vaccines. With regard to malaria, as mentioned above, an effective vaccine requires long-lasting primary and memory antigen-specific CD4+ T cell, B cell, and CTL responses. However, age-old adjuvants like alum, which skews the immune response to Th2, are not suitable for malaria vaccine development. Importantly, a combination of Th1 and Th2 adjuvants with a proper delivery system is needed for malaria vaccine development. Many prediction models have confirmed that activation of the IFN-γ signaling pathway is the best target to confer vaccine-induced protection against P. falciparum [256]. Next-generation vaccines should have wide spectrum activity against parasites, as some of the endemic residents encounter infection with more than one parasite simultaneously [257]. On top of that, vaccines should not induce suppressive Tregs, which are shown to aid in the parasite immune evasion mechanisms [110]. Further, an ideal adjuvant should be able to produce a protective immune response with a less frequent number of administrations. Nevertheless, we noticed that many adjuvants enhance effective immune responses with experimental antigens or model antigens, whereas a similar trend might not happen with the real antigens and unpaired antigens (Bonam, SR and Kumar HMS, personal observation; unpublished data). All the above details should help in the selection of specific adjuvant systems for the development of novel vaccines that elicit the highest humoral and cellular responses. Nowadays, analytical characterization of formulations to obtain vital information (i.e., particle size, morphology, and concentration of components, physicochemical interactions, stability, and others) are routinely performed. On the other hand, detailed studies are required to establish the biological interactions and determine the exact mechanism of action [201]. Systems vaccinology has helped considerably to uncover the unidentified vaccine adjuvant mechanism(s). Finally, the repository of novel Th1/Th2 adjuvants should be available to vaccine researchers to test their antigens for developing novel, promising malaria vaccines.
In summary, there is a need to coordinate research in multiple directions to develop an efficient malaria vaccine, such as (1) clear understanding of the life cycle of the malaria parasite, (2) identification and characterization of targets of interest with a broad range of strains covered, (3) identification of suitable adjuvant, which could enhance the magnitude and quality of the immunogenicity of antigen(s) without age factor [172], and (4) characterization and selection of a suitable ‘perfect mix’ (e.g., emulsion, liposomes, nanoparticles, etc.) for the delivery of the final vaccine to the host [173]. In essence, a detailed spatiotemporal analysis of parasite invasion biology will provide more information for effective targets. By tackling all these parameters, we could also support the poor responders in the vaccination.

Author Contributions

S.R.B., L.R. and G.T. came up with the idea and wrote the first draft. The draft was revised by L.R., J.B. and H.M.S.K. All authors have read and agreed to the published version of the manuscript.

Funding

Laurent Rénia was supported by core funding to the A*STAR ID labs given by the Agency of Science, Technology and Research (A*STAR). G.T. is a Research Associate-II funded by the Department of Biotechnology (DBT), Government of India, India.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Authors would like to thank Samarchith P. Kurup (Center for Tropical and Emerging Global Diseases & Department of Cellular Biology, The University of Georgia, US) and Giuseppe Del Giudice (Translational Science Leader, R&D Center Italy—GSK Vaccines Srl, Siena, Italy) for scientific discussion. G.T. and H.M.S.K. are thankful to Director CSIR—Indian Institute of Chemical Technology (IICT) for the support (IICT/Pubs./2020/080). J.B. acknowledges the support of the European Union Horizon H2020 programme under Grant Agreement No. H2020-SC1-2019-874653-INDIGO and the Department of Biotechnology (DBT), Government of India. (Figure 2) was created with BioRender.com under a paid subscription.

Conflicts of Interest

We declare that the research was conducted in the absence of any relationships that could construe as a potential conflict of interest.

Abbreviations

ADCCantibody-dependent cell cytotoxicity
AIM2absent in melanoma 2
AP-1activator protein 1
ASCapoptosis-associated speck-like protein containing a CARD (caspase activation and recruitment domains)
CCL2/MCP-1monocyte chemoattractant protein-1
CCL4/MIP-1βmacrophage inflammatory protein-1β
cGAScyclic GMP-AMP synthase
DAMPsdamage-associated molecular patterns
DCsdendritic cells
dsDNAdouble-stranded DNA
ERKextracellular signal-regulated kinases
GPIglycosylphosphatidylinositol
IKKIκB kinase
IRFIFN (interferon) regulatory factor
ILinterleukin
JNKc-Jun N-terminal kinases
MAPKmitogen-activated protein kinases
MDA5melanoma differentiation-associated gene 5
MHCmajor histocompatibility complex
NF-kBnuclear factor kappa-light-chain-enhancer of activated B cells
NK cellsnatural killer cells
NLRP3NOD-, LRR- and pyrin domain-containing 3
PAMPspathogen-associated molecular patterns
PRRspathogen-recognition receptors
STINGstimulator of IFN genes
TCRT cell receptor
Tfhfollicular T cell
TGFtransforming growth factor
Thhelper T cell
TLRstoll-like receptors
TNFtumor-necrosis factor
TRAF6tumor necrosis factor receptor-associated factor 6
Tregregulatory T cell

Appendix A

Key Points in Malaria Vaccine Development

  • Spatiotemporal relation between liver anatomy and sporozoite biology should be explored in detail [258].
  • Invasion strategies of sporozoites (in liver) and merozoites (in RBCs) should be elucidated [259,260].
  • Unlike other infections, vaccine adjuvants, which induces both Th1 and Th2 mediated immunity is essential against P. falciparum malaria.
  • Dominant MHC I-presenting antigen epitopes should be selected for cell-mediated, especially CD8+ T cells and tissue-resident memory T cells in the liver, which are essential to confer vaccine-induced protection [261].
  • Protective malaria vaccines should elicit IgG1 and IgG3 antibody responses, which are protective against P. falciparum malaria [147,262,263].
  • Protective vaccines (e.g., RTS,S) to pre-erythrocytic stage infections are greatly needed [264].
  • In malaria vaccines, along with target antigen, development of adjuvants (e.g., AS01) is also considered as major discipline of research [240,265].
  • Prime-boost and/or prime-target immunization strategies are probably better options to confer protection against liver-stage infections [266].
  • Subcutaneous vaccination has shown increased response than the intradermal vaccination [267]. In addition, vaccines are preferably effective, if given by intravenous route [79].

Appendix B

Appendix B.1. RTS, S Malaria Vaccine

  • RTS, S is a genetically engineered vaccine where “RT” denotes C-terminal (190 amino acids) part of P. falciparum (strain NF54) circumsporozoite protein (CSP) protein, which contains both T and B cell epitopes (central repeat region). “S” is a surface antigen of hepatitis-B virus and the combination is expressed in Saccharomyces cerevisiae (S) [268].
  • RTS, S was produced by Walter Reed Army Institute of Research (WRAIR) in collaboration with GlaxoSmithKline (GSK) biologicals and the Programs for Appropriate Technologies in Health (PATH) Malaria Vaccine Initiative (MVI).
  • The idea behind the development of the RTS, S vaccine is, like other vaccines (e.g., ChAd63/MVA ME-TRAP), to inhibit sporozoite motility at pre-erythrocytic stage and preventing the entry to liver cells.
  • The first licensed vaccine for children living in endemic areas in sub-Saharan Africa, and is also approved by European Medicines Agency (EMA) in 2015 [269].
  • Presently under clinical evaluation in infants and children in Africa (Phase IV/NCT00866619). Previous regimens are partly effective, now four dose- schedule have been initiated in Kenya [270].
  • Individuals vaccinated with RTS,S/AS01 has shown increased NK cell activation in response to increased T cell activation [58,265].
  • Studies in infants and children vaccinated with RTS, S/AS01 confirmed the antibody (IgG subclass)-mediated protection against P. falciparum malaria by altering the natural acquired immunity [147].
  • Safe, immunogenic and potent for individuals who experienced malaria.
  • Like other majority vaccines, RTS, S also has hurdles in the stability maintenance of specified temperatures (2–8 °C), which is certainly expensive in developing countries.
  • RTS, S vaccine with nearly or entirely different adjuvants also exhibit promising results against malaria P. falciparum [264,271,272].
  • Adult vaccination regimen containing RTS,S/AS01B vaccine with ChAd63/MVA ME-TRAP vaccine has not shown interesting results [273].

Appendix B.2. Highlights

  • Fine strategies to eradicate and eliminate malaria through drugs or vaccines have gained momentum.
  • Fundamental functions of CD8+ T cells and NK cells have proven their effective role in preventing malaria transmission.
  • Majority of prophylactic/therapeutic malaria vaccines are in the clinical evaluation.
  • RTS, S/AS01 is the only licensed malaria vaccine.
  • Novel delivery systems of malarial vaccines with QS-21 have yielded increased potency and reduced toxicity.
  • Though subunit vaccines are an effective strategy, the search for novel approaches like live and/or whole parasite vaccinations should be evaluated in detail.

Appendix B.3. Glossary

Plasmodium falciparum
P. falciparum is a parasite protozoan and one of the most infectious species among the other Plasmodium such as P. malariae, P. vivax, P. ovale and P. knowlesi, which causes malaria in humans by using female anopheles’ mosquito as a vector.
Cerebral malaria
Cerebral malaria caused by P. falciparum, affects total neurological functions, and leads to death. Two hypotheses have tried to elucidate the pathogenesis of disease: mechanical and inflammatory. According to mechanical hypothesis, impaired cerebral blood flow due to sequestration of erythrocytes leads to hypoxia whereas inflammatory theory suggests that P. falciparum infection alters brain function via Th1 inflammatory immune response. Symptoms of cerebral malaria include headache, behavioral changes, confusion, coma, and mortality (rate exceeding 10%) [274,275].
Heterologous prime-boost strategy
Multiple immunization of vaccine with or without discrete antigens in DNA vectored priming followed by protein boosting. Strategy is useful in targeting highly complex and pathogenic infections for inducing both humoral and cell-mediated immune responses.
Danger-associated molecular patterns (DAMPs)
Signals from the damaged or dying cells or necrotic cells, which alert the immune system via acting on PRRs.
Clinical immunity
Natural antibody response developed against P. falciparum following repeated infection. It reduces the parasite load and regulates the inflammatory responses.
Sterile immunity
A cent percent immunity achieved by malaria vaccine is termed as sterile immunity. Clinically immune individuals can still act as a reservoir for the parasite. Thus, to confer complete protection against the P. falciparum malaria sterile immunity is needed.
Pre-erythrocytic vaccine
This vaccine with respective antigens prevents the generation of sporozoites into merozoites in the liver thereby reduce the clinically asymptomatic pre-erythrocytic infection. Some of the examples include RTS, S/AS01 (CSP), ChAd63/MVA ME–TRAP (TRAP, CSP, TRAP, CSP, and others), PfSPZ (whole sporozoites), CVac (whole sporozoites), GAP (whole sporozoites), PfCelTOS FMP012 (PfCelTOS), CSVAC (CSP), R21 (CSP), R21/ME–TRAP (TRAP, CSP, and others).
Erythrocytic stage vaccine
This vaccine with respective antigens prevents merozoites’ entry into the RBCs, thereby reducing the clinical symptoms and mortality. Examples include GMZ2 (GLURP MSP3), pfAMA1-DiCo, (AMA1) P27A (P27A), MSP3 (MSP3), SE36 (SERA5), PfPEBS, CHAd63RH5/MVARH5 (RH5), PRIMVAC (Var2CSA), and PAMVAC (Var2CSA) [80].
Transmission-blocking vaccines
These vaccines with respective antigens interrupt malaria transmission by impeding the parasite cycle in the mosquito. Antigens like Pfs25 (Pfs25 VLP & Pfs25-EPA), Pfs28, Pfs48/45, and Pfs230 (Pfs25-EPA) have been assessed as vaccine candidates among which ookinetes of P. falciparum Pfs25 expressed in S. cerevisiae have shown moderate immunogenicity and suboptimal levels of transmission-blocking activity in Phase Ia trials [165,276].
TLRs
Class of PRRs located on the surface and in the cytosol of the cells. Upon recognition of PAMPs/DAMPs, these receptors activate the downstream signaling to secrete diverse inflammatory mediators.
Antibody-dependent cell-mediated cytotoxicity (ADCC)
Antigen-binding fragment (Fab) of antibodies (developed against target protein) binds to the target cells and recruit’s effector cells, which possess receptors for the fragment crystallizable region (Fc) of the antibody (FcγR) to interact with target cell. Activated myeloid effector cells kill the target cells via various mechanisms (release of cytotoxic granule, Fas signaling, induction of reactive oxygen species).
Vaccine delivery systems
These are antigenic formulations (emulsions, liposomes, polymeric particles, adenoviral systems, and others) with or without immunostimulants for targeted delivery and inducing protective immunity (cellular and humoral immunity).
Adjuvant systems (mostly licensed by GlaxoSmithKline plc)
Liposomes and emulsion formulations, which contain one or more immunostimulants to induce antigen-specific immune responses.
QS-21
A water-soluble triterpene glycoside obtained from the purified fractions (fraction 21) of mixture of Quil-A saponins. Quil-A saponins are sourced from the bark of Quillaja saponaria tree. It is potent inducer of both cellular and humoral immunity. QS-21 induces the MHC I antigen cross-presentation followed by the activation of CD8+ T cells.
AS01
Liposomal formulation composed of two synergistic discrete immunostimulatory components, i.e., synthetic 3-O-desacyl-4′-monophosphoryl lipid A (MPL) (a TLR4 ligand) and natural QS-21.
Virosomes
Virosomes are derived from the inactivated/non-replicating virus. Virosomes are composed of bilayer phospholipids, like liposomes and viral glycoproteins. The targeted antigen of interest is encapsulated or cross-linked to the virosomes for effective delivery.
CpG
ODN sequences containing unmethylated CpG motifs (highly methylated in vertebrates); cytosine triphosphate deoxynucleotide (C) linked to the guanine triphosphate deoxynucleotide (G) via phosphodiester bond (p), which specifically binds to TLR9 to trigger immune response. Synthetic CpG ODN over the natural ones has a different level of phosphorothioate backbone.
Structural-activity relationship
It defines the relationship between the chemical structure of a compound to its pharmacological activity and toxicity.

Appendix B.4. Outstanding Questions: P. falciparum Vaccines and Vaccine Adjuvants

  • Knowledge on the optimum combination of multi-stage malarial antigens for the vaccines
  • Influence of natural immunity towards vaccine response and performance in endemic countries
  • Fine incorporation of parameters of anti-disease immunity of the host to conceive effective and safe anti-malarial vaccines.
  • The role of genetic background of the population in determining the vaccine response to malaria.
  • Need of appropriate experimental models that closely mimic human for the pre-screening of malaria vaccine candidates.

References

  1. Andrews, K.T.; Fisher, G.; Skinner-Adams, T.S. Drug repurposing and human parasitic protozoan diseases. Int. J. Parasitol. Drugs Drug Resist. 2014, 4, 95–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. WHO. The “World Malaria Report 2019” at a Glance. Available online: https://www.who.int/news-room/feature-stories/detail/world-malaria-report-2019 (accessed on 1 July 2020).
  3. Key Points: World Malaria Report 2017. Available online: https://www.who.int/malaria/media/world-malaria-report-2017/en/ (accessed on 1 January 2021).
  4. World Health Organization. This Year’s World Malaria Report at a Glance. Available online: https://www.who.int/malaria/media/world-malaria-report-2018/en/ (accessed on 1 July 2020).
  5. Head, M.G.; Goss, S.; Gelister, Y.; Alegana, V.; Brown, R.J.; Clarke, S.C.; Fitchett, J.R.A.; Atun, R.; Scott, J.A.G.; Newell, M.L.; et al. Global funding trends for malaria research in sub-Saharan Africa: A systematic analysis. Lancet Glob. Health 2017, 5, e772–e781. [Google Scholar] [CrossRef] [Green Version]
  6. Kumar, A.; Valecha, N.; Jain, T.; Dash, A.P. Burden of malaria in india: Retrospective and prospective view. Am. J. Trop. Med. Hyg. 2007, 77, 69–78. [Google Scholar] [CrossRef] [PubMed]
  7. Hemingway, J.; Shretta, R.; Wells, T.N.; Bell, D.; Djimde, A.A.; Achee, N.; Qi, G. Tools and strategies for malaria control and elimination: What do we need to achieve a grand convergence in malaria? PLoS Biol. 2016, 14, e1002380. [Google Scholar] [CrossRef]
  8. Cox, F.E.G. History of the discovery of the malaria parasites and their vectors. Parasite Vector 2010, 3, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Krettli, A.U.; Miller, L.H. Malaria: A sporozoite runs through it. Curr. Biol. 2001, 11, R409–R412. [Google Scholar] [CrossRef] [Green Version]
  10. Baer, K.; Klotz, C.; Kappe, S.H.; Schnieder, T.; Frevert, U. Release of hepatic Plasmodium yoelii merozoites into the pulmonary microvasculature. PLoS Pathog. 2007, 3, e171. [Google Scholar] [CrossRef] [PubMed]
  11. Gomes, P.S.; Bhardwaj, J.; Rivera-Correa, J.; Freire-De-Lima, C.G.; Morrot, A. Immune escape strategies of malaria parasites. Front. Microbiol. 2016, 7, 1617. [Google Scholar] [CrossRef]
  12. Phillips, M.A.; Burrows, J.N.; Manyando, C. Nature reviews disease primers. Malaria 2017, 3, 17050. [Google Scholar] [CrossRef]
  13. Yap, X.Z.; Lundie, R.J.; Beeson, J.G.; O’Keeffe, M. Dendritic cell responses and function in malaria. Front. Immunol. 2019, 10, 357. [Google Scholar] [CrossRef]
  14. Cowman, A.F.; Healer, J.; Marapana, D.; Marsh, K. Malaria: Biology and disease. Cell 2016, 167, 610–624. [Google Scholar] [CrossRef] [Green Version]
  15. Thu, A.M.; Phyo, A.P.; Landier, J.; Parker, D.M.; Nosten, F.H. Combating multidrug-resistant Plasmodium falciparum malaria. FEBS J. 2017, 284, 2569–2578. [Google Scholar] [CrossRef] [Green Version]
  16. Ross, L.S.; Fidock, D.A. Elucidating mechanisms of drug-resistant Plasmodium falciparum. Cell Host Microbe 2019, 26, 35–47. [Google Scholar] [CrossRef] [Green Version]
  17. Dagen, M. History of malaria and its treatment. In Antimalarial Agents; Patrick, G.L., Ed.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 1–48. [Google Scholar] [CrossRef]
  18. World Health Organization. Guidelines for the Treatment of Malaria. Available online: https://apps.who.int/iris/bitstream/handle/10665/162441/9789241549127_eng.pdf (accessed on 1 July 2020).
  19. Blasco, B.; Leroy, D.; Fidock, D.A. Antimalarial drug resistance: Linking Plasmodium falciparum parasite biology to the clinic. Nat. Med. 2017, 23, 917–928. [Google Scholar] [CrossRef] [PubMed]
  20. Lubell, Y.; Dondorp, A.; Guerin, P.J.; Drake, T.; Meek, S.; Ashley, E.; Day, N.P.; White, N.J.; White, L.J. Artemisinin resistance—Modelling the potential human and economic costs. Malar. J. 2014, 13, 452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Plebanski, M.; Hannan, C.M.; Behboudi, S.; Flanagan, K.L.; Apostolopoulos, V.; Sinden, R.E.; Hill, A.V. Direct processing and presentation of antigen from malaria sporozoites by professional antigen-presenting cells in the induction of CD8 T-cell responses. Immunol. Cell Biol. 2005, 83, 307–312. [Google Scholar] [CrossRef] [PubMed]
  22. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef]
  23. Bousheri, S.; Cao, H. New insight into the role of dendritic cells in malaria immune pathogenesis. Trends Parasitol. 2008, 24, 199–200. [Google Scholar] [CrossRef]
  24. Draheim, M.; Wlodarczyk, M.F.; Crozat, K.; Saliou, J.M.; Alayi, T.D.; Tomavo, S.; Hassan, A.; Salvioni, A.; Demarta-Gatsi, C.; Sidney, J.; et al. Profiling mhc ii immunopeptidome of blood-stage malaria reveals that cDC1 control the functionality of parasite-specific CD4 T cells. EMBO Mol. Med. 2017, 9, 1605–1621. [Google Scholar] [CrossRef] [Green Version]
  25. Gazzinelli, R.T.; Kalantari, P.; Fitzgerald, K.A.; Golenbock, D.T. Innate sensing of malaria parasites. Nat. Rev. Immunol. 2014, 14, 744–757. [Google Scholar] [CrossRef]
  26. Liehl, P.; Zuzarte-Luís, V.; Chan, J.; Zillinger, T.; Baptista, F.; Carapau, D.; Konert, M.; Hanson, K.K.; Carret, C.; Lassnig, C.; et al. Host-cell sensors for plasmodium activate innate immunity against liver-stage infection. Nat. Med. 2014, 20, 47–53. [Google Scholar] [CrossRef] [PubMed]
  27. Osii, R.S.; Otto, T.D.; Garside, P.; Ndungu, F.M.; Brewer, J.M. The impact of malaria parasites on dendritic cell–T cell interaction. Front. Immunol. 2020, 11, 1597. [Google Scholar] [CrossRef]
  28. deWalick, S.; Amante, F.H.; McSweeney, K.A.; Randall, L.M.; Stanley, A.C.; Haque, A.; Kuns, R.D.; MacDonald, K.P.; Hill, G.R.; Engwerda, C.R. Cutting edge: Conventional dendritic cells are the critical required for the induction of experimental cerebral malaria. J. Immunol. 2007, 178, 6033–6037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Claudianos, C.; Dessens, J.T.; Trueman, H.E.; Arai, M.; Mendoza, J.; Butcher, G.A.; Crompton, T.; Sinden, R.E. A malaria scavenger receptor-like protein essential for parasite development. Mol. Microbiol. 2002, 45, 1473–1484. [Google Scholar] [CrossRef] [Green Version]
  30. Gowda, D.C.; Wu, X. Parasite recognition and signaling mechanisms in innate immune responses to malaria. Front. Immunol. 2018, 9, 3006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Kalantari, P. The emerging role of pattern recognition receptors in the pathogenesis of malaria. Vaccines 2018, 6, 13. [Google Scholar] [CrossRef] [Green Version]
  32. McGuinness, D.H.; Dehal, P.K.; Pleass, R.J. Pattern recognition molecules and innate immunity to parasites. Trends Parasitol. 2003, 19, 312–319. [Google Scholar] [CrossRef]
  33. Jide, C.; Ying, H.; Wenyue, X.; Fusheng, H. Toll-like receptors, a double-edged sword in immunity to malaria. J. Med. Coll. PLA 2009, 24, 118–124. [Google Scholar] [CrossRef]
  34. Wykes, M.N.; Lewin, S.R. Immune checkpoint blockade in infectious diseases. Nat. Rev. Immunol. 2018, 18, 91–104. [Google Scholar] [CrossRef] [PubMed]
  35. Yap, X.Z.; Hustin, L.S.P.; Sauerwein, R.W. Th1-polarized tfh cells delay naturally-acquired immunity to malaria. Front. Immunol. 2019, 10, 1096. [Google Scholar] [CrossRef]
  36. Mauduit, M.; See, P.; Peng, K.; Renia, L.; Ginhoux, F. Dendritic cells and the malaria pre-erythrocytic stage. Immunol. Res. 2012, 53, 115–126. [Google Scholar] [CrossRef]
  37. Winkel, B.M.F.; Pelgrom, L.R.; van Schuijlenburg, R.; Baalbergen, E.; Ganesh, M.S.; Gerritsma, H.; de Korne, C.M.; Duszenko, N.; Langenberg, M.C.C.; Chevalley-Maurel, S.C.; et al. Plasmodium sporozoites induce regulatory macrophages. PLoS Pathog. 2020, 16, e1008799. [Google Scholar] [CrossRef]
  38. Leiriao, P.; Mota, M.M.; Rodriguez, A. Apoptotic plasmodium-infected hepatocytes provide antigens to liver dendritic cells. J. Infect. Dis. 2005, 191, 1576–1581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Chakravarty, S.; Cockburn, I.A.; Kuk, S.; Overstreet, M.G.; Sacci, J.B.; Zavala, F. CD8+ T lymphocytes protective against malaria liver stages are primed in skin-draining lymph nodes. Nat. Med. 2007, 13, 1035–1041. [Google Scholar] [CrossRef] [PubMed]
  40. Ferreira, A.; Enea, V.; Morimoto, T.; Nussenzweig, V. Infectivity of Plasmodium berghei sporozoites measured with a DNA probe. Mol. Biochem. Parasitol. 1986, 19, 103–109. [Google Scholar] [CrossRef]
  41. Kurup, S.P.; Anthony, S.M.; Hancox, L.S.; Vijay, R.; Pewe, L.L.; Moioffer, S.J.; Sompallae, R.; Janse, C.J.; Khan, S.M.; Harty, J.T. Monocyte-derived CD11c+ cells acquire plasmodium from hepatocytes to prime CD8 T cell immunity to liver-stage malaria. Cell Host Microbe 2019, 25, 565–577.e6. [Google Scholar] [CrossRef] [PubMed]
  42. Rénia, L.; Marañón, C.; Hosmalin, A.; Grüner, A.C.; Silvie, O.; Snounou, G. Do apoptotic plasmodium-infected hepatocytes initiate protective immune responses? J. Infect. Dis. 2006, 193, 163–164. [Google Scholar] [CrossRef] [Green Version]
  43. Nussenzweig, R.; Chen, D.F. Sporozoite-induced immunity in mammalian malaria. Am. J. Trop. Med. Hyg. 1972, 21, 722–728. [Google Scholar] [CrossRef] [Green Version]
  44. Seder, R.A.; Chang, L.J.; Enama, M.E.; Zephir, K.L.; Sarwar, U.N.; Gordon, I.J.; Holman, L.A.; James, E.R.; Billingsley, P.F.; Gunasekera, A.; et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 2013, 341, 1359–1365. [Google Scholar] [CrossRef] [Green Version]
  45. Yang, A.S.P.; O’Neill, M.T.; Jennison, C.; Lopaticki, S.; Allison, C.C.; Armistead, J.S.; Erickson, S.M.; Rogers, K.L.; Ellisdon, A.M.; Whisstock, J.C.; et al. Cell traversal activity is important for Plasmodium falciparum liver infection in humanized mice. Cell Rep. 2017, 18, 3105–3116. [Google Scholar] [CrossRef] [Green Version]
  46. Sack, B.K.; Mikolajczak, S.A.; Fishbaugher, M.; Vaughan, A.M.; Flannery, E.L.; Nguyen, T.; Betz, W.; Jane Navarro, M.; Foquet, L.; Steel, R.W.J.; et al. Humoral protection against mosquito bite-transmitted Plasmodium falciparum infection in humanized mice. NPJ Vaccines 2017, 2, 27. [Google Scholar] [CrossRef] [Green Version]
  47. Huisjes, R.; Bogdanova, A.; van Solinge, W.W.; Schiffelers, R.M.; Kaestner, L.; van Wijk, R. Squeezing for life—Properties of red blood cell deformability. Front. Physiol. 2018, 9, 656. [Google Scholar] [CrossRef] [PubMed]
  48. Cooke, B.M.; Stuart, J.; Nash, G.B. The cellular and molecular rheology of malaria. Biorheology 2014, 51, 99–119. [Google Scholar] [CrossRef] [PubMed]
  49. Buffet, P.A.; Safeukui, I.; Deplaine, G.; Brousse, V.; Prendki, V.; Thellier, M.; Turner, G.D.; Mercereau-Puijalon, O. The pathogenesis of Plasmodium falciparum malaria in humans: Insights from splenic physiology. Blood 2011, 117, 381–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Lee, W.C.; Russell, B.; Renia, L. Sticking for a cause: The falciparum malaria parasites cytoadherence paradigm. Front. Immunol. 2019, 10, 1444. [Google Scholar] [CrossRef] [Green Version]
  51. Urban, B.C.; Willcox, N.; Roberts, D.J. A role for CD36 in the regulation of dendritic cell function. Proc. Natl. Acad. Sci. USA 2001, 98, 8750–8755. [Google Scholar] [CrossRef] [Green Version]
  52. Urban, B.C.; Ferguson, D.J.; Pain, A.; Willcox, N.; Plebanski, M.; Austyn, J.M.; Roberts, D.J. Plasmodium falciparum-infected erythrocytes modulate the maturation of dendritic cells. Nature 1999, 400, 73–77. [Google Scholar] [CrossRef] [PubMed]
  53. Loharungsikul, S.; Troye-Blomberg, M.; Amoudruz, P.; Pichyangkul, S.; Yongvanitchit, K.; Looareesuwan, S.; Mahakunkijcharoen, Y.; Sarntivijai, S.; Khusmith, S. Expression of Toll-like receptors on antigen-presenting cells in patients with falciparum malaria. Acta Trop. 2008, 105, 10–15. [Google Scholar] [CrossRef]
  54. Pinzon-Charry, A.; Woodberry, T.; Kienzle, V.; McPhun, V.; Minigo, G.; Lampah, D.A.; Kenangalem, E.; Engwerda, C.; Lopez, J.A.; Anstey, N.M.; et al. Apoptosis and dysfunction of blood dendritic cells in patients with falciparum and vivax malaria. J. Exp. Med. 2013, 210, 1635–1646. [Google Scholar] [CrossRef] [Green Version]
  55. Van Kooyk, Y. Next-generation malarial vaccines. Nat. Mater. 2019, 18, 94–96. [Google Scholar] [CrossRef]
  56. Brown, J.; Smalley, M.E. Specific antibody-dependent cellular cytotoxicity in human malaria. Clin. Exp. Immunol. 1980, 41, 423–429. [Google Scholar] [PubMed]
  57. Arora, G.; Hart, G.T.; Manzella-Lapeira, J.; Doritchamou, J.Y.; Narum, D.L.; Thomas, L.M.; Brzostowski, J.; Rajagopalan, S.; Doumbo, O.K.; Traore, B. NK cells inhibit Plasmodium falciparum growth in red blood cells via antibody-dependent cellular cytotoxicity. eLife 2018, 7, e36806. [Google Scholar] [CrossRef]
  58. Wolf, A.-S.; Sherratt, S.; Riley, E.M. NK cells: Uncertain allies against malaria. Front. Immunol. 2017, 8, 212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Chen, Q.; Amaladoss, A.; Ye, W.; Liu, M.; Dummler, S.; Kong, F.; Wong, L.H.; Loo, H.L.; Loh, E.; Tan, S.Q. Human natural killer cells control Plasmodium falciparum infection by eliminating infected red blood cells. Proc. Natl. Acad. Sci. USA 2014, 111, 1479–1484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Hart, G.T.; Tran, T.M.; Theorell, J.; Schlums, H.; Arora, G.; Rajagopalan, S.; Sangala, A.J.; Welsh, K.J.; Traore, B.; Pierce, S.K. Adaptive NK cells in people exposed to Plasmodium falciparum correlate with protection from malaria. J. Exp. Med. 2019, 216, 1280–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Kurup, S.P.; Butler, N.S.; Harty, J.T. T cell-mediated immunity to malaria. Nat. Rev. Immunol. 2019, 19, 457–471. [Google Scholar] [CrossRef]
  62. Schofield, L.; Villaquiran, J.; Ferreira, A.; Schellekens, H.; Nussenzweig, R.; Nussenzweig, V. Gamma interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature 1987, 330, 664–666. [Google Scholar] [CrossRef]
  63. Weiss, W.R.; Sedegah, M.; Beaudoin, R.L.; Miller, L.H.; Good, M.F. CD8+ T cells (cytotoxic/suppressors) are required for protection in mice immunized with malaria sporozoites. Proc. Natl. Acad. Sci. USA 1988, 85, 573–576. [Google Scholar] [CrossRef] [Green Version]
  64. Silvie, O.; Amino, R.; Hafalla, J.C. Tissue-specific cellular immune responses to malaria pre-erythrocytic stages. Curr. Opin. Microbiol. 2017, 40, 160–167. [Google Scholar] [CrossRef]
  65. Druilhe, P.; Rénia, L.; Fidock, D. Immunity to Liver Stages. In Malaria: Parasite Biology, Pathogenesis, and Protection; Sherman, I., Ed.; ASM Press: Washington, DC, USA, 1998. [Google Scholar]
  66. Rénia, L.; Marussig, M.S.; Grillot, D.; Pied, S.; Corradin, G.; Miltgen, F.; Del Giudice, G.; Mazier, D. In vitro activity of CD4+ and CD8+ T lymphocytes from mice immunized with a synthetic malaria peptide. Proc. Natl. Acad. Sci. USA 1991, 88, 7963–7967. [Google Scholar] [CrossRef] [Green Version]
  67. Renia, L.; Grillot, D.; Marussig, M.; Corradin, G.; Miltgen, F.; Lambert, P.H.; Mazier, D.; Delgiudice, G. Effector functions of circumsporozoite peptide-primed CD4+ T-cell clones against plasmodium-yoelii liver stages. J. Immunol. 1993, 150, 1471–1478. [Google Scholar] [PubMed]
  68. Bongfen, S.E.; Torgler, R.; Romero, J.F.; Renia, L.; Corradin, G. Plasmodium berghei-infected primary hepatocytes process and present the circumsporozoite protein to specific CD8+ T cells in vitro. J. Immunol. 2007, 178, 7054–7063. [Google Scholar] [CrossRef] [Green Version]
  69. Doll, K.L.; Pewe, L.L.; Kurup, S.P.; Harty, J.T. Discriminating protective from nonprotective plasmodium-specific CD8+ T cell responses. J. Immunol. 2016, 196, 4253–4262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Cockburn, I.A.; Amino, R.; Kelemen, R.K.; Kuo, S.C.; Tse, S.-W.; Radtke, A.; Mac-Daniel, L.; Ganusov, V.V.; Zavala, F.; Ménard, R. In vivo imaging of CD8+ T cell-mediated elimination of malaria liver stages. Proc. Natl. Acad. Sci. USA 2013, 110, 9090–9095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Nussler, A.K.; Beger, H.G.; Liu, Z.Z.; Billiar, T.R. Nitric oxide, hepatocytes and inflammation. Res. Immunol. 1995, 146, 671–677. [Google Scholar] [CrossRef]
  72. Belnoue, E.; Costa, F.T.; Frankenberg, T.; Vigario, A.M.; Voza, T.; Leroy, N.; Rodrigues, M.M.; Landau, I.; Snounou, G.; Renia, L. Protective T cell immunity against malaria liver stage after vaccination with live sporozoites under chloroquine treatment. J. Immunol. 2004, 172, 2487–2495. [Google Scholar] [CrossRef]
  73. Belnoue, E.; Voza, T.; Costa, F.T.; Gruner, A.C.; Mauduit, M.; Rosa, D.S.; Depinay, N.; Kayibanda, M.; Vigario, A.M.; Mazier, D.; et al. Vaccination with live Plasmodium yoelii blood stage parasites under chloroquine cover induces cross-stage immunity against malaria liver stage. J. Immunol. 2008, 181, 8552–8558. [Google Scholar] [CrossRef] [Green Version]
  74. Trimnell, A.; Takagi, A.; Gupta, M.; Richie, T.L.; Kappe, S.H.; Wang, R. Genetically attenuated parasite vaccines induce contact-dependent CD8+ T cell killing of Plasmodium yoelii liver stage-infected hepatocytes. J. Immunol. 2009, 183, 5870–5878. [Google Scholar] [CrossRef] [Green Version]
  75. Carvalho, L.H.; Sano, G.; Hafalla, J.C.; Morrot, A.; de Lafaille, M.A.C.; Zavala, F. Il-4-secreting CD4+ T cells are crucial to the development of CD8+ T-cell responses against malaria liver stages. Nat. Med. 2002, 8, 166–170. [Google Scholar] [CrossRef]
  76. Lefebvre, M.N.; Harty, J.T. You shall not pass: Memory CD8 T cells in liver-stage malaria. Trends Parasitol. 2020, 36, 147–157. [Google Scholar] [CrossRef]
  77. Valencia-Hernandez, A.M.; Ng, W.Y.; Ghazanfari, N.; Ghilas, S.; de Menezes, M.N.; Holz, L.E.; Huang, C.; English, K.; Naung, M.; Tan, P.S.; et al. A natural peptide antigen within the Plasmodium ribosomal protein RPL6 confers liver TRM cell-mediated immunity against malaria in mice. Cell Host Microbe 2020, 27, 950–962.e7. [Google Scholar] [CrossRef]
  78. Fernandez-Ruiz, D.; Ng, W.Y.; Holz, L.E.; Ma, J.Z.; Zaid, A.; Wong, Y.C.; Lau, L.S.; Mollard, V.; Cozijnsen, A.; Collins, N.; et al. Liver-resident memory CD8+ T cells form a front-line defense against malaria liver-stage infection. Immunity 2016, 45, 889–902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Epstein, J.E.; Tewari, K.; Lyke, K.E.; Sim, B.K.; Billingsley, P.F.; Laurens, M.B.; Gunasekera, A.; Chakravarty, S.; James, E.R.; Sedegah, M.; et al. Live attenuated malaria vaccine designed to protect through hepatic CD8+ T cell immunity. Science 2011, 334, 475–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Cockburn, I.A.; Seder, R.A. Malaria prevention: From immunological concepts to effective vaccines and protective antibodies. Nat. Immunol. 2018, 19, 1199–1211. [Google Scholar] [CrossRef] [PubMed]
  81. Gola, A.; Silman, D.; Walters, A.A.; Sridhar, S.; Uderhardt, S.; Salman, A.M.; Halbroth, B.R.; Bellamy, D.; Bowyer, G.; Powlson, J.; et al. Prime and target immunization protects against liver-stage malaria in mice. Sci. Transl. Med. 2018, 10, eaap9128. [Google Scholar] [CrossRef] [Green Version]
  82. Hill, A.V.; Reyes-Sandoval, A.; O’Hara, G.; Ewer, K.; Lawrie, A.; Goodman, A.; Nicosia, A.; Folgori, A.; Colloca, S.; Cortese, R.; et al. Prime-boost vectored malaria vaccines: Progress and prospects. Hum Vaccin 2010, 6, 78–83. [Google Scholar] [CrossRef] [Green Version]
  83. Rutishauser, T.; Lepore, M.; Di Blasi, D.; Dangy, J.-P.; Abdulla, S.; Jongo, S.; Ramadhani, K.; Sim, B.K.L.; Hoffman, S.L.; Tanner, M.; et al. Activation of tcr vδ1+ and vδ1vδ2 γδ T cells upon controlled infection with Plasmodium falciparum in tanzanian volunteers. J. Immunol. 2019. [Google Scholar] [CrossRef]
  84. Mpina, M.; Maurice, N.J.; Yajima, M.; Slichter, C.K.; Miller, H.W.; Dutta, M.; McElrath, M.J.; Stuart, K.D.; De Rosa, S.C.; McNevin, J.P.; et al. Controlled human malaria infection leads to long-lasting changes in innate and innate-like lymphocyte populations. J. Immunol. 2017, 199, 107–118. [Google Scholar] [CrossRef]
  85. Perez-Mazliah, D.; Langhorne, J. CD4 T-cell subsets in malaria: Th1/th2 revisited. Front. Immunol. 2014, 5, 671. [Google Scholar] [CrossRef]
  86. Perez-Mazliah, D.; Nguyen, M.P.; Hosking, C.; McLaughlin, S.; Lewis, M.D.; Tumwine, I.; Levy, P.; Langhorne, J. Follicular helper T cells are essential for the elimination of plasmodium infection. EBioMedicine 2017, 24, 216–230. [Google Scholar] [CrossRef] [Green Version]
  87. Soon, M.S.F.; Lee, H.J.; Engel, J.A.; Straube, J.; Thomas, B.S.; Pernold, C.P.S.; Clarke, L.S.; Laohamonthonkul, P.; Haldar, R.N.; Williams, C.G.; et al. Transcriptome dynamics of CD4+ T cells during malaria maps gradual transit from effector to memory. Nat. Immunol. 2020, 21, 1597–1610. [Google Scholar] [CrossRef] [PubMed]
  88. Deroost, K.; Langhorne, J. Gamma/delta T cells and their role in protection against malaria. Front. Immunol. 2018, 9, 2973. [Google Scholar] [CrossRef] [PubMed]
  89. Goodier, M.R.; Wolf, A.S.; Riley, E.M. Differentiation and adaptation of natural killer cells for anti-malarial immunity. Immunol. Rev. 2020, 293, 25–37. [Google Scholar] [CrossRef] [PubMed]
  90. Horne-Debets, J.M.; Faleiro, R.; Karunarathne, D.S.; Liu, X.Q.; Lineburg, K.E.; Poh, C.M.; Grotenbreg, G.M.; Hill, G.R.; MacDonald, K.P.; Good, M.F.; et al. Pd-1 dependent exhaustion of CD8+ T cells drives chronic malaria. Cell Rep. 2013, 5, 1204–1213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Horne-Debets, J.M.; Karunarathne, D.S.; Faleiro, R.J.; Poh, C.M.; Renia, L.; Wykes, M.N. Mice lacking programmed cell death-1 show a role for CD8+ T cells in long-term immunity against blood-stage malaria. Sci. Rep. 2016, 6, 26210. [Google Scholar] [CrossRef] [PubMed]
  92. Imai, T.; Ishida, H.; Suzue, K.; Taniguchi, T.; Okada, H.; Shimokawa, C.; Hisaeda, H. Cytotoxic activities of CD8+ T cells collaborate with macrophages to protect against blood-stage murine malaria. eLife 2015, 4, e04232. [Google Scholar] [CrossRef]
  93. Kaminski, L.C.; Riehn, M.; Abel, A.; Steeg, C.; Yar, D.D.; Addai-Mensah, O.; Aminkiah, F.; Owusu Dabo, E.; Jacobs, T.; Mackroth, M.S. Cytotoxic T cell-derived granzyme B is increased in severe Plasmodium falciparum malaria. Front. Immunol. 2019, 10, 2917. [Google Scholar] [CrossRef] [Green Version]
  94. Riggle, B.A.; Manglani, M.; Maric, D.; Johnson, K.R.; Lee, M.H.; Neto, O.L.A.; Taylor, T.E.; Seydel, K.B.; Nath, A.; Miller, L.H.; et al. CD8+ T cells target cerebrovasculature in children with cerebral malaria. J. Clin. Investig. 2020, 130, 1128–1138. [Google Scholar] [CrossRef] [Green Version]
  95. Belnoue, E.; Kayibanda, M.; Vigario, A.M.; Deschemin, J.C.; van Rooijen, N.; Viguier, M.; Snounou, G.; Renia, L. On the pathogenic role of brain-sequestered alphabeta CD8+ T cells in experimental cerebral malaria. J. Immunol. 2002, 169, 6369–6375. [Google Scholar] [CrossRef] [Green Version]
  96. Engwerda, C.; Belnoue, E.; Gruner, A.C.; Renia, L. Experimental models of cerebral malaria. Curr. Top. Microbiol. Immunol. 2005, 297, 103–143. [Google Scholar]
  97. Renia, L.; Goh, Y.S. Malaria parasites: The great escape. Front. Immunol. 2016, 7, 463. [Google Scholar] [CrossRef] [Green Version]
  98. Scholzen, A.; Mittag, D.; Rogerson, S.J.; Cooke, B.M.; Plebanski, M. Plasmodium falciparum-mediated induction of human CD25hiFoxp3hi CD4 T cells is independent of direct tcr stimulation and requires IL-2, IL-10 and TGFβ. PLoS Pathog. 2009, 5, e1000543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Kurup, S.P.; Obeng-Adjei, N.; Anthony, S.M.; Traore, B.; Doumbo, O.K.; Butler, N.S.; Crompton, P.D.; Harty, J.T. Regulatory T cells impede acute and long-term immunity to blood-stage malaria through CTLA-4. Nat. Med. 2017, 23, 1220–1225. [Google Scholar] [CrossRef] [PubMed]
  100. Nyirenda, T.S.; Molyneux, M.E.; Kenefeck, R.; Walker, L.S.; MacLennan, C.A.; Heyderman, R.S.; Mandala, W.L. T-regulatory cells and inflammatory and inhibitory cytokines in malawian children residing in an area of high and an area of low malaria transmission during acute uncomplicated malaria and in convalescence. J. Pediatr. Infect. Dis. Soc. 2015, 4, 232–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Goncalves, R.M.; Salmazi, K.C.; Santos, B.A.; Bastos, M.S.; Rocha, S.C.; Boscardin, S.B.; Silber, A.M.; Kallas, E.G.; Ferreira, M.U.; Scopel, K.K. CD4+ CD25+ Foxp3+ regulatory T cells, dendritic cells, and circulating cytokines in uncomplicated malaria: Do different parasite species elicit similar host responses? Infect. Immun. 2010, 78, 4763–4772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Todryk, S.M.; Bejon, P.; Mwangi, T.; Plebanski, M.; Urban, B.; Marsh, K.; Hill, A.V.; Flanagan, K.L. Correlation of memory T cell responses against trap with protection from clinical malaria, and CD4 CD25 high T cells with susceptibility in Kenyans. PLoS ONE 2008, 3, e2027. [Google Scholar] [CrossRef] [Green Version]
  103. Illingworth, J.; Butler, N.S.; Roetynck, S.; Mwacharo, J.; Pierce, S.K.; Bejon, P.; Crompton, P.D.; Marsh, K.; Ndungu, F.M. Chronic exposure to Plasmodium falciparum is associated with phenotypic evidence of B and T cell exhaustion. J. Immunol. 2013, 190, 1038–1047. [Google Scholar] [CrossRef] [Green Version]
  104. Van Braeckel-Budimir, N.; Kurup, S.P.; Harty, J.T. Regulatory issues in immunity to liver and blood-stage malaria. Curr. Opin. Immunol. 2016, 42, 91–97. [Google Scholar] [CrossRef]
  105. Boyle, M.J.; Jagannathan, P.; Farrington, L.A.; Eccles-James, I.; Wamala, S.; McIntyre, T.I.; Vance, H.M.; Bowen, K.; Nankya, F.; Auma, A.; et al. Decline of Foxp3+ regulatory CD4 T cells in peripheral blood of children heavily exposed to malaria. PLoS Pathog. 2015, 11, e1005041. [Google Scholar] [CrossRef]
  106. Dups, J.N.; Pepper, M.; Cockburn, I.A. Antibody and B cell responses to Plasmodium sporozoites. Front. Microbiol. 2014, 5, 625. [Google Scholar] [CrossRef] [Green Version]
  107. Blank, A.; Fürle, K.; Jäschke, A.; Mikus, G.; Lehmann, M.; Hüsing, J.; Heiss, K.; Giese, T.; Carter, D.; Böhnlein, E.; et al. Immunization with full-length Plasmodium falciparum merozoite surface protein 1 is safe and elicits functional cytophilic antibodies in a randomized first-in-human trial. NPJ Vaccines 2020, 5, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Aitken, E.H.; Mahanty, S.; Rogerson, S.J. Antibody effector functions in malaria and other parasitic diseases: A few needles and many haystacks. Immunol. Cell Biol. 2020, 98, 264–275. [Google Scholar] [CrossRef] [PubMed]
  109. Hermsen, C.C.; Verhage, D.F.; Telgt, D.S.; Teelen, K.; Bousema, J.T.; Roestenberg, M.; Bolad, A.; Berzins, K.; Corradin, G.; Leroy, O. Glutamate-rich protein (GLURP) induces antibodies that inhibit in vitro growth of Plasmodium falciparum in a phase 1 malaria vaccine trial. Vaccine 2007, 25, 2930–2940. [Google Scholar] [CrossRef] [PubMed]
  110. Plebanski, M.; Locke, E.; Kazura, J.W.; Coppel, R.L. Malaria vaccines: Into a mirror, darkly? Trends Parasitol. 2008, 24, 532–536. [Google Scholar] [CrossRef]
  111. Gupta, S.; Snow, R.W.; Donnelly, C.A.; Marsh, K.; Newbold, C. Immunity to non-cerebral severe malaria is acquired after one or two infections. Nat. Med. 1999, 5, 340–343. [Google Scholar] [CrossRef]
  112. Julien, J.-P.; Wardemann, H. Antibodies against Plasmodium falciparum malaria at the molecular level. Nat. Rev. Immunol. 2019, 19, 761–775. [Google Scholar] [CrossRef] [PubMed]
  113. Vanderberg, J.P.; Frevert, U. Intravital microscopy demonstrating antibody-mediated immobilisation of Plasmodium berghei sporozoites injected into skin by mosquitoes. Int. J. Parasitol. 2004, 34, 991–996. [Google Scholar] [CrossRef]
  114. Aliprandini, E.; Tavares, J.; Panatieri, R.H.; Thiberge, S.; Yamamoto, M.M.; Silvie, O.; Ishino, T.; Yuda, M.; Dartevelle, S.; Traincard, F.; et al. Cytotoxic anti-circumsporozoite antibodies target malaria sporozoites in the host skin. Nat. Microbiol. 2018, 3, 1224–1233. [Google Scholar] [CrossRef]
  115. Schwenk, R.; Lumsden, J.M.; Rein, L.E.; Juompan, L.; Kester, K.E.; Heppner, D.G.; Krzych, U. Immunization with the RTS,S/AS malaria vaccine induces IFN-γ+ CD4 T cells that recognize only discrete regions of the circumsporozoite protein and these specificities are maintained following booster immunizations and challenge. Vaccine 2011, 29, 8847–8854. [Google Scholar] [CrossRef]
  116. Nudelman, S.; Renia, L.; Charoenvit, Y.; Yuan, L.; Miltgen, F.; Beaudoin, R.L.; Mazier, D. Dual action of anti-sporozoite antibodies in vitro. J. Immunol. 1989, 143, 996–1000. [Google Scholar]
  117. Vidarsson, G.; Dekkers, G.; Rispens, T. IgG subclasses and allotypes: From structure to effector functions. Front. Immunol. 2014, 5, 520. [Google Scholar] [CrossRef] [Green Version]
  118. Boyle, M.J.; Reiling, L.; Feng, G.; Langer, C.; Osier, F.H.; Aspeling-Jones, H.; Cheng, Y.S.; Stubbs, J.; Tetteh, K.K.; Conway, D.J.; et al. Human antibodies fix complement to inhibit Plasmodium falciparum invasion of erythrocytes and are associated with protection against malaria. Immunity 2015, 42, 580–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Osier, F.H.A.; Feng, G.; Boyle, M.J.; Langer, C.; Zhou, J.; Richards, J.S.; McCallum, F.J.; Reiling, L.; Jaworowski, A.; Anders, R.F.; et al. Opsonic phagocytosis of Plasmodium falciparummerozoites: Mechanism in human immunity and a correlate of protection against malaria. BMC Med. 2014, 12, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Boyle, M.J.; Chan, J.A.; Handayuni, I.; Reiling, L.; Feng, G.; Hilton, A.; Kurtovic, L.; Oyong, D.; Piera, K.A.; Barber, B.E.; et al. IgM in human immunity to Plasmodium falciparum malaria. Sci. Adv. 2019, 5, eaax4489. [Google Scholar] [CrossRef] [Green Version]
  121. Barfod, L.; Dalgaard, M.B.; Pleman, S.T.; Ofori, M.F.; Pleass, R.J.; Hviid, L. Evasion of immunity to Plasmodium falciparum malaria by IgM masking of protective IgG epitopes in infected erythrocyte surface-exposed pfemp1. Proc. Natl. Acad. Sci. USA 2011, 108, 12485–12490. [Google Scholar] [CrossRef] [Green Version]
  122. Hopp, C.S.; Diouf, A.; Miura, K.; Boswell, K.; Sekar, P.; Skinner, J.; Tipton, C.M.; Chambers, M.; Andrews, S.; Tan, J.; et al. Plasmodium falciparum-specific IgM B cells dominate in children, expand with malaria and produce parasite inhibitory IgM. bioRxiv 2020. [Google Scholar] [CrossRef]
  123. Renia, L.; Mattei, D.; Goma, J.; Pied, S.; Dubois, P.; Miltgen, F.; Nussler, A.; Matile, H.; Menegaux, F.; Gentilini, M.; et al. A malaria heat-shock-like determinant expressed on the infected hepatocyte surface is the target of antibody-dependent cell-mediated cytotoxic mechanisms by nonparenchymal liver cells. Eur. J. Immunol. 1990, 20, 1445–1449. [Google Scholar] [CrossRef]
  124. Dutta, S.; Kaushal, D.C.; Ware, L.A.; Puri, S.K.; Kaushal, N.A.; Narula, A.; Upadhyaya, D.S.; Lanar, D.E. Merozoite surface protein 1 of plasmodium vivax induces a protective response against plasmodium cynomolgi challenge in rhesus monkeys. Infect. Immun. 2005, 73, 5936–5944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Jiang, L.; López-Barragán, M.J.; Jiang, H.; Mu, J.; Gaur, D.; Zhao, K.; Felsenfeld, G.; Miller, L.H. Epigenetic control of the variable expression of a Plasmodium falciparum receptor protein for erythrocyte invasion. Proc. Natl. Acad. Sci. USA 2010, 107, 2224–2229. [Google Scholar] [CrossRef] [Green Version]
  126. Michon, P.; Fraser, T.; Adams, J.H. Naturally acquired and vaccine-elicited antibodies block erythrocyte cytoadherence of the plasmodium vivax duffy binding protein. Infect. Immun. 2000, 68, 3164–3171. [Google Scholar] [CrossRef] [Green Version]
  127. Marsh, K.; Kinyanjui, S. Immune effector mechanisms in malaria. Parasite Immunol. 2006, 28, 51–60. [Google Scholar] [CrossRef]
  128. Bouharoun-Tayoun, H.; Oeuvray, C.; Lunel, F.; Druilhe, P. Mechanisms underlying the monocyte-mediated antibody-dependent killing of Plasmodium falciparum asexual blood stages. J. Exp. Med. 1995, 182, 409–418. [Google Scholar] [CrossRef] [PubMed]
  129. Bouharoun-Tayoun, H.; Attanath, P.; Sabchareon, A.; Chongsuphajaisiddhi, T.; Druilhe, P. Antibodies that protect humans against Plasmodium falciparum blood stages do not on their own inhibit parasite growth and invasion in vitro, but act in cooperation with monocytes. J. Exp. Med. 1990, 172, 1633–1641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Su, X.Z.; Heatwole, V.M.; Wertheimer, S.P.; Guinet, F.; Herrfeldt, J.A.; Peterson, D.S.; Ravetch, J.A.; Wellems, T.E. The large diverse gene family var encodes proteins involved in cytoadherence and antigenic variation of Plasmodium falciparum-infected erythrocytes. Cell 1995, 82, 89–100. [Google Scholar] [CrossRef] [Green Version]
  131. Cheng, Q.; Cloonan, N.; Fischer, K.; Thompson, J.; Waine, G.; Lanzer, M.; Saul, A. Stevor and rif are Plasmodium falciparum multicopy gene families which potentially encode variant antigens. Mol. Biochem. Parasitol. 1998, 97, 161–176. [Google Scholar] [CrossRef]
  132. Kyes, S.A.; Rowe, J.A.; Kriek, N.; Newbold, C.I. Rifins: A second family of clonally variant proteins expressed on the surface of red cells infected with Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 1999, 96, 9333–9338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Chan, J.A.; Fowkes, F.J.; Beeson, J.G. Surface antigens of Plasmodium falciparum-infected erythrocytes as immune targets and malaria vaccine candidates. Cell. Mol. Life Sci. 2014, 71, 3633–3657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Tan, J.; Pieper, K.; Piccoli, L.; Abdi, A.; Perez, M.F.; Geiger, R.; Tully, C.M.; Jarrossay, D.; Maina Ndungu, F.; Wambua, J.; et al. A LAIR1 insertion generates broadly reactive antibodies against malaria variant antigens. Nature 2016, 529, 105–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Olivier, M.; Van Den Ham, K.; Shio, M.T.; Kassa, F.A.; Fougeray, S. Malarial pigment hemozoin and the innate inflammatory response. Front. Immunol. 2014, 5, 25. [Google Scholar] [CrossRef] [PubMed]
  136. Schofield, L.; Tachado, S.D. Regulation of host cell function by glycosylphosphatidylinositols of the parasitic protozoa. Immunol. Cell Biol. 1996, 74, 555–563. [Google Scholar] [CrossRef]
  137. Chang, Z.; Jiang, N.; Zhang, Y.; Lu, H.; Yin, J.; Wahlgren, M.; Cheng, X.; Cao, Y.; Chen, Q. The tatd-like dnase of plasmodium is a virulence factor and a potential malaria vaccine candidate. Nat. Commun. 2016, 7, 11537. [Google Scholar] [CrossRef] [Green Version]
  138. Bhatt, T.K.; Khan, S.; Dwivedi, V.P.; Banday, M.M.; Sharma, A.; Chandele, A.; Camacho, N.; Ribas de Pouplana, L.; Wu, Y.; Craig, A.G.; et al. Malaria parasite tyrosyl-tRNA synthetase secretion triggers pro-inflammatory responses. Nat. Commun. 2011, 2, 530. [Google Scholar] [CrossRef] [Green Version]
  139. Schofield, L.; Hewitt, M.C.; Evans, K.; Siomos, M.A.; Seeberger, P.H. Synthetic GPI as a candidate anti-toxic vaccine in a model of malaria. Nature 2002, 418, 785–789. [Google Scholar] [CrossRef] [PubMed]
  140. De Jong, R.M.; Tebeje, S.K.; Meerstein-Kessel, L.; Tadesse, F.G.; Jore, M.M.; Stone, W.; Bousema, T. Immunity against sexual stage Plasmodium falciparum and plasmodium vivax parasites. Immunol. Rev. 2020, 293, 190–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Dantzler, K.W.; Ma, S.; Ngotho, P.; Stone, W.J.R.; Tao, D.; Rijpma, S.; De Niz, M.; Nilsson Bark, S.K.; Jore, M.M.; Raaijmakers, T.K.; et al. Naturally acquired immunity against immature Plasmodium falciparum gametocytes. Sci. Transl. Med. 2019, 11, eaav3963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Read, D.; Lensen, A.H.; Begarnie, S.; Haley, S.; Raza, A.; Carter, R. Transmission-blocking antibodies against multiple, non-variant target epitopes of the Plasmodium falciparum gamete surface antigen Pfs230 are all complement-fixing. Parasite Immunol. 1994, 16, 511–519. [Google Scholar] [CrossRef]
  143. Blagborough, A.M.; Sinden, R.E. Plasmodium berghei HAP2 induces strong malaria transmission-blocking immunity in vivo and in vitro. Vaccine 2009, 27, 5187–5194. [Google Scholar] [CrossRef]
  144. Volohonsky, G.; Steinert, S.; Levashina, E.A. Focusing on complement in the antiparasitic defense of mosquitoes. Trends Parasitol. 2010, 26, 1–3. [Google Scholar] [CrossRef]
  145. Sauerwein, R.W.; Richie, T.L. Malaria vaccines getting close to clinical reality. Vaccine 2015, 33, 7423–7424. [Google Scholar] [CrossRef] [Green Version]
  146. Kwenti, T.E.; Kukwah, T.A.; Kwenti, T.D.B.; Nyassa, B.R.; Dilonga, M.H.; Enow-Orock, G.; Tendongfor, N.; Anong, N.D.; Wanji, S.; Njunda, L.A.; et al. Comparative analysis of IgG and IgG subclasses against Plasmodium falciparum MSP-119 in children from five contrasting bioecological zones of cameroon. Malar. J. 2019, 18, 16. [Google Scholar] [CrossRef] [Green Version]
  147. Dobano, C.; Santano, R.; Vidal, M.; Jimenez, A.; Jairoce, C.; Ubillos, I.; Dosoo, D.; Aguilar, R.; Williams, N.A.; Diez-Padrisa, N.; et al. Differential patterns of IgG subclass responses to Plasmodium falciparum antigens in relation to malaria protection and RTS,S vaccination. Front. Immunol. 2019, 10, 439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Stanisic, D.I.; Richards, J.S.; McCallum, F.J.; Michon, P.; King, C.L.; Schoepflin, S.; Gilson, P.R.; Murphy, V.J.; Anders, R.F.; Mueller, I.; et al. Immunoglobulin g subclass-specific responses against Plasmodium falciparum merozoite antigens are associated with control of parasitemia and protection from symptomatic illness. Infect. Immun. 2009, 77, 1165–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Abagna, H.B.; Acquah, F.K.; Okonu, R.; Aryee, N.A.; Theisen, M.; Amoah, L.E. Assessment of the quality and quantity of naturally induced antibody responses to EBA175RIII–V in ghanaian children living in two communities with varying malaria transmission patterns. Malar. J. 2018, 17, 14. [Google Scholar] [CrossRef] [Green Version]
  150. Kana, I.H.; Garcia-Senosiain, A.; Singh, S.K.; Tiendrebeogo, R.W.; Chourasia, B.K.; Malhotra, P.; Sharma, S.K.; Das, M.K.; Singh, S.; Adu, B.; et al. Cytophilic antibodies against key Plasmodium falciparum blood stage antigens contribute to protection against clinical malaria in a high transmission region of eastern India. J. Infect. Dis. 2018, 218, 956–965. [Google Scholar] [CrossRef] [PubMed]
  151. Lusingu, J.P.A.; Vestergaard, L.S.; Alifrangis, M.; Mmbando, B.P.; Theisen, M.; Kitua, A.Y.; Lemnge, M.M.; Theander, T.G. Cytophilic antibodies to Plasmodium falciparum glutamate rich protein are associated with malaria protection in an area of holoendemic transmission. Malar. J. 2005, 4, 48. [Google Scholar] [CrossRef] [Green Version]
  152. Courtin, D.; Oesterholt, M.; Huismans, H.; Kusi, K.; Milet, J.; Badaut, C.; Gaye, O.; Roeffen, W.; Remarque, E.J.; Sauerwein, R.; et al. The quantity and quality of african children’s IgG responses to merozoite surface antigens reflect protection against Plasmodium falciparum malaria. PLoS ONE 2009, 4, e7590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. De Souza, J.B. Protective immunity against malaria after vaccination. Parasite Immunol. 2014, 36, 131–139. [Google Scholar] [CrossRef] [Green Version]
  154. Pinkevych, M.; Petravic, J.; Chelimo, K.; Kazura, J.W.; Moormann, A.M.; Davenport, M.P. The dynamics of naturally acquired immunity to Plasmodium falciparum infection. PLoS Comp. Biol. 2012, 8, e1002729. [Google Scholar] [CrossRef]
  155. John, C.C.; Moormann, A.M.; Pregibon, D.C.; Sumba, P.O.; McHugh, M.M.; Narum, D.L.; Lanar, D.E.; Schluchter, M.D.; Kazura, J.W. Correlation of high levels of antibodies to multiple pre-erythrocytic Plasmodium falciparum antigens and protection from infection. Am. J. Trop. Med. Hyg. 2005, 73, 222–228. [Google Scholar] [CrossRef]
  156. John, C.C.; Tande, A.J.; Moormann, A.M.; Sumba, P.O.; Lanar, D.E.; Min, X.M.; Kazura, J.W. Antibodies to pre-erythrocytic Plasmodium falciparum antigens and risk of clinical malaria in Kenyan children. J. Infect. Dis. 2008, 197, 519–526. [Google Scholar] [CrossRef] [Green Version]
  157. Dobbs, K.R.; Dent, A.E. Plasmodium malaria and antimalarial antibodies in the first year of life. Parasitology 2016, 143, 129–138. [Google Scholar] [CrossRef]
  158. Osier, F.H.; Fegan, G.; Polley, S.D.; Murungi, L.; Verra, F.; Tetteh, K.K.; Lowe, B.; Mwangi, T.; Bull, P.C.; Thomas, A.W.; et al. Breadth and magnitude of antibody responses to multiple Plasmodium falciparum merozoite antigens are associated with protection from clinical malaria. Infect. Immun. 2008, 76, 2240–2248. [Google Scholar] [CrossRef] [Green Version]
  159. Dent, A.E.; Nakajima, R.; Liang, L.; Baum, E.; Moormann, A.M.; Sumba, P.O.; Vulule, J.; Babineau, D.; Randall, A.; Davies, D.H.; et al. Plasmodium falciparum protein microarray antibody profiles correlate with protection from symptomatic malaria in Kenya. J. Infect. Dis. 2015, 212, 1429–1438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Stanisic, D.I.; Fowkes, F.J.; Koinari, M.; Javati, S.; Lin, E.; Kiniboro, B.; Richards, J.S.; Robinson, L.J.; Schofield, L.; Kazura, J.W.; et al. Acquisition of antibodies against Plasmodium falciparum merozoites and malaria immunity in young children and the influence of age, force of infection, and magnitude of response. Infect. Immun. 2015, 83, 646–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Kusi, K.A.; Manu, E.A.; Manful Gwira, T.; Kyei-Baafour, E.; Dickson, E.K.; Amponsah, J.A.; Remarque, E.J.; Faber, B.W.; Kocken, C.H.M.; Dodoo, D.; et al. Variations in the quality of malaria-specific antibodies with transmission intensity in a seasonal malaria transmission area of northern Ghana. PLoS ONE 2017, 12, e0185303. [Google Scholar] [CrossRef] [PubMed]
  162. McGregor, I.A. The Passive Transfer of Human Malarial Immunity. Am. J. Trop. Med. Hyg. 1964, 13, 237–239. [Google Scholar] [CrossRef] [PubMed]
  163. Cohen, S.; McGregor, I.A.; Carrington, S. Gamma-globulin and acquired immunity to human malaria. Nature 1961, 192, 733–737. [Google Scholar] [CrossRef] [PubMed]
  164. Schwartz, L.; Brown, G.V.; Genton, B.; Moorthy, V.S. A review of malaria vaccine clinical projects based on the who rainbow table. Malar. J. 2012, 11, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Genton, B. Malaria vaccines: A toy for travelers or a tool for eradication? Expert Rev. Vaccines 2008, 7, 597–611. [Google Scholar] [CrossRef]
  166. Draper, S.J.; Angov, E.; Horii, T.; Miller, L.H.; Srinivasan, P.; Theisen, M.; Biswas, S. Recent advances in recombinant protein-based malaria vaccines. Vaccine 2015, 33, 7433–7443. [Google Scholar] [CrossRef] [Green Version]
  167. Coppel, R.L. Vaccinating with the genome: A sisyphean task? Trends Parasitol. 2009, 25, 205–212. [Google Scholar] [CrossRef]
  168. Florens, L.; Washburn, M.P.; Raine, J.D.; Anthony, R.M.; Grainger, M.; Haynes, J.D.; Moch, J.K.; Muster, N.; Sacci, J.B.; Tabb, D.L.; et al. A proteomic view of the Plasmodium falciparum life cycle. Nature 2002, 419, 520–526. [Google Scholar] [CrossRef]
  169. Draper, S.J.; Sack, B.K.; King, C.R.; Nielsen, C.M.; Rayner, J.C.; Higgins, M.K.; Long, C.A.; Seder, R.A. Malaria vaccines: Recent advances and new horizons. Cell Host Microbe 2018, 24, 43–56. [Google Scholar] [CrossRef] [Green Version]
  170. O’Hagan, D.T.; Valiante, N.M. Recent advances in the discovery and delivery of vaccine adjuvants. Nat. Rev. Drug Discov. 2003, 2, 727–735. [Google Scholar] [CrossRef]
  171. Guy, B. The perfect mix: Recent progress in adjuvant research. Nat. Rev. Microbiol. 2007, 5, 505–517. [Google Scholar] [CrossRef] [PubMed]
  172. Coffman, R.L.; Sher, A.; Seder, R.A. Vaccine adjuvants: Putting innate immunity to work. Immunity 2010, 33, 492–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Reed, S.G.; Orr, M.T.; Fox, C.B. Key roles of adjuvants in modern vaccines. Nat. Med. 2013, 19, 1597–1608. [Google Scholar] [CrossRef] [PubMed]
  174. Bonam, S.R.; Partidos, C.D.; Halmuthur, S.K.M.; Muller, S. An overview of novel adjuvants designed for improving vaccine efficacy. Trends Pharmacol. Sci. 2017, 38, 771–793. [Google Scholar] [CrossRef]
  175. Coelho, C.H.; Doritchamou, J.Y.A.; Zaidi, I.; Duffy, P.E. Advances in malaria vaccine development: Report from the 2017 malaria vaccine symposium. NPJ Vaccines 2017, 2, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Spellberg, B.; Edwards, J.E., Jr. Type 1/type 2 immunity in infectious diseases. Clin. Infect. Dis. 2001, 32, 76–102. [Google Scholar] [CrossRef]
  177. Hansen, D.S.; Obeng-Adjei, N.; Ly, A.; Ioannidis, L.J.; Crompton, P.D. Emerging concepts in t follicular helper cell responses to malaria. Int. J. Parasitol. 2017, 47, 105–110. [Google Scholar] [CrossRef]
  178. Radtke, A.J.; Anderson, C.F.; Riteau, N.; Rausch, K.; Scaria, P.; Kelnhofer, E.R.; Howard, R.F.; Sher, A.; Germain, R.N.; Duffy, P. Adjuvant and carrier protein-dependent t-cell priming promotes a robust antibody response against the Plasmodium falciparum Pfs25 vaccine candidate. Sci. Rep. 2017, 7, 40312. [Google Scholar] [CrossRef] [PubMed]
  179. Figueiredo, M.M.; Costa, P.A.C.; Diniz, S.Q.; Henriques, P.M.; Kano, F.S.; Tada, M.S.; Pereira, D.B.; Soares, I.S.; Martins-Filho, O.A.; Jankovic, D.; et al. T follicular helper cells regulate the activation of B lymphocytes and antibody production during plasmodium vivax infection. PLoS Pathog. 2017, 13, e1006484. [Google Scholar] [CrossRef]
  180. Moon, J.J.; Suh, H.; Li, A.V.; Ockenhouse, C.F.; Yadava, A.; Irvine, D.J. Enhancing humoral responses to a malaria antigen with nanoparticle vaccines that expand Tfh cells and promote germinal center induction. Proc. Natl. Acad. Sci. USA 2012, 109, 1080–1085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Goh, Y.S.; Peng, K.; Chia, W.N.; Siau, A.; Chotivanich, K.; Gruner, A.C.; Preiser, P.; Mayxay, M.; Pukrittayakamee, S.; Sriprawat, K.; et al. Neutralizing antibodies against Plasmodium falciparum associated with successful cure after drug therapy. PLoS ONE 2016, 11, e0159347. [Google Scholar] [CrossRef]
  182. Kurtovic, L.; Behet, M.C.; Feng, G.; Reiling, L.; Chelimo, K.; Dent, A.E.; Mueller, I.; Kazura, J.W.; Sauerwein, R.W.; Fowkes, F.J.I.; et al. Human antibodies activate complement against Plasmodium falciparum sporozoites, and are associated with protection against malaria in children. BMC Med. 2018, 16, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Radosevic, K.; Rodriguez, A.; Lemckert, A.A.; van der Meer, M.; Gillissen, G.; Warnar, C.; von Eyben, R.; Pau, M.G.; Goudsmit, J. The Th1 immune response to Plasmodium falciparum circumsporozoite protein is boosted by adenovirus vectors 35 and 26 with a homologous insert. Clin. Vaccine Immunol. 2010, 17, 1687–1694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Tom, J.K.; Albin, T.J.; Manna, S.; Moser, B.A.; Steinhardt, R.C.; Esser-Kahn, A.P. Applications of immunomodulatory immune synergies to adjuvant discovery and vaccine development. Trends Biotechnol. 2019, 37, 373–388. [Google Scholar] [CrossRef] [PubMed]
  185. Del Giudice, G.; Rappuoli, R.; Didierlaurent, A.M. Correlates of adjuvanticity: A review on adjuvants in licensed vaccines. Semin. Immunol. 2018, 39, 14–21. [Google Scholar] [CrossRef]
  186. Garcon, N.; Di Pasquale, A. From discovery to licensure, the adjuvant system story. Hum. Vaccin. Immunother. 2017, 13, 19–33. [Google Scholar] [CrossRef] [PubMed]
  187. Draper, S.J.; Moore, A.C.; Goodman, A.L.; Long, C.A.; Holder, A.A.; Gilbert, S.C.; Hill, F.; Hill, A.V. Effective induction of high-titer antibodies by viral vector vaccines. Nat. Med. 2008, 14, 819–821. [Google Scholar] [CrossRef]
  188. HogenEsch, H.; O’Hagan, D.T.; Fox, C.B. Optimizing the utilization of aluminum adjuvants in vaccines: You might just get what you want. NPJ Vaccines 2018, 3, 51. [Google Scholar] [CrossRef] [PubMed]
  189. Aimanianda, V.; Haensler, J.; Lacroix-Desmazes, S.; Kaveri, S.V.; Bayry, J. Novel cellular and molecular mechanisms of induction of immune responses by aluminum adjuvants. Trends Pharmacol. Sci. 2009, 30, 287–295. [Google Scholar] [CrossRef] [PubMed]
  190. Mata, E.; Salvador, A.; Igartua, M.; Hernandez, R.M.; Pedraz, J.L. Malaria vaccine adjuvants: Latest update and challenges in preclinical and clinical research. Biomed Res. Int. 2013, 2013, 282913. [Google Scholar] [CrossRef]
  191. Near, K.A.; Stowers, A.W.; Jankovic, D.; Kaslow, D.C. Improved immunogenicity and efficacy of the recombinant 19-kilodalton merozoite surface protein 1 by the addition of oligodeoxynucleotide and aluminum hydroxide gel in a murine malaria vaccine model. Infect. Immun. 2002, 70, 692–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Roeffen, W.; Theisen, M.; van de Vegte-Bolmer, M.; van Gemert, G.; Arens, T.; Andersen, G.; Christiansen, M.; Sevargave, L.; Singh, S.K.; Kaviraj, S.; et al. Transmission-blocking activity of antibodies to Plasmodium falciparum GLURP.10C chimeric protein formulated in different adjuvants. Malar. J. 2015, 14, 443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Nalla, N.; Pallavi, P.; Reddy, B.S.; Miryala, S.; Kumar, V.N.; Mahboob, M.; Halmuthur, M.S.K. Design, synthesis and immunological evaluation of 1,2,3-triazole-tethered carbohydrate–Pam3Cys conjugates as TLR2 agonists. Biorg. Med. Chem. 2015, 23, 5846–5855. [Google Scholar] [CrossRef] [PubMed]
  194. Bhunia, D.; Pallavi, P.M.; Bonam, S.R.; Reddy, S.A.; Verma, Y.; Halmuthur, M.S.K. Design, synthesis, and evaluation of novel 1,2,3-triazole-tethered glycolipids as vaccine adjuvants. Arch. Pharm. 2015, 348, 689–703. [Google Scholar] [CrossRef] [PubMed]
  195. Chioato, A.; Noseda, E.; Felix, S.; Stevens, M.; Del Giudice, G.; Fitoussi, S.; Kleinschmidt, A. Influenza and meningococcal vaccinations are effective in healthy subjects treated with the interleukin-1β-blocking antibody canakinumab: Results of an open-label, parallel group, randomized, single-center study. Clin. Vaccine Immunol. 2010, 17, 1952–1957. [Google Scholar] [CrossRef] [Green Version]
  196. Liang, F.; Lindgren, G.; Sandgren, K.J.; Thompson, E.A.; Francica, J.R.; Seubert, A.; De Gregorio, E.; Barnett, S.; O’Hagan, D.T.; Sullivan, N.J.; et al. Vaccine priming is restricted to draining lymph nodes and controlled by adjuvant-mediated antigen uptake. Sci. Transl. Med. 2017, 9, eaal2094. [Google Scholar] [CrossRef]
  197. López, J.A.; Weilenman, C.; Audran, R.; Roggero, M.A.; Bonelo, A.; Tiercy, J.-M.; Spertini, F.; Corradin, G. A synthetic malaria vaccine elicits a potent CD8+ and CD4+ T lymphocyte immune response in humans. Implications for vaccination strategies. Eur. J. Immunol. 2001, 31, 1989–1998. [Google Scholar] [CrossRef]
  198. Sirima, S.B.; Richert, L.; Chene, A.; Konate, A.T.; Campion, C.; Dechavanne, S.; Semblat, J.P.; Benhamouda, N.; Bahuaud, M.; Loulergue, P.; et al. Primvac vaccine adjuvanted with alhydrogel or GLA-SE to prevent placental malaria: A first-in-human, randomised, double-blind, placebo-controlled study. Lancet Infect. Dis. 2020, 20, 585–597. [Google Scholar] [CrossRef]
  199. Carter, R.; Mendis, K.N.; Miller, L.H.; Molineaux, L.; Saul, A. Malaria transmission-blocking vaccines—How can their development be supported? Nat. Med. 2000, 6, 241–244. [Google Scholar] [CrossRef] [PubMed]
  200. Sagara, I.; Healy, S.A.; Assadou, M.H.; Gabriel, E.E.; Kone, M.; Sissoko, K.; Tembine, I.; Guindo, M.A.; Doucoure, M.; Niare, K.; et al. Safety and immunogenicity of Pfs25h-EPA/Alhydrogel, a transmission-blocking vaccine against Plasmodium falciparum: A randomised, double-blind, comparator-controlled, dose-escalation study in healthy malian adults. Lancet Infect. Dis. 2018, 18, 969–982. [Google Scholar] [CrossRef]
  201. O’Hagan, D.T.; Fox, C.B. New generation adjuvants–from empiricism to rational design. Vaccine 2015, 33, B14–B20. [Google Scholar] [CrossRef]
  202. Gregoriadis, G. Immunological adjuvants: A role for liposomes. Immunol. Today 1990, 11, 89–97. [Google Scholar] [CrossRef]
  203. Peek, L.J.; Middaugh, C.R.; Berkland, C. Nanotechnology in vaccine delivery. Adv. Drug Deliv. Rev. 2008, 60, 915–928. [Google Scholar] [CrossRef] [PubMed]
  204. Rosenkrands, I.; Vingsbo-Lundberg, C.; Bundgaard, T.J.; Lindenstrom, T.; Enouf, V.; van der Werf, S.; Andersen, P.; Agger, E.M. Enhanced humoral and cell-mediated immune responses after immunization with trivalent influenza vaccine adjuvanted with cationic liposomes. Vaccine 2011, 29, 6283–6291. [Google Scholar] [CrossRef] [PubMed]
  205. Didierlaurent, A.M.; Laupeze, B.; Di Pasquale, A.; Hergli, N.; Collignon, C.; Garcon, N. Adjuvant system AS01: Helping to overcome the challenges of modern vaccines. Expert Rev. Vaccines 2017, 16, 55–63. [Google Scholar] [CrossRef] [Green Version]
  206. Coccia, M.; Collignon, C.; Herve, C.; Chalon, A.; Welsby, I.; Detienne, S.; van Helden, M.J.; Dutta, S.; Genito, C.J.; Waters, N.C.; et al. Cellular and molecular synergy in AS01-adjuvanted vaccines results in an early ifngamma response promoting vaccine immunogenicity. NPJ Vaccines 2017, 2, 25. [Google Scholar] [CrossRef]
  207. Didierlaurent, A.M.; Collignon, C.; Bourguignon, P.; Wouters, S.; Fierens, K.; Fochesato, M.; Dendouga, N.; Langlet, C.; Malissen, B.; Lambrecht, B.N.; et al. Enhancement of adaptive immunity by the human vaccine adjuvant AS01 depends on activated dendritic cells. J. Immunol. 2014, 193, 1920–1930. [Google Scholar] [CrossRef] [Green Version]
  208. Leroux-Roels, I.; Forgus, S.; De Boever, F.; Clement, F.; Demoitie, M.A.; Mettens, P.; Moris, P.; Ledent, E.; Leroux-Roels, G.; Ofori-Anyinam, O.; et al. Improved CD4+ T cell responses to mycobacterium tuberculosis in ppd-negative adults by M72/AS01 as compared to the M72/AS02 and MTB72F/AS02 tuberculosis candidate vaccine formulations: A randomized trial. Vaccine 2013, 31, 2196–2206. [Google Scholar] [CrossRef] [PubMed]
  209. Fox, C.B.; Baldwin, S.L.; Vedvick, T.S.; Angov, E.; Reed, S.G. Effects on immunogenicity by formulations of emulsion-based adjuvants for malaria vaccines. Clin. Vaccine Immunol. 2012, 19, 1633–1640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Pallikkuth, S.; Chaudhury, S.; Lu, P.; Pan, L.; Jongert, E.; Wille-Reece, U.; Pahwa, S. A delayed fractionated dose RTS,S AS01 vaccine regimen mediates protection via improved t follicular helper and B cell responses. eLife 2020, 9, e51889. [Google Scholar] [CrossRef] [PubMed]
  211. Marty-Roix, R.; Vladimer, G.I.; Pouliot, K.; Weng, D.; Buglione-Corbett, R.; West, K.; MacMicking, J.D.; Chee, J.D.; Wang, S.X.; Lu, S.; et al. Identification of QS-21 as an inflammasome-activating molecular component of saponin adjuvants. J. Biol. Chem. 2016, 291, 1123–1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Regules, J.A.; Cummings, J.F.; Ockenhouse, C.F. The RTS,S vaccine candidate for malaria. Expert Rev. Vaccines 2011, 10, 589–599. [Google Scholar] [CrossRef] [Green Version]
  213. Coler, R.N.; Bertholet, S.; Moutaftsi, M.; Guderian, J.A.; Windish, H.P.; Baldwin, S.L.; Laughlin, E.M.; Duthie, M.S.; Fox, C.B.; Carter, D.; et al. Development and characterization of synthetic glucopyranosyl lipid adjuvant system as a vaccine adjuvant. PLoS ONE 2011, 6, e16333. [Google Scholar] [CrossRef] [Green Version]
  214. Sravanthi, V.; Pallavi, M.C.P.; Bonam, S.R.; Sathyabama, S.; Kumar, H.M.S. Oleic acid nanoemulsion for nasal vaccination: Impact on adjuvanticity based immune response. J. Drug Deliv. Sci. Technol. 2015, 28, 56–63. [Google Scholar] [CrossRef]
  215. Kantipakala, R.; Bonam, S.R.; Vemireddy, S.; Miryala, S.; Halmuthur, M.S. Squalane-based emulsion vaccine delivery system: Composition with murabutide activate Th1 response. Pharm. Dev. Technol. 2019, 24, 269–275. [Google Scholar] [CrossRef] [PubMed]
  216. Mosca, F.; Tritto, E.; Muzzi, A.; Monaci, E.; Bagnoli, F.; Iavarone, C.; O’Hagan, D.; Rappuoli, R.; De Gregorio, E. Molecular and cellular signatures of human vaccine adjuvants. Proc. Natl. Acad. Sci. USA 2008, 105, 10501–10506. [Google Scholar] [CrossRef] [Green Version]
  217. Ott, G.; Barchfeld, G.L.; Chernoff, D.; Radhakrishnan, R.; van Hoogevest, P.; Van Nest, G. Vaccine Design; Springer: Berlin, Germany, 1995; pp. 277–296. [Google Scholar]
  218. Corradin, G.; Giudice, G.D. Novel adjuvants for vaccines. Curr. Med. Chem. 2005, 4, 185–191. [Google Scholar] [CrossRef] [Green Version]
  219. Wack, A.; Baudner, B.C.; Hilbert, A.K.; Manini, I.; Nuti, S.; Tavarini, S.; Scheffczik, H.; Ugozzoli, M.; Singh, M.; Kazzaz, J.; et al. Combination adjuvants for the induction of potent, long-lasting antibody and T-cell responses to influenza vaccine in mice. Vaccine 2008, 26, 552–561. [Google Scholar] [CrossRef] [PubMed]
  220. Seubert, A.; Monaci, E.; Pizza, M.; O’Hagan, D.T.; Wack, A. The adjuvants aluminum hydroxide and MF59 induce monocyte and granulocyte chemoattractants and enhance monocyte differentiation toward dendritic cells. J. Immunol. 2008, 180, 5402–5412. [Google Scholar] [CrossRef]
  221. Baudner, B.C.; Ronconi, V.; Casini, D.; Tortoli, M.; Kazzaz, J.; Singh, M.; Hawkins, L.D.; Wack, A.; O’Hagan, D.T. MF59 emulsion is an effective delivery system for a synthetic TLR4 agonist (E6020). Pharm. Res. 2009, 26, 1477–1485. [Google Scholar] [CrossRef] [PubMed]
  222. Roestenberg, M.; Remarque, E.; de Jonge, E.; Hermsen, R.; Blythman, H.; Leroy, O.; Imoukhuede, E.; Jepsen, S.; Ofori-Anyinam, O.; Faber, B.; et al. Safety and immunogenicity of a recombinant Plasmodium falciparum AMA1 malaria vaccine adjuvanted with alhydrogel™, montanide ISA 720 or AS02. PLoS ONE 2008, 3, e3960. [Google Scholar] [CrossRef] [Green Version]
  223. Brando, C.; Ware, L.A.; Freyberger, H.; Kathcart, A.; Barbosa, A.; Cayphas, S.; Demoitie, M.A.; Mettens, P.; Heppner, D.G.; Lanar, D.E. Murine immune responses to liver-stage antigen 1 protein FMP011, a malaria vaccine candidate, delivered with adjuvant AS01B or AS02A. Infect. Immun. 2007, 75, 838–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Audran, R.; Lurati-Ruiz, F.; Genton, B.; Blythman, H.E.; Ofori-Anyinam, O.; Reymond, C.; Corradin, G.; Spertini, F. The synthetic Plasmodium falciparum circumsporozoite peptide PFCS102 as a malaria vaccine candidate: A randomized controlled phase I trial. PLoS ONE 2009, 4, e7304. [Google Scholar] [CrossRef] [PubMed]
  225. Kester, K.E.; Cummings, J.F.; Ofori-Anyinam, O.; Ockenhouse, C.F.; Krzych, U.; Moris, P.; Schwenk, R.; Nielsen, R.A.; Debebe, Z.; Pinelis, E.; 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, 337–346. [Google Scholar] [CrossRef]
  226. Polhemus, M.E.; Remich, S.A.; Ogutu, B.R.; Waitumbi, J.N.; Otieno, L.; Apollo, S.; Cummings, J.F.; Kester, K.E.; Ockenhouse, C.F.; Stewart, A.; et al. Evaluation of RTS,S/AS02A and RTS,S/AS01B in adults in a high malaria transmission area. PLoS ONE 2009, 4, e6465. [Google Scholar] [CrossRef] [Green Version]
  227. Kester, K.E.; Cummings, J.F.; Ockenhouse, C.F.; Nielsen, R.; Hall, B.T.; Gordon, D.M.; Schwenk, R.J.; Krzych, U.; Holland, C.A.; Richmond, G.; et al. Phase 2a trial of 0, 1, and 3 month and 0, 7, and 28 day immunization schedules of malaria vaccine RTS,S/AS02 in malaria-naive adults at the walter reed army institute of research. Vaccine 2008, 26, 2191–2202. [Google Scholar] [CrossRef] [Green Version]
  228. Alonso, P.L.; Sacarlal, J.; Aponte, J.J.; Leach, A.; Macete, E.; Aide, P.; Sigauque, B.; Milman, J.; Mandomando, I.; Bassat, Q.; 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, 2012–2018. [Google Scholar] [CrossRef]
  229. Leroux-Roels, G.; Leroux-Roels, I.; Clement, F.; Ofori-Anyinam, O.; Lievens, M.; Jongert, E.; Moris, P.; Ballou, W.R.; Cohen, J. Evaluation of the immune response to RTS,S/AS01 and RTS,S/AS02 adjuvanted vaccines: Randomized, double-blind study in malaria-naive adults. Hum. Vaccines Immunother. 2014, 10, 2211–2219. [Google Scholar] [CrossRef] [Green Version]
  230. Stoute, J.A.; Kester, K.E.; Krzych, U.; Wellde, B.T.; Hall, T.; White, K.; Glenn, G.; Ockenhouse, C.F.; Garcon, N.; Schwenk, R.; et al. Long-term efficacy and immune responses following immunization with the RTS,S malaria vaccine. J. Infect. Dis. 1998, 178, 1139–1144. [Google Scholar] [CrossRef] [Green Version]
  231. Bojang, K.A.; Olodude, F.; Pinder, M.; Ofori-Anyinam, O.; Vigneron, L.; Fitzpatrick, S.; Njie, F.; Kassanga, A.; Leach, A.; Milman, J.; et al. Safety and immunogenicty of RTS,S/AS02A candidate malaria vaccine in gambian children. Vaccine 2005, 23, 4148–4157. [Google Scholar] [CrossRef]
  232. Wu, Y.; Ellis, R.D.; Shaffer, D.; Fontes, E.; Malkin, E.M.; Mahanty, S.; Fay, M.P.; Narum, D.; Rausch, K.; Miles, A.P.; et al. Phase 1 trial of malaria transmission blocking vaccine candidates Pfs25 and Pvs25 formulated with montanide ISA 51. PLoS ONE 2008, 3, e2636. [Google Scholar] [CrossRef]
  233. Aucouturier, J.; Dupuis, L.; Deville, S.; Ascarateil, S.; Ganne, V. Montanide ISA 720 and 51: A new generation of water in oil emulsions as adjuvants for human vaccines. Expert Rev. Vaccines 2002, 1, 111–118. [Google Scholar] [CrossRef] [PubMed]
  234. Verma, Y.K.; Reddy, B.S.; Pawar, M.S.; Bhunia, D.; Sampath Kumar, H.M. Design, synthesis, and immunological evaluation of benzyloxyalkyl-substituted 1,2,3-triazolyl alpha-galcer analogues. ACS Med. Chem. Lett. 2016, 7, 172–176. [Google Scholar] [CrossRef] [Green Version]
  235. Sandeep, A.; Reddy, B.S.; Hyder, I.; Kumar, H.M.S. Synthesis of a new class of glycolipids and the evaluation of their immunogenicity using murine splenocytes. J. Carbohydr. Chem. 2016, 35, 326–343. [Google Scholar] [CrossRef]
  236. Chirke, S.S.; Krishna, J.S.; Rathod, B.B.; Bonam, S.R.; Khedkar, V.M.; Rao, B.V.; Sampath Kumar, H.M.; Shetty, P.R. Synthesis of triazole derivatives of 9-ethyl-9H-carbazole and dibenzo[b,d]furan and evaluation of their antimycobacterial and immunomodulatory activity. ChemistrySelect 2017, 2, 7309–7318. [Google Scholar] [CrossRef]
  237. Lingamurthy, M.; Nalliboina, G.R.; Rao, M.V.; Rao, B.V.; Reddy, B.S.; Kumar, H.M.S. Ddq mediated stereoselective intermolecular benzylic cn bond formation: Synthesis of (−)-cytoxazone,(−)-4-epi-cytoxazone and their analogues and immunological evaluation of their cytokine modulating activity. Tetrahedron 2017, 73, 1473–1481. [Google Scholar] [CrossRef]
  238. Reddy Bonam, S.; Naidu Gorantla, J.; Thangarasu, A.K.; Lankalapalli, R.S.; Sampath Kumar, H.M. Polyhydroxy-n-alkyl-2-pyrrolidinones as a new class of glycolipid analogues with immune modulation potential. J. Carbohydr. Chem. 2018, 37, 30–43. [Google Scholar] [CrossRef]
  239. Bonam, S.R.; Bhunia, D.; Muller, S.; Nerella, S.G.; Alvala, M.; Halmuthur Mahabalarao, S.K. Novel trisaccharide based phospholipids as immunomodulators. Int. Immunopharmacol. 2019, 74, 105684. [Google Scholar] [CrossRef] [PubMed]
  240. Detienne, S.; Welsby, I.; Collignon, C.; Wouters, S.; Coccia, M.; Delhaye, S.; Van Maele, L.; Thomas, S.; Swertvaegher, M.; Detavernier, A.; et al. Central role of CD169+ lymph node resident macrophages in the adjuvanticity of the QS-21 component of AS01. Sci. Rep. 2016, 6, 39475. [Google Scholar] [CrossRef]
  241. Welsby, I.; Detienne, S.; N’Kuli, F.; Thomas, S.; Wouters, S.; Bechtold, V.; De Wit, D.; Gineste, R.; Reinheckel, T.; Elouahabi, A.; et al. Lysosome-dependent activation of human dendritic cells by the vaccine adjuvant QS-21. Front. Immunol. 2016, 7, 663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Ng, H.I.; Fernando, G.J.P.; Depelsenaire, A.C.I.; Kendall, M.A.F. Potent response of QS-21 as a vaccine adjuvant in the skin when delivered with the nanopatch, resulted in adjuvant dose sparing. Sci. Rep. 2016, 6, 29368. [Google Scholar] [CrossRef] [PubMed]
  243. Collins, K.A.; Snaith, R.; Cottingham, M.G.; Gilbert, S.C.; Hill, A.V.S. Enhancing protective immunity to malaria with a highly immunogenic virus-like particle vaccine. Sci. Rep. 2017, 7, 46621. [Google Scholar] [CrossRef] [Green Version]
  244. Coler, R.N.; Carter, D.; Friede, M.; Reed, S.G. Adjuvants for malaria vaccines. Parasite Immunol. 2009, 31, 520–528. [Google Scholar] [CrossRef]
  245. Petrovsky, N. Comparative safety of vaccine adjuvants: A summary of current evidence and future needs. Drug Saf. 2015, 38, 1059–1074. [Google Scholar] [CrossRef]
  246. Klinman, D.M. Immunotherapeutic uses of CpG oligodeoxynucleotides. Nat. Rev. Immunol. 2004, 4, 249–258. [Google Scholar] [CrossRef] [PubMed]
  247. Pirahmadi, S.; Zakeri, S.; Mehrizi, A.A.; Djadid, N.D.; Raz, A.A.; Sani, J.J. Combining monophosphoryl lipid a (MPL), CpG oligodeoxynucleotide (ODN), and QS-21 adjuvants induces strong and persistent functional antibodies and T cell responses against cell-traversal protein for ookinetes and sporozoites (CelTOS) of Plasmodium falciparum in BALB/c mice. Infect. Immun. 2019, 87, e00911-18. [Google Scholar] [CrossRef] [Green Version]
  248. Mullen, G.E.; Ellis, R.D.; Miura, K.; Malkin, E.; Nolan, C.; Hay, M.; Fay, M.P.; Saul, A.; Zhu, D.; Rausch, K.; et al. Phase 1 trial of AMA1-C1/alhydrogel plus CpG 7909: An asexual blood-stage vaccine for Plasmodium falciparum malaria. PLoS ONE 2008, 3, e2940. [Google Scholar] [CrossRef] [Green Version]
  249. Traore, B.; Kone, Y.; Doumbo, S.; Doumtabe, D.; Traore, A.; Crompton, P.D.; Mircetic, M.; Huang, C.Y.; Kayentao, K.; Dicko, A.; et al. The TLR9 agonist CpG fails to enhance the acquisition of Plasmodium falciparum-specific memory B cells in semi-immune adults in mali. Vaccine 2009, 27, 7299–7303. [Google Scholar] [CrossRef] [Green Version]
  250. Horii, T.; Shirai, H.; Jie, L.; Ishii, K.J.; Palacpac, N.Q.; Tougan, T.; Hato, M.; Ohta, N.; Bobogare, A.; Arakaki, N.; et al. Evidences of protection against blood-stage infection of Plasmodium falciparum by the novel protein vaccine SE36. Parasitol. Int. 2010, 59, 380–386. [Google Scholar] [CrossRef] [PubMed]
  251. Tougan, T.; Aoshi, T.; Coban, C.; Katakai, Y.; Kai, C.; Yasutomi, Y.; Ishii, K.J.; Horii, T. TLR9 adjuvants enhance immunogenicity and protective efficacy of the SE36/AHG malaria vaccine in nonhuman primate models. Hum. Vaccines Immunother. 2013, 9, 283–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Duncan, C.J.; Sheehy, S.H.; Ewer, K.J.; Douglas, A.D.; Collins, K.A.; Halstead, F.D.; Elias, S.C.; Lillie, P.J.; Rausch, K.; Aebig, J.; et al. Impact on malaria parasite multiplication rates in infected volunteers of the protein-in-adjuvant vaccine AMA1-C1/alhydrogel+CpG 7909. PLoS ONE 2011, 6, e22271. [Google Scholar] [CrossRef] [Green Version]
  253. Antony, H.A.; Parija, S.C. Antimalarial drug resistance: An overview. Trop. Parasitol. 2016, 6, 30–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Rappuoli, R.; Aderem, A. A 2020 vision for vaccines against HIV, tuberculosis and malaria. Nature 2011, 473, 463–469. [Google Scholar] [CrossRef] [PubMed]
  255. Voepel, N.; Boes, A.; Edgue, G.; Beiss, V.; Kapelski, S.; Reimann, A.; Schillberg, S.; Pradel, G.; Fendel, R.; Scheuermayer, M. Malaria vaccine candidate antigen targeting the pre-erythrocytic stage of Plasmodium falciparum produced at high level in plants. Biotechnol. J. 2014, 9, 1435–1445. [Google Scholar] [CrossRef] [PubMed]
  256. van den Berg, R.A.; Coccia, M.; Ballou, W.R.; Kester, K.E.; Ockenhouse, C.F.; Vekemans, J.; Jongert, E.; Didierlaurent, A.M.; van der Most, R.G. Predicting RTS,S vaccine-mediated protection from transcriptomes in a malaria-challenge clinical trial. Front. Immunol. 2017, 8, 557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Sauerwein, R.W.; Roestenberg, M.; Moorthy, V.S. Experimental human challenge infections can accelerate clinical malaria vaccine development. Nat. Rev. Immunol. 2011, 11, 57–64. [Google Scholar] [CrossRef]
  258. Vaughan, A.M.; Kappe, S.H.I. Malaria parasite liver infection and exoerythrocytic biology. Cold Spring Harb. Perspect. Med. 2017, 7, a025486. [Google Scholar] [CrossRef] [Green Version]
  259. Paul, A.S.; Egan, E.S.; Duraisingh, M.T. Host–parasite interactions that guide red blood cell invasion by malaria parasites. Curr. Opin. Hematol. 2015, 22. [Google Scholar] [CrossRef] [PubMed]
  260. Sinnis, P.; Sim, B.K. Cell invasion by the vertebrate stages of plasmodium. Trends Microbiol. 1997, 5, 52–58. [Google Scholar] [CrossRef] [Green Version]
  261. Holz, L.E.; Prier, J.E.; Freestone, D.; Steiner, T.M.; English, K.; Johnson, D.N.; Mollard, V.; Cozijnsen, A.; Davey, G.M.; Godfrey, D.I.; et al. CD8+ T cell activation leads to constitutive formation of liver tissue-resident memory T cells that seed a large and flexible niche in the liver. Cell Rep. 2018, 25, 68–79.e4. [Google Scholar] [CrossRef] [Green Version]
  262. Weaver, R.; Reiling, L.; Feng, G.; Drew, D.R.; Mueller, I.; Siba, P.M.; Tsuboi, T.; Richards, J.S.; Fowkes, F.J.I.; Beeson, J.G. The association between naturally acquired IgG subclass specific antibodies to the pfrh5 invasion complex and protection from Plasmodium falciparum malaria. Sci. Rep. 2016, 6, 33094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Berry, A.A.; Gottlieb, E.R.; Kouriba, B.; Diarra, I.; Thera, M.A.; Dutta, S.; Coulibaly, D.; Ouattara, A.; Niangaly, A.; Kone, A.K.; et al. Immunoglobulin G subclass and antibody avidity responses in Malian children immunized with Plasmodium falciparum apical membrane antigen 1 vaccine candidate FMP2.1/AS02A. Malar. J. 2019, 18, 13. [Google Scholar] [CrossRef]
  264. Ndungu, F.M.; Mwacharo, J.; Wambua, J.; Njuguna, P.; Marsh, K.; Drakeley, C.; Bejon, P. A seven-year study on the effect of the pre-erythrocytic malaria vaccine candidate RTS,S/AS01E on blood stage immunity in young Kenyan children. Wellcome Open Res. 2019, 4, 42. [Google Scholar] [CrossRef] [Green Version]
  265. Horowitz, A.; Hafalla, J.C.; King, E.; Lusingu, J.; Dekker, D.; Leach, A.; Moris, P.; Cohen, J.; Vekemans, J.; Villafana, T.; et al. Antigen-specific IL-2 secretion correlates with NK cell responses after immunization of tanzanian children with the RTS,S/AS01 malaria vaccine. J. Immunol. 2012, 188, 5054–5062. [Google Scholar] [CrossRef]
  266. Dunachie, S.J.; Hill, A.V. Prime-boost strategies for malaria vaccine development. J. Exp. Biol. 2003, 206, 3771–3779. [Google Scholar] [CrossRef] [Green Version]
  267. Hoffman, S.L.; Vekemans, J.; Richie, T.L.; Duffy, P.E. The march toward malaria vaccines. Vaccine 2015, 33 (Suppl. 4), D13–D23. [Google Scholar] [CrossRef] [Green Version]
  268. López, C.; Yepes-Pérez, Y.; Hincapié-Escobar, N.; Díaz-Arévalo, D.; Patarroyo, M.A. What is known about the immune response induced by plasmodium vivax malaria vaccine candidates? Front. Immunol. 2017, 8, 126. [Google Scholar] [CrossRef] [Green Version]
  269. Nega, D.; Alemu, A.; Tasew, G. Malaria vaccine development: Recent advances alongside the barriers. J. Bacteriol. Parasitol. 2016, 7, 300. [Google Scholar] [CrossRef]
  270. Villa, E.; Marchetti, S.; Ricci, J.E. No parkin zone: Mitophagy without parkin. Trends Cell Biol. 2018, 28, 882–895. [Google Scholar] [CrossRef] [PubMed]
  271. Bejon, P.; Cook, J.; Bergmann-Leitner, E.; Olotu, A.; Lusingu, J.; Mwacharo, J.; Vekemans, J.; Njuguna, P.; Leach, A.; Lievens, M.; et al. Effect of the pre-erythrocytic candidate malaria vaccine RTS,S/AS01E on blood stage immunity in young children. J. Infect. Dis. 2011, 204, 9–18. [Google Scholar] [CrossRef] [PubMed]
  272. Abdulla, S.; Salim, N.; Machera, F.; Kamata, R.; Juma, O.; Shomari, M.; Kubhoja, S.; Mohammed, A.; Mwangoka, G.; Aebi, T.; et al. Randomized, controlled trial of the long term safety, immunogenicity and efficacy of RTS,S/AS02D malaria vaccine in infants living in a malaria-endemic region. Malar. J. 2013, 12, 11. [Google Scholar] [CrossRef] [Green Version]
  273. Rampling, T.; Ewer, K.J.; Bowyer, G.; Edwards, N.J.; Wright, D.; Sridhar, S.; Payne, R.; Powlson, J.; Bliss, C.; Venkatraman, N.; et al. Safety and efficacy of novel malaria vaccine regimens of RTS,S/AS01B alone, or with concomitant ChAd63-MVA-vectored vaccines expressing ME-TRAP. NPJ Vaccines 2018, 3, 49. [Google Scholar] [CrossRef] [PubMed]
  274. Berendt, A.R.; Tumer, G.D.; Newbold, C.I. Cerebral malaria: The sequestration hypothesis. Parasitol. Today 1994, 10, 412–414. [Google Scholar] [CrossRef]
  275. Clark, I.A.; Rockett, K.A. The cytokine theory of human cerebral malaria. Parasitol. Today 1994, 10, 410–412. [Google Scholar] [CrossRef]
  276. Vekemans, J.; Ballou, W.R. Plasmodium falciparum malaria vaccines in development. Expert Rev. Vaccines 2008, 7, 223–240. [Google Scholar] [CrossRef]
Figure 1. Complex life cycle of P. falciparum. The life cycle has 3 stages: the pre-erythrocytic and erythrocytic stages in humans (host) and sexual process in the mosquito vector. a|Pre-erythrocytic stage. b|Erythrocytic stage. c|Mosquito/sexual stage.
Figure 1. Complex life cycle of P. falciparum. The life cycle has 3 stages: the pre-erythrocytic and erythrocytic stages in humans (host) and sexual process in the mosquito vector. a|Pre-erythrocytic stage. b|Erythrocytic stage. c|Mosquito/sexual stage.
Vaccines 09 01072 g001
Figure 2. Immunity in malaria infection. A|Activation of dendritic cells. During the infection, both liver- and blood-stage parasites are recognized by the several PRRs present in the cells of the innate immune system. Majorly, monocytes, macrophages, and DCs have a significant role in the recognition. Vast literature has documented the TLR-mediated signals by parasite components, such as glycosylphosphatidylinositol (GPI), hemozoin, and DNA, upon infection. In addition, hemozoin and uric acid (released from the dying cells) activate the inflammasome. Furthermore, many other malaria parasite PAMPs are recognized by the different other unknown receptors and sensors. Once DCs/macrophages recognize the PAMPs and DAMPs, they phagocyte and process the pathogen, followed by the antigen presentation, to the T cells (reviewed in [25]). In addition, Plasmodium RNA (particularly in the hepatocytes) is sensed by the MDA5 ((RIG-I)-like receptor) and activates a type 1 IFN response via IRF3 and IRF7 signaling [26]. The above-mentioned innate immune responses help in the development of antigen-specific adaptive immunity. B|Activation of adaptive immunity. CD4+ T cells are activated by the DCs, and under the influence of cytokine milieu, CD4+ T cells are polarized into Th1, Th2, Th17, Treg, and Tfh cells. Each CD4+ T cell subset has a significant role in shaping the immune response (reviewed in [27]). NK cells perceive the signals by cytokines, which are produced from DCs, monocytes, and/or macrophages. Once activated, NK cells secrete inflammatory cytokines (IL-8, IFN-γ, TNF-α, CCL4, and others), which act as danger signals to gain other immune cells’ attention. NK cells also perform cytotoxicity as one of their main functions. NK cells either directly or indirectly kill the infected cells using perforin and granzyme and antibody-dependent cell cytotoxicity (ADCC), respectively. Antigen cross-presentation by DCs activates the CD8+ T cells, which produce IFN-γ upon activation. IFN-γ-producing CD8+ T cells perform inflammatory as well as cytotoxic (perforin and granzyme B mediated) functions. Though DCs induce a balanced immune response, the parasite could promote a severe pathology of cerebral malaria as a part of the immune evasion strategy [28].
Figure 2. Immunity in malaria infection. A|Activation of dendritic cells. During the infection, both liver- and blood-stage parasites are recognized by the several PRRs present in the cells of the innate immune system. Majorly, monocytes, macrophages, and DCs have a significant role in the recognition. Vast literature has documented the TLR-mediated signals by parasite components, such as glycosylphosphatidylinositol (GPI), hemozoin, and DNA, upon infection. In addition, hemozoin and uric acid (released from the dying cells) activate the inflammasome. Furthermore, many other malaria parasite PAMPs are recognized by the different other unknown receptors and sensors. Once DCs/macrophages recognize the PAMPs and DAMPs, they phagocyte and process the pathogen, followed by the antigen presentation, to the T cells (reviewed in [25]). In addition, Plasmodium RNA (particularly in the hepatocytes) is sensed by the MDA5 ((RIG-I)-like receptor) and activates a type 1 IFN response via IRF3 and IRF7 signaling [26]. The above-mentioned innate immune responses help in the development of antigen-specific adaptive immunity. B|Activation of adaptive immunity. CD4+ T cells are activated by the DCs, and under the influence of cytokine milieu, CD4+ T cells are polarized into Th1, Th2, Th17, Treg, and Tfh cells. Each CD4+ T cell subset has a significant role in shaping the immune response (reviewed in [27]). NK cells perceive the signals by cytokines, which are produced from DCs, monocytes, and/or macrophages. Once activated, NK cells secrete inflammatory cytokines (IL-8, IFN-γ, TNF-α, CCL4, and others), which act as danger signals to gain other immune cells’ attention. NK cells also perform cytotoxicity as one of their main functions. NK cells either directly or indirectly kill the infected cells using perforin and granzyme and antibody-dependent cell cytotoxicity (ADCC), respectively. Antigen cross-presentation by DCs activates the CD8+ T cells, which produce IFN-γ upon activation. IFN-γ-producing CD8+ T cells perform inflammatory as well as cytotoxic (perforin and granzyme B mediated) functions. Though DCs induce a balanced immune response, the parasite could promote a severe pathology of cerebral malaria as a part of the immune evasion strategy [28].
Vaccines 09 01072 g002
Figure 3. Structures of QS-21 (a) and MPL (b).
Figure 3. Structures of QS-21 (a) and MPL (b).
Vaccines 09 01072 g003
Figure 4. Proposed mechanisms of action of QS-21 in AS01 and QS-21 alone. a|Upon intramuscular injection, QS-21 (within the liposomes) drains to the lymph nodes and localizes at the subcapsular sinus (SCS) macrophages [240]. Macrophages activated via NLRP3 inflammasome pathways recruit other bystander cells including neutrophils, monocytes, and DCs [241]. b|QS-21 (within the liposomes) helps in the MHC-II and MHC-I antigen cross-presentation by forming a lipid body in the DCs and activating T cells [240,242]. Occasionally, in another way, QS-21 (alone) containing aldehyde moiety interacts with the T cell surface receptor (ε-amino acid) (e.g., CD2) and induces costimulatory signals [211]. These interactions, followed by ion exchanges (K+, Na+, Ca2+) and the activation of mitogen-activated protein (MAP) kinase, ERK2, and transcription factors, lead to cytokine release and cell proliferation, differentiation, and migration. c|QS-21 (within the liposomes) enters DCs via cholesterol-mediated endocytosis and situates itself in the lysosomes, where it destabilizes the lysosomes by forming size-specific pores. Cathepsin B (cysteine proteases) released from the lysosomes has an effective role in the immune functions mediated by QS-21. QS-21 (alone or in liposomes) activates the inflammasome (NLRP3), like other adjuvants, which induces active caspase 1 production. Activated caspase 1 coverts the pro-inflammatory cytokines (IL-1β and IL-18) into active forms [240].
Figure 4. Proposed mechanisms of action of QS-21 in AS01 and QS-21 alone. a|Upon intramuscular injection, QS-21 (within the liposomes) drains to the lymph nodes and localizes at the subcapsular sinus (SCS) macrophages [240]. Macrophages activated via NLRP3 inflammasome pathways recruit other bystander cells including neutrophils, monocytes, and DCs [241]. b|QS-21 (within the liposomes) helps in the MHC-II and MHC-I antigen cross-presentation by forming a lipid body in the DCs and activating T cells [240,242]. Occasionally, in another way, QS-21 (alone) containing aldehyde moiety interacts with the T cell surface receptor (ε-amino acid) (e.g., CD2) and induces costimulatory signals [211]. These interactions, followed by ion exchanges (K+, Na+, Ca2+) and the activation of mitogen-activated protein (MAP) kinase, ERK2, and transcription factors, lead to cytokine release and cell proliferation, differentiation, and migration. c|QS-21 (within the liposomes) enters DCs via cholesterol-mediated endocytosis and situates itself in the lysosomes, where it destabilizes the lysosomes by forming size-specific pores. Cathepsin B (cysteine proteases) released from the lysosomes has an effective role in the immune functions mediated by QS-21. QS-21 (alone or in liposomes) activates the inflammasome (NLRP3), like other adjuvants, which induces active caspase 1 production. Activated caspase 1 coverts the pro-inflammatory cytokines (IL-1β and IL-18) into active forms [240].
Vaccines 09 01072 g004
Table 1. List of vaccines under clinical testing without specific vaccine adjuvants.
Table 1. List of vaccines under clinical testing without specific vaccine adjuvants.
Malaria VaccineClinical Trial IdentifierCurrent Stage
ChAd63 RH5 (chimpanzee adenovirus serotype 63 reticulocyte-binding protein homolog 5)NCT02181088Phase 1
MVA (modified vaccinia virus Ankara) RH5NCT02181088Phase 1
PEBS-POC1 (synthetic protein containing 131 amino acids)NCT01605786Phase 1
ChAd63-METRAP (multiple epitope string and thrombospondin-related adhesion protein)NCT03084289Phase 1
MVA METRAPNCT03084289Phase 1
DNA-Ad (contains a combination of circumsporozoite (CS) protein and AMA1)NCT00870987Phase 2
PfSPZ (P. falciparum (Pf) sporozoite (SPZ))NCT02601716Phase 2
Ad35.CS.01 (P. falciparum CS surface antigen is inserted in a replication deficient Adenovirus 35 backbone)NCT01018459Phase 1
CS protein expressed either in MVA, or an attenuated Fowl pox virus strain (FP9).NCT00121771Phase 1
AdCh63-MSP1 (merozoite surface protein-1) and MVA-MSP1NCT01003314Phase 2
GMZ2 (recombinant hybrid of the glutamate rich protein (GLURP) and the merozoite surface protein 3 (MSP 3))NCT00424944Phase 1
FP9-PP and MVA-PP (FP9 polyprotein, modified virus Ankara polyprotein)NCT00374998Phase 1
p52-p36-GAP (genetically attenuated parasite malaria vaccine)NCT01024686Phase 2
PfSPZ-GA1 (genetically attenuated PfSPZ)NCT03163121Phase 1
ChAdOx1 LS2 (malaria liver-stage dual antigen LS2 (LSA1 and LSAP2) fused with the transmembrane domain from shark invariant chain) and MVA LS2NCT03203421Phase 2/Phase 1
DNA-Ad (it contains a liver-stage antigen (circumsporozoite protein) and an antigen (apical membrane antigen 1))NCT00870987Phase 1
NMRC-M3V-Ad-PfCA (NMRC + multi-antigen multi-stage, malaria vaccine + adenovectored + P. falciparum CSP and AMA1 antigens), is a combination of two recombinant adenovirus-derived constructs (adenovectors)NCT00392015Phase 1
Table 2. Status of malarial vaccines with vaccine adjuvants #.
Table 2. Status of malarial vaccines with vaccine adjuvants #.
AdjuvantVaccineLife-Cycle StageClinical Trial IdentifierCurrent Stage
Aluminium hydroxide/Alhydrogel®Lyophilized PEBS synthetic protein (PfPEBS) (synthetic protein containing 131 amino acids)Pre-erythrocytic and blood stageNCT01605786Phase 2
AMA1-C1 (combination of the 3D7 and FVO alleles of P. falciparum apical membrane antigen-1 (AMA1))Blood stageNCT00984763
NCT00114010
Phase 2
Phase 1
PRIMVAC (VAR2CSA protein)Blood stageNCT02658253Phase 1
P27A proteinBlood stageNCT01949909Phase 1
Pfs25-EPA (Pfs25 has been conjugated to Pseudomonas aeruginosa ExoProtein A) (EPA)Sexual stageNCT01434381Phase 1
Pfs25 VLPSexual stageNCT02013687Phase 1
AMA1-DiCoBlood stageNCT02014727Phase 1
Pfs25M-EPA, Pfs230D1M-EPASexual stageNCT02334462Phase 1
AdCh63 AMA1 + MVA AMA1 + AMA1-C1Blood stageNCT01351948Phase 1
BSAM-2 (mixture of two proteins found on the surface of merozoites, AMA1 and MSP1 (42))Blood stageNCT00889616Phase 1
ICC-1132Pre-erythrocytic stageNCT00587249Phase 1
PAMVACBlood stageNCT02647489Phase 1
MSP3-LSPBlood stageNCT01341704Phase 2
MSP1 42-C1Blood stageNCT00320658Phase 1
Aluminum phosphateErythrocyte-binding antigen 175 kDA region II-non-glycosylated (EBA-175 RII-NG)Blood stageNCT01026246Phase 1
AS01RH5.1 (protein ectodomain of the PfRH5 (amino acids E26—Q526) antigen)Blood stageNCT02927145Phase 2
Pfs25M-EPA, Pfs230D1M-EPA (PfS230D1M conjugated to Pseudomonas aeruginosa ExoProtein A (EPA))Sexual stageNCT02942277Phase 1
AS01BR21 (RTS, S-like vaccine)Pre-erythrocytic stageNCT02600975Phase 1
FMP012 (Escherichia coli-expressed P. falciparum cell-traversal protein for ookinetes and sporozoites (PfCelTOS))Sexual stageNCT02174978Phase 1
P. falciparum malaria protein (FMP)010Blood stageNCT00666380Phase 1
FMP2.1 (AMA1 malaria antigen)Blood stageNCT00385047Phase 1
AS01ERTS, SPre-erythrocytic stageNCT00380393Phase 2
AS02AFalciparum Merozoite Protein-1 (FMP1)Blood stageNCT00308061Phase 1
FMP2.1 (AMA1 malaria antigen)Blood stageNCT00385047Phase 2
Falciparum Malaria Protein 11Pre-erythrocytic stageNCT00312702Phase 2
AS02DRTS, SPre-erythrocytic stageNCT00289185Phase 2
ODN 2006 (7909)MSP1 42-C1Blood stageNCT00320658Phase 1
AMA1-C1 (combination of the 3D7 and FVO alleles of P. falciparum apical membrane antigen-1 (AMA1))Blood stageNCT00984763Phase 2
AdCh63 AMA1 + MVA AMA1 + AMA1-C1Blood stageNCT01351948Phase 1
BSAM-2Blood stageNCT00889616Phase 1
Glucopyranosyl Lipid Adjuvant-Liposome-QS-21 Formulation (GLA-LSQ)PAMVACBlood stageNCT02647489Phase 1
Recombinant circumsporozoite protein (rCSP) malaria vaccine administered with and without AP 10-602 (GLA-LSQ)Pre-erythrocytic stageNCT03589794Phase 1
Glucopyranosyl Lipid Adjuvant-Stable Emulsion (GLA-SE)PAMVACBlood stageNCT02647489Phase 1
PRIMVAC (VAR2CSA protein)Blood stageNCT02658253Phase 1
P27A proteinBlood stageNCT01949909Phase 1
AMA1-DiCoBlood stageNCT02014727Phase 1
FMP012Sexual stageNCT01540474Phase 1
Matrix-M1 (a bifunctional matrix protein of influenza virus)R21Pre-erythrocytic stageNCT02925403Phase 1
Montanide ISA51PpPfs25Sexual stageNCT00295581Phase 1
Montanide ISA 720PfCS102 (antigen of the sporozoite protein)Pre-erythrocytic stageNCT01031524Phase 1
VirosomesPEV301 & 302 (it includes two antigens (CSP and AMA1-derived))Pre-erythrocytic stage and blood stageNCT00513669Phase 1
Abbreviations: AMA1-DiCo, apical membrane antigen 1 diversity covering; ICC-1132, a vaccine candidate composed of P. falciparum hepatitis B virus core particle, which contains T- and B-cell epitopes from the repeat region of the C terminus circumsporozoite protein; METRAP, multiple-epitope thrombospondin-related adhesion protein; MSP3-LSP, merozoite surface protein-3 long synthetic peptide; PfCelTOS, P. falciparum cell traversal protein of ookinetes and sporozoites; PfAMA1, P. falciparum apical membrane antigen 1; PfPEBS, P. falciparum pre-erythrocytic and blood stage; PfMSP1, P. falciparum merozoite surface protein 1; PfRH5, P. falciparum reticulocyte-binding protein homolog 5; PfSERA5, P. falciparum Serine Repeat Antigen-5; P27A protein, unstructured 104 mer synthetic peptide from P. falciparum trophozoite exported protein 1; VAR2CSA, variant surface antigen 2CSA; VLP, virus-like particles. # We searched for malarial vaccines in clinical trials website (https://clinicaltrials.gov/ accessed on 1 June 2020) and selected the studies pertinent to P. falciparum. Many vaccines use alhydrogel as adjuvant, however, at different doses and different conditions. To our knowledge, many vaccines with a similar type of antigens using a diversified combination of adjuvants have been under clinical trials, which are not listed here.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bonam, S.R.; Rénia, L.; Tadepalli, G.; Bayry, J.; Kumar, H.M.S. Plasmodium falciparum Malaria Vaccines and Vaccine Adjuvants. Vaccines 2021, 9, 1072. https://doi.org/10.3390/vaccines9101072

AMA Style

Bonam SR, Rénia L, Tadepalli G, Bayry J, Kumar HMS. Plasmodium falciparum Malaria Vaccines and Vaccine Adjuvants. Vaccines. 2021; 9(10):1072. https://doi.org/10.3390/vaccines9101072

Chicago/Turabian Style

Bonam, Srinivasa Reddy, Laurent Rénia, Ganesh Tadepalli, Jagadeesh Bayry, and Halmuthur Mahabalarao Sampath Kumar. 2021. "Plasmodium falciparum Malaria Vaccines and Vaccine Adjuvants" Vaccines 9, no. 10: 1072. https://doi.org/10.3390/vaccines9101072

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop