6,087
Views
8
CrossRef citations to date
0
Altmetric
Signature Reviews

Pathogenicity and virulence of malaria: Sticky problems and tricky solutions

& ORCID Icon
Article: 2150456 | Received 05 Jun 2022, Accepted 19 Oct 2022, Published online: 04 Jan 2023

ABSTRACT

Infections with Plasmodium falciparum and Plasmodium vivax cause over 600,000 deaths each year, concentrated in Africa and in young children, but much of the world’s population remain at risk of infection. In this article, we review the latest developments in the immunogenicity and pathogenesis of malaria, with a particular focus on P. falciparum, the leading malaria killer. Pathogenic factors include parasite-derived toxins and variant surface antigens on infected erythrocytes that mediate sequestration in the deep vasculature. Host response to parasite toxins and to variant antigens is an important determinant of disease severity. Understanding how parasites sequester, and how antibody to variant antigens could prevent sequestration, may lead to new approaches to treat and prevent disease. Difficulties in malaria diagnosis, drug resistance, and specific challenges of treating P. vivax pose challenges to malaria elimination, but vaccines and other preventive strategies may offer improved disease control.

Introduction

There are 241 million cases of malaria per year, predominantly in sub-Saharan Africa. Malaria can be life threatening and caused 627,000 deaths in 2021 [Citation1]. Of the five species of Plasmodium spp. that commonly infect humans – P. falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesiP. falciparum causes 95% of the cases and the great majority of the severe disease [Citation1], although severe disease can also be caused by the less prevalent P. vivax and P. knowlesi [Citation2,Citation3].

Malaria is one of the oldest diseases on record and has shaped the human genome, leading to the selection of protective genetic traits such as sickle cell trait and beta-thalassaemia. Similarly, Plasmodium spp. have developed mechanisms to prolong infection in the human host, including the ability to evade immunity by expressing highly diverse surface antigens and to sequester in the host microvasculature, and continue to develop resistance to chemotherapeutics. This review will summarize the key virulence factors involved in progression to or protection from severe disease and highlight important areas of future research.

Epidemiological risk factors for severe disease

Clinical manifestations of malaria range from mild to life threatening. Uncomplicated malaria symptoms are non-specific and include fever, nausea, vomiting, headache, and diarrhoea, with no indication of organ dysfunction. Uncomplicated malaria can rapidly become severe malaria, defined by the World Health Organization (WHO) as one or more of a range of conditions, including impaired consciousness, respiratory distress, convulsions, prostration, shock, pulmonary oedema, abnormal bleeding, and jaundice, that can be recognized by several laboratory indicators including severe anaemia, hyperparasitaemia, hyperlactaemia, acidosis, and renal impairment [Citation4].

The incidence of severe malaria is higher in high transmission settings [Citation5] and children under 5 are most susceptible [Citation6]. As such, children under 5 account for approximately 40% of global malaria cases and the majority of malaria related deaths [Citation1]. In high transmission settings, Plasmodium spp. infections in adults are generally asymptomatic, due to acquisition of protective immunity after repeat infections. In areas of low or unstable transmission, or in previously unexposed individuals, older children and adults are also vulnerable to severe disease [Citation5,Citation7]. In addition to young children, pregnant women are highly susceptible to P. falciparum infection and severe P. falciparum malaria, particularly severe anaemia. Malaria during pregnancy can also affect the baby, by causing miscarriage, stillbirth, or low birthweight delivery, which predisposes to neonatal and infant mortality [Citation8].

There are many environmental factors that contribute to transmission of malaria, such as rainfall, proximity to stagnant water, and human activity. In general, urbanization is thought to be a key contributor to the reduction of malaria [Citation9] and urban areas have less transmission compared to rural areas [Citation10]. However, some city centres have high estimated inoculation rates, likely due to several factors including poor drainage, unplanned urban dwellings [Citation10,Citation11] or nearby deforestation [Citation12]. Additionally, Anopheles mosquitoes have adapted to breed in urban environments, such as polluted water and insecticide treated water [Citation13,Citation14]. Anopheles stephensi is a prevalent vector for both P. falciparum and P. vivax malaria in South Asia that is well adapted to breed in urban settings [Citation15] and has expanded to the Horn of Africa where it is predicted to continue to spread and increase urban malaria outbreaks [Citation16].

Diagnostics

Prompt treatment is key to averting severe malaria and relies on access to accurate diagnosis and effective therapeutics. Blood-stage malaria can be diagnosed by light microscopic examination of stained thick and thin smears of peripheral blood, and microscopy remains the field standard for malaria diagnostics. However, light microscopy requires electricity and false negatives and incorrect speciation are common in microscopic diagnosis of P. malariae, P. ovale, and P. vivax, due to low density and similarities in morphology [Citation17]. Microscopy is also unlikely to detect low-density “sub-microscopic” infections (approximately <100 parasites per microliter of blood) that are reservoirs for transmission and are important to identify in countries targeting malaria elimination; in screening pregnant women who may have parasite sequestered in the placenta; as well as for blood safety in transfusion [Citation18]. Ultrasensitive PCR offers higher sensitivity than microscopy but the reliance on electricity, costly reagents and laboratory facilities for sample preparation have limited PCR to reference laboratories, rather than point of care [Citation19]. Loop-mediated isothermal amplification (LAMP) is a more portable alternative to PCR but the high cost and sample preparation requirements have prevented extensive use in the field, although a recent study suggests it may be cost effective to include in surveillance programs [Citation20].

Rapid diagnostic tests (RDTs) are affordable and easy to use and a large number of RDTs are commercially available that meet the WHO minimum performance criteria [Citation21]. Malaria RDTs mostly detect P. falciparum histidine-rich protein 2 (HRP2), as well as P. falciparum lactate dehydrogenase (LDH), P. vivax LDH, pan species LDH, and aldolase [Citation21]. Similarly to microscopy, RDTs lack sensitivity to detect low-density infections [Citation18], limiting their use in elimination settings. They are further challenged by the presence of HRP2 and/or HRP3 deletions in South America and sub-Saharan Africa [Citation22]. HRP2 antibodies can cross react with HRP3 at a certain antigen threshold and so RDTs can still be effective in regions with only HPR2 or HRP3 deletions, but not both [Citation23]. Interestingly, both modelling and field studies have shown that HRP2 deletions are driven by low transmission and the use of PfHRP2 RDTs to correctly treat a high proportion of infections [Citation24–26].

Life cycle and parasite density

Life cycle

Malaria was first confirmed to be caused by the Plasmodium parasite when observed by microscopy in a peripheral blood smear, in 1880 [Citation27]. Female Anopheles mosquitoes transmit the sporozoite form of P. falciparum to humans with saliva whilst taking a blood feed. Sporozoites are activated by migrating through hepatocytes and Kupffer cells [Citation28]. The activated sporozoites replicate by mitosis in hepatocytes, producing up to 40,000 parasites per hepatocyte, for 1–2 weeks. Hepatocytes release merozoite filled merosomes into the blood stream where they infect and replicate within erythrocytes. P. vivax and P. ovale can remain dormant (non-replicating) in the liver as hypnozoites for weeks, months and sometimes years following infection and can reactivate to cause relapses, although the mechanisms by which this occurs are unclear.

Malaria symptoms occur during the blood stage of the parasite lifecycle, a process which repeatedly cycles every 24–72 h, depending on the species. P. falciparum merozoites invade erythrocytes of all ages, whereas P. vivax will only invade reticulocytes recently released from the bone marrow. Merozoites use an actomyosin motor to undergo gliding motility across erythrocyte surfaces before attachment and invasion [Citation29]. Invasion is a multi-step process involving complex protein interactions. Initial low affinity attachment to the erythrocyte surface is followed by reorientation to align the apical end of the merozoite with the erythrocyte surface; a “tight junction” of high affinity interactions is then formed and finally, the merozoite surface is “shed” as it enters the erythrocyte [Citation30]. During the first hours inside the erythrocyte, the parasite is recognizable by a ring shape under Giemsa staining and is known as the ring stage. Ring-stage parasites establish a protein trafficking network and begin remodelling the infected erythrocyte (IE) [Citation31].

In P. falciparum infection, as rings develop into trophozoite stage parasites, they begin exporting knob-associated histidine-rich protein through the red cell cytoplasm to the plasma membrane which becomes deformed with knob-like protrusions (knobs) [Citation32]. Knobs are unique to P. falciparum, and anchor P. falciparum erythrocyte membrane protein 1 (PfEMP1) – a key parasite virulence factor – on the IE surface during the trophozoite stage [Citation33]. Trophozoites develop into schizonts, which burst to release 8–32 merozoites that infect erythrocytes and continue the asexual lifecycle. The parasite metabolizes host haemoglobin during the blood stage and detoxifies the released free haem by polymerization into a pigmented crystal known as haemozoin [Citation34].

A small percentage of ring-stage parasites differentiate into male or female gametocytes (reviewed in [Citation35]), the sexual form of the parasite, driven in part by the parasite’s ability to sense host physiological signals [Citation36]. Gametocytes are taken up by mosquitoes during a blood meal and enter the mosquito midgut. Gametocytes shed the erythrocyte membrane and the released male microgametes fertilize the released macrogametes to form a zygote, which develops into a motile ookinete. Ookinetes invade the midgut wall and become oocysts, which mitotically replicate and eventually rupture to release sporozoites. Sporozoites migrate to, and remain in, the salivary glands to be transmitted to a human host [Citation37].

Parasite biomass

It has long been established that a high parasite load is associated with poorer clinical outcomes, and hyperparasitaemia (>10% parasitaemia) is one of the WHO criteria for severe malaria () [Citation4]. Circulating parasitaemia does not accurately reflect total parasites in the body as P. falciparum sequesters in the microvasculature, out of circulation. Instead, plasma level of HRP2 is a stronger correlate of severe malaria and predictor of mortality than circulating parasitaemia [Citation38,Citation39]. The relationship between HRP2 and severity of malaria has been reported in several studies [Citation39–41], although others have found no relationship [Citation42]. A similar relationship has also been observed for P. vivax, in which circulating P. vivax specific LDH reflects the emerging importance of a sequestered parasite biomass in the spleen and bone marrow (as will be discussed below) [Citation43].

The threshold at which parasitaemia is associated with fever increases with age and is higher in holoendemic settings than mesoendemic settings [Citation44], suggesting that exposure-dependent acquisition of immunity is a key regulator or parasite density. Other factors including parasite multiplication rate, sequestration of mature IEs, and RBC mutations likely also contribute to parasitaemia, as will be discussed in this review.

The clinical impact of low-density infections is unclear. In a study of 2801 febrile children, there were no differences in severe clinical outcomes or secondary hospitalizations for individuals with low-density P. falciparum (who were not treated with antimalarials) compared to uninfected children [Citation45]. In a 2-year longitudinal study, persistent, low-density infections were found to oscillate between low and high density over time but individuals with oscillating parasitaemia remained afebrile and were untreated [Citation46]. In contrast, submicroscopic infection in Ugandan children was associated with both febrile and non-febrile illness [Citation47]. The consequences or benefits of low-density or chronic asymptomatic infection are largely unknown, although some researchers believe that all such infections should be treated [Citation48].

In the absence of mass drug administration regimes, low-density infections are untreated. Although individuals with submicroscopic infection are less likely to infect mosquitoes than microscopy detected infection [Citation49], submicroscopic infections can carry gametocytes [Citation50] and therefore are a potential “reservoir” for transmission. In a low, seasonal transmission area of Vietnam, 9% of P. falciparum and 46% of P. vivax infections lasted for over 6 months (during periods of low transmission) [Citation46], suggesting that chronic, low-density infections may maintain endemicity between malaria seasons.

Figure 1. Key pathogenic factors that contribute to severe malaria. High parasite biomass and parasite products released into circulation during cell lysis are stimulants for inflammation and may directly contribute to severe anaemia by reducing erythropoiesis. A strong inflammatory response, including Th1 type cytokine bias and sequestration of innate immune cells, such as monocytes, can activate and damage endothelial cells, enhance sequestration and contribute to severe anaemia by excessive complement mediated lysis of uninfected erythrocytes. A lack of antibodies targeting IE surface antigens may contribute to high parasite biomass and sequestration, although the key targets of antibodies are unclear. Sequestration of IE, mediated by PfEMP1, allows the parasite to evade clearance by the spleen and sequestration in the brain of young children is key to the pathogenesis of cerebral malaria. Sequestration in the placenta is strongly associated with poor pregnancy outcomes. Sequestration in the placenta is mediated by PfEMP1 expressed from VAR2CSA and sequestration in the brain microvasculature is associated with Group A PfEMP1 with domains that bind to ICAM-1 and EPCR. Protective genetic variants can reduce PfEMP1 expression, sequestration and rosetting.

Figure 1. Key pathogenic factors that contribute to severe malaria. High parasite biomass and parasite products released into circulation during cell lysis are stimulants for inflammation and may directly contribute to severe anaemia by reducing erythropoiesis. A strong inflammatory response, including Th1 type cytokine bias and sequestration of innate immune cells, such as monocytes, can activate and damage endothelial cells, enhance sequestration and contribute to severe anaemia by excessive complement mediated lysis of uninfected erythrocytes. A lack of antibodies targeting IE surface antigens may contribute to high parasite biomass and sequestration, although the key targets of antibodies are unclear. Sequestration of IE, mediated by PfEMP1, allows the parasite to evade clearance by the spleen and sequestration in the brain of young children is key to the pathogenesis of cerebral malaria. Sequestration in the placenta is strongly associated with poor pregnancy outcomes. Sequestration in the placenta is mediated by PfEMP1 expressed from VAR2CSA and sequestration in the brain microvasculature is associated with Group A PfEMP1 with domains that bind to ICAM-1 and EPCR. Protective genetic variants can reduce PfEMP1 expression, sequestration and rosetting.

Genetic variation in red blood cells

Several RBC polymorphisms occur at higher frequency amongst populations that have historically experienced a high burden of malaria and therefore are likely to offer an evolutionary survival advantage [Citation51]. It is well established that thalassaemia (reduced α or β haemoglobin) and heterozygous sickle cell haemoglobin (HbS) confer significant protection from severe malaria and are widespread in historically malaria endemic settings, particularly the Mediterranean and Sub Saharan Africa [Citation52]. Blood group O is also more common amongst malaria endemic populations and has been associated with protection from severe malaria [Citation51]. Interestingly, a recent study found a strong positive correlation between children with HbS and 3 genetic variants in the parasite genome and the protective effect of HbS against severe malaria was not observed in the presence of these parasite variants. This small study suggests a more complex evolutionary competition between host and parasite genomes may be at play [Citation53].

There are likely to be a number of host genetic traits that contribute to protection and are not yet characterized [Citation51]. Of recent interest, populations with Dantu (a polymorphism in RBC surface glycophorins) and mutations in the RBC membrane protein ATP2B4 have been associated with protection from severe malaria [Citation54–56]. However, these and other known polymorphisms (including HbS) only account for 5% of the total estimated genetic effect on the risk of severe malaria [Citation54], suggesting there are many other genetic variants influencing malaria pathogenesis.

Understanding the mechanisms by which naturally selected genetic traits contribute to protection enhances our understanding of malaria pathogenesis that may open doors to novel therapeutic strategies. For example, the Dantu mutation increases membrane tension and in turn inhibits merozoite invasion, suggesting that a membrane tension threshold may be a key factor for successful merozoite invasion [Citation57]. Several mechanisms of protection for haemoglobin mutations have been proposed, including reduced haemoglobin digestion by parasites and increased splenic clearance [Citation51]. Studies have shown that sickle cell trait [Citation58], haemoglobin C [Citation59] and a gain-of-function mutation in the mechanosensitive ion channel PIEZO1 E756del [Citation60] reduce surface presentation of the major variant surface antigen PfEMP1, in turn reducing adhesion to host receptors involved in parasite sequestration [Citation61]. In the case of blood types, IE have impaired ability to form rosettes (the adhesion of IE to uninfected erythrocytes) in blood group O erythrocytes [Citation62]. Both rosetting and sequestration are important virulence mechanisms that will be discussed below.

Cytoadherence, clumping, and rosetting

Cytoadherence and sequestration

Asexual P. falciparum IEs sequester in the heart, lungs, brain, skin, gastrointestinal tract, placenta, and other organs [Citation63]. Sequestration is generally thought of as a mechanism to avoid clearance by the spleen and may help avoid detection by circulating immune cells [Citation64]. Some sequestration sites have been linked to severe clinical outcomes, including cerebral malaria and placental malaria (as will be discussed in section 7).

Sequestration occurs when P. falciparum IE cytoadhere to receptors on endothelial cells or syncytiotrophoblast lining the placenta via cell surface antigens, particularly PfEMP1. Several possible endothelial adhesion receptors have been identified by in vitro and post-mortem histological studies, including Cluster of Differentiation 36 (CD36), intercellular adhesion molecule-1 (ICAM-1), E selectin, P-Selectin, platelet endothelial cell adhesion molecule-1 (PECAM-1), vascular cell adhesion molecule-1 (VCAM-1), endothelial protein C receptor (EPCR), integrins and fibronectins [Citation65], while chondroitin sulphate A (CSA) mediates placental sequestration [Citation33]. In addition to PfEMP1-mediated adhesion, the altered structure of the IE compared to non-IE is also important for sequestration. Knobless parasites that express PfEMP1 have impaired infectivity in monkeys [Citation66] and in humans [Citation67,Citation68] and this is thought to be due to reduced sequestration, allowing for efficient splenic clearance. Using a 3D in vitro model of the arteriole-capillary venules, parasites lacking knobs or PfEMP1 had reduced sequestration in narrow capillary constriction zones [Citation69]. The authors suggest a mechanism whereby reduced deformability and irregular surface structure decelerates IE flow in capillaries to allow for PfEMP1 adhesion and sequestration in post capillary venules [Citation69].

In addition to asexual parasites, post-mortem histology has revealed sequestration of immature gametocyte IE in the extravascular bone marrow parenchyma [Citation70], accounting for the lack of immature gametocytes seen in circulation. This has led to the hypothesis that gametocytes mature in the bone marrow parenchyma and re-enter circulation in preparation for being taken up by the mosquito. The mechanisms of sequestration of sexual stage parasites are unclear. In mice, P. berghei migrate through the sinusoidal barrier of the bone marrow, spleen, and liver and translocation into the bone marrow parenchyma was partially dependent on host P-selectin expression [Citation71]. Adhesion of gametocytes to human endothelial cell lines has been observed in some studies [Citation72] but not others [Citation73]. A recent study found that immature but not mature gametocytes bind to primary human bone marrow mesenchymal stromal cells in vitro and this activated the release of cytokines and growth factors, many of which are involved in angiogenesis [Citation74]. Adhesion was mediated by a trypsin-sensitive surface molecule that is unlikely to be PfEMP1, due to its low expression on gametocyte IEs [Citation74]. Gametocyte sequestration in bone marrow may be critical to sexual development and a clearer understanding of the mechanisms of sequestration may lead to opportunities for transmission blocking therapies. Interestingly, a recent study found that primaquine reacts with cytochrome P450 2D6 (CYP2D6), followed by cytochrome P450 NADPH:oxidoreductase (CPR) to generate excessive H2O2 that ultimately kills gametocytes, and proposed that the colocalization of CYP2D6 and CPR in the liver and bone marrow explains the efficacy of primaquine to kill hypnozoites and P. falciparum and P. vivax gametocytes [Citation75].

Sequestration in P. vivax

Asexual P. vivax-infected reticulocytes do not display PfEMP1-like antigens or knobs; however, there is some evidence for their cytoadherence to lung and placental endothelial cells in vitro [Citation76]. Low levels of apparent sequestration of asexual parasites have been observed ex vivo in the lung [Citation77], liver, and bone marrow parenchyma [Citation78], and like P. falciparum, immature P. vivax gametocytes sequester in the bone marrow parenchyma as a potential site of development and reservoir for infection [Citation78,Citation79].

Interestingly, a recent study of Plasmodium spp. infected, asymptomatic individuals undergoing splenectomy found massive sequestration of intact P. vivax IE of all stages in the spleen, at a rate 3590 times higher than the peripheral blood [Citation80]. P. falciparum IE also sequestered in the spleen but to a far lesser extent [Citation80]. It remains open to speculation whether these parasites can enter circulation and how this influences disease outcome.

Sequestration and the spleen

The spleen has multiple important functions in infection. Splenic macrophages and polymorphonuclear cells remove IE by phagocytosis [Citation81] and this may be impaired at high parasitaemia [Citation82]. The spleen also recycles erythrocytes by “pitting”- squeezing the IE through narrow passages into the red pulp sinuses that removes trophozoite, schizont, and some ring-stage parasites and releases the uninfected cell portion into circulation [Citation83,Citation84]. Studies of splenectomies in humans and monkeys have found that the spleen also modulates expression of knobs and variant surface antigens in P. falciparum IE [Citation82,Citation85] and modulates expression of parasite factors in P. vivax [Citation86]. This suggests that although the spleen may help the host by clearing or possibly with-holding parasites from circulation to minimize sequestration at other sites, sequestration in the spleen may perpetuate infection by acting as a chronic reservoir or by increasing surface antigen and knob expression that leads to sequestration.

Rosetting and clumping

Trophozoite IE infected with P. falciparum can bind to two or more uninfected red blood cells to form “rosettes.” This phenomenon is observed with some parasite lines but not others, suggesting it is a variant specific mechanism. Rosetting is also not associated with parasite density [Citation87–89], suggesting rosetting specific epitopes are involved rather than non-specific aggregation due to morphological or other generic changes. Rosetting is probably mediated by both variants of PfEMP1 and RIFINS (repetitive interspersed family proteins), although STEVORs (subtelomeric variable open reading frame proteins) may also be involved [Citation62,Citation90]. Rosetting is enhanced in non-blood group O compared to blood group O erythrocytes [Citation62], in particular blood group A and AB increase rosette rate and rosette strength [Citation91,Citation92]. Rosettes still form in blood group O suggesting other RBC surface receptors are also involved, and complement receptor 1, glycosaminoglycans (heparan sulphate) and glycophorin C on the surface of uninfected erythrocytes have been implicated [Citation62]. Serum factors including IgM and α2 macroglobulin binding to IEs can also mediate adhesion to uninfected erythrocytes in laboratory adapted and field isolates [Citation93,Citation94].

In several studies, rosetting was more common in isolates taken from individuals with cerebral malaria or with severe malaria anaemia [Citation87–89] but not in others [Citation95]. In line with the importance of blood group antigens and rosetting in severe malaria, blood group A/B/AB are at greater risk of severe malaria compared to blood group O [Citation96].

The mechanisms by which rosetting contributes to severe malaria are still unclear. Rosetting has been hypothesized to block or slow capillary blood flow, possibly enhancing cytoadherence [Citation97]. Rosetting may also inhibit immune mechanisms of parasite clearance. Non-O blood group rosettes block parasite epitopes (PfEMP1) from antibody binding and heparin disruption [Citation92,Citation98] and rosetting has been shown to reduce phagocytosis of IEs [Citation99,Citation100]. In a recent study, artesunate stimulated rosetting and artemisinin resistant parasites formed rosettes faster and at a higher rate than non-resistant parasites. The authors propose that rapid and frequent rosetting protects resistant parasites from intracellular artemisinin accumulation to perpetuate resistance [Citation100].

Rosetting has been occasionally described in other parasite species [Citation101], although its importance is less well understood. Rosettes produced by clinical isolates of P. vivax were similar in strength to those of P. falciparum and may alter IE deformability to enhance sequestration [Citation102]. Rosetting may be mediated by glycophorin C receptor or serum factors such as IgM [Citation62,Citation99]. P. vivax rosetting has been associated with anaemia [Citation103]; however, the contribution to severe disease, sequestration in the spleen or bone marrow, or influence on parasite load in vivo requires further study.

Aside from adhesion to uninfected erythrocytes, parasite IEs are also able to bind to CD36 or gC1qR/HABP1 on the surface of platelets to mediate clumping of IEs [Citation104,Citation105]. Clumping of IEs has been shown to be more common in parasites causing severe malaria than those causing uncomplicated malaria in some studies [Citation104,Citation106]. However, clumping was shown to be strongly associated with parasitaemia, rather than disease outcome, in another study [Citation107]. High assay variability in vitro makes it difficult to discern the importance of clumping in severe malaria [Citation108], and it is unknown whether specific PfEMP1 or other IE surface receptors are involved.

Virulent surface antigen diversity

Variant surface antigens (VSAs) are central to the pathogenesis of P. falciparum malaria. There are 4 main families of VSAs expressed during the blood stage of P. falciparum: PfEMP1, encoded by   60 var genes; RIFINs, encoded by 150–200 rif genes; Surface-associated interspersed gene family proteins (SURFINs), encoded by   10 surf genes; and STEVOR, encoded by 30–40 stevor genes [Citation109]. The large variation in surface antigens gives the parasite a survival advantage by evading epitope recognition and destruction by the humoral immune system [Citation110,Citation111]. Bull et al. showed that at the start of an infection, children had significantly fewer agglutinating antibodies to the IEs causing their infection than to other parasite isolates from the community, supporting the idea that the parasite exploits “holes in the antibody repertoire” [Citation112]. PfEMP1 is generally considered the most important VSA family due to its cytoadhering properties. In general, only one PfEMP1 type is expressed by a single parasite and there is often a dominant transcript in an infected individual. In addition to switching of PfEMP1 expression upon a5ntibody recognition, PfEMP1 expression is influenced by availability of adhesion receptors. In vitro, cultures of IEs panned on a particular receptor will favour expression of PfEMP1 that can adhere to the receptor [Citation113]. A well-characterized example in vivo is that var2csa becomes the dominant var gene to be expressed by parasites infecting pregnant women and encodes for PfEMP1 that binds to placental CSA receptors [Citation114]. The wide variation in var genes allows the parasite to adapt to different adhesion environments to avoid splenic clearance. Additionally, multiple var genes of the same binding phenotype exist, allowing the parasite to maintain adhesion to particular receptors despite antibody recognition [Citation115].

Multigene families have been identified in P. vivax, including the vir gene family, but are less well characterized due to high antigenic variation and difficulty maintaining cultures in vitro. Different VIR protein subfamilies are trafficked to different cellular locations, including the reticulocyte surface (for VIR14 of subfamily C) where they have been speculated to be involved in cytoadherence [Citation116]. Transgenic P. falciparum IE expressing VIR14 has been shown to bind to ICAM-1 and human spleen fibroblasts in vitro [Citation86,Citation116]. A small number of VIR proteins have been shown to be immunogenic and antibody responses to 2 VIR antigens were associated with protection from low birthweight in P. vivax-infected pregnant women [Citation117]. It remains open to speculation as to whether VIR antigens contribute to immune evasion as with VSAs in P. falciparum and whether other VIR or non-VIR proteins are involved in cytoadherence of P. vivax.

PfEMP1 types associated with disease outcomes

Several studies have found associations between expression of specific PfEMP1 subclasses, domains or combinations of domains (domain cassettes or DCs) and disease outcomes, supporting the idea that there are virulent var genes/PfEMP1 types. Var genes have been classified into groups A, B, C, and E and two intermediate groups B/A and B/C, based on their chromosomal location and type of upstream promotor sequence (UPS A, B, C, or E) [Citation118]. Interestingly, parasites infecting cerebral and non-cerebral severe malaria patients often have upregulated expression of Group A and B/A var genes compared to uncomplicated malaria patients [Citation119–122]. Additionally, PfEMP1 with adhesion phenotypes that are associated with severe or cerebral malaria are typically encoded by group A or B/A var genes, rather than group B or C [Citation123,Citation124]. Expression of group B PfEMP1 has also been associated with clinical or severe malaria in some studies [Citation120,Citation125] but not in others [Citation126], as has expression of Group C var genes [Citation119].

Individual var genes are composed of arrays of Duffy binding-like domains (DBLs) and cysteine-rich interdomain regions (CIDRs), in various combinations that can be subclassified into DBL α0.1–2, β1–10, γ1–18, δ1–9, ε1–14, ζ1–6 and CIDR α1–6, β1–7, γ1–12, γ1–2 [Citation118,Citation127]. Domain subclasses range from 38% to 98% sequence homology between parasite genomes of laboratory adapted parasite lines [Citation118]. Some domains have been shown to adhere to specific endothelial cell receptors and have been associated with disease presentations. Adhesion to CD36 is the most common property of PfEMP1 and occurs via the CIDRα of the head structure – specifically, CIDRα2–6 domains of Group B or C PfEMP1 [Citation128,Citation129] that are present in 70% of var genes [Citation130]. Adhesion to CD36 has been frequently associated with parasites causing uncomplicated, rather than severe malaria [Citation130–133]. CD36 is not expressed by brain endothelial cells [Citation134] and IE binding to human brain endothelial cells in vitro is independent of CD36 [Citation135], but platelets which express CD36 can act as a bridge between IEs and brain endothelial cells [Citation136]. Thus, the role of CD36 adhesion in cerebral malaria remains unclear. The major variants of PfEMP1 that have been associated with specific malaria syndromes are those that bind EPCR (involved in severe malaria); variants that bind both ICAM-1 and EPCR (involved in CM); a unique variant, VAR2CSA, that binds CSA (involved in placental malaria); and variants that bind uninfected RBCs (involved in rosetting), as will be discussed below.

EPCR binding PfEMP1 associated with severe malaria

Group A PfEMP1s with head structures containing a CIDRα1.1 or 1.4–1.8 domain bind to EPCR and this includes conserved domain cassette 13 (DC13, group A) and domain cassette 8 (DC8, group B/A) [Citation137]. EPCR is expressed on multiple endothelial cell types, and IEs containing parasites that express DC8 and DC13 are able to bind to brain, lung, heart, dermis and bone marrow endothelial cells [Citation138]. Transcription of DC8 and DC13 has been shown to be upregulated in parasites causing cerebral malaria [Citation121,Citation123] and various individual group A and B CIDRα1 domains predicted to bind EPCR have also been associated with cerebral malaria [Citation132,Citation139–141]. Evidence is inconsistent as to whether particular CIDRα subclasses may be contributing to a specific malaria pathology, such as brain swelling [Citation140], retinopathy positive cerebral malaria [Citation140,Citation142], severe malarial anaemia [Citation123,Citation142], and respiratory distress [Citation143]. Further studies are needed to understand the mechanisms by which EPCR binding IE directly or indirectly contribute to severe or cerebral malaria.

Dual ICAM-1-EPCR binding PfEMP1 associated with cerebral malaria

Post-mortem immunohistochemical studies of cerebral malaria patients first showed ICAM-1 mediated adhesion of IEs to cerebral blood vessels [Citation134]. ICAM-1 is upregulated on brain vascular endothelial cells by inflammatory cytokines such as TNF [Citation135] and is widely expressed in the brain, like EPCR, but unlike CD36 or E-Selectin [Citation134]. ICAM-1 binding is mediated by DBLβ domains, including the DBLβ3 domain of group A DC4 [Citation124,Citation144]. A short sequence of amino acids known as the Lennartz ICAM-1 binding motif, can be used to predict ICAM-1 binding for all group A and some group B/A DBLβ3 domains [Citation124,Citation145]. Group A, ICAM-1 binders that contain the Lennartz ICAM-1 binding motif also have an upstream CIDRα1 domain that can bind to EPCR. Group B or C ICAM-1 binding IE (that do not have the motif) often bind to both CD36 and ICAM-1 [Citation124].

Expression of var genes containing the ICAM-1-binding motif, that are predicted to induce binding to ICAM-1 and EPCR, is thought to be involved in the pathogenesis of cerebral malaria [Citation33]. Transcripts of var genes containing the ICAM-1-binding motif are upregulated in parasites from patients with cerebral malaria compared to severe anaemia and uncomplicated malaria [Citation124,Citation140] and in severe malaria compared to uncomplicated malaria [Citation146]. Dual ICAM-1-EPCR binding IE were more common in patients with cerebral malaria than in patients with uncomplicated malaria, whereas CD36 binders were more frequently found in patients with uncomplicated malaria [Citation132]. In a study of parasite transcripts from 45 children, expression of var genes predicted to bind to EPCR but not ICAM-1 was associated with severe malaria but not with cerebral malaria or severe malaria anaemia alone, whereas transcription of var genes encoding both ICAM-1 and EPCR binding was associated specifically with cerebral malaria [Citation124]. In a recent study, CIDRα-DBLβ1/3 domains predicted to bind ICAM1 and EPCR were similarly expressed in isolates causing uncomplicated and cerebral malaria [Citation139].

CSA binding PfEMP1 VAR2CSA is associated with placental malaria

Perhaps the most well-characterized var gene is var2csa. Var2csa is highly conserved – the 6 commonly-identified DBL domains and interdomain regions have 58–90% sequence homology across 7 different parasite genomes – and has a unique promotor sequence, UPS E [Citation118]. Var2csa is the dominant var gene transcribed in parasites infecting the placenta [Citation114,Citation147,Citation148]. VAR2CSA binds to chondroitin sulphate A, a glycosaminoglycan expressed on placental syncytiotrophoblasts [Citation114], via the DBL2× domain and adjacent interdomain regions [Citation149], to mediate placental sequestration of IE. Placental malaria infection is associated with poor birth outcomes [Citation1].

Protective immunity against severe malaria

Sterile immunity (i.e. absence of infection) is not acquired with natural exposure to malaria, however protective immunity against high-density parasitaemia and clinical disease is acquired with repeated exposure and is lost if exposure is interrupted (reviewed in [Citation150]). Exposure-dependent acquisition of immunity likely explains why severe and clinical malaria is most prevalent in children under 5, although maturation of the immune system with age also likely contributes to protection [Citation151]. Immunity to severe, non-cerebral malaria is acquired after 1 or 2 childhood infections in high transmission settings [Citation152], whereas in low and moderate transmission settings the acquisition of immunity may be slower and result in a higher likelihood of severe disease [Citation150]. This raises concerns about the impacts of interventions that reduce the incidence of malaria partially but not dramatically [Citation153] and highlights the importance of understanding markers and mechanisms of naturally acquired immunity.

A pioneering study in the 1960s showed that transfer of immunoglobulin G (IgG) from serum of immune adults to non-immune children with malaria could dramatically reduce blood-stage parasite burden and fever, whereas transfer of non-immune serum and immunoglobulin-depleted serum had a far lesser effect [Citation154]. This study suggested that malaria antigen-specific antibodies are important mediators of parasite clearance and protection from blood-stage malaria infection, and initiated the search for targets of IgG that may be valuable vaccine candidates or biomarkers of disease. Protein microarrays have allowed for large scale screening of immunogenicity to hundreds of antigens, including whole protein families [Citation155] or dominant antigens that are associated with severe malaria. There are now multiple known targets of antibodies at different stages of the parasite life cycle, however a relatively small number of antigens have been associated with protective immunity and fewer have been developed into vaccines. The following sections focus on the contribution of antibody mediated immunity to protection from malaria at the various parasite lifecycle stages.

Immunity to mosquito/sexual stage antigens

Little is known about the contribution of the immune system to reduced levels of circulating or sequestering gametocyte IEs. Stage 1 gametocyte IEs are phagocytosed by monocytes and macrophages in vitro, likely via CD36 recognition of trypsin-sensitive surface antigens [Citation156]. Naturally acquired antibodies to the surface of gametocyte IEs have been reported [Citation157] however antibody levels are dramatically lower than those to PfEMP1 on IEs [Citation158] and the functions of these antibodies are largely unknown. Antibodies to gametocyte surface antigens, such as Pfs230 and Pfs48/45, are more prominent than responses to the gametocyte IEs surface [Citation158] and are associated with reduced infectivity of mosquitoes fed on P. falciparum gametocytes grown in vitro [Citation159] or in individuals [Citation160]. Early studies reported gametocyte-specific antibodies that reduce mosquito infectivity with P. vivax [Citation161] although the target antigens are not well characterized. Antibodies targeting gametocyte stage antigens are taken up with gametocytes by mosquitoes during a blood feed, and they can inhibit mosquito stage development of the parasite’s lifecycle [Citation162]. Gametocyte, zygote, and oocyst antigens present attractive opportunities to develop transmission blocking vaccines against antigens such as Pfs25/Pvs25 and Pfs28 that are not exposed to the human immune system but may be targets of vaccine induced immunity [Citation163].

Immunity to pre-erythrocytic stage antigens

CD8+ T cells are important in defence against pre-erythrocytic stage parasites [Citation164]. Individuals develop CD8+ T cells specific for the circumsporozoite protein (CSP) in malaria endemic areas and following vaccination [Citation165], and in vivo imaging of mouse livers has shown CD8+ T cells primed for CSP can locate and destroy Plasmodium in infected hepatocytes [Citation166].

CSP is the major antigen coating sporozoites prior to invasion of hepatocytes. Antibodies to P. falciparum CSP are naturally acquired with age [Citation167], albeit slowly, and can promote opsonic phagocytosis [Citation167], inhibit hepatocyte invasion and inhibit sporozoite motility [Citation168,Citation169]. Antibodies to P. vivax CSP are acquired following controlled human malaria infection and are sustained in the absence of re-infection [Citation170]. Sterile protection does not occur naturally and naturally acquired antibodies to P. falciparum or P. vivax CSP have been associated with protection from clinical malaria in some studies but not others [Citation168,Citation171–173]. Despite this, the first WHO endorsed vaccine for malaria, RTS,S/AS01, is a virus-like particle made up of a recombinant fragment of CSP coupled with hepatitis-B surface antigen. It has been successfully implemented in Ghana, Kenya and Malawi [Citation174], and endorsed for wider use. Antibodies to CSP are induced by vaccination and can promote phagocytosis and fix complement [Citation175]. RTS,S/AS01 was estimated to provide 36% protective efficacy for clinical malaria over 4 years in a phase 3 clinical trial [Citation176] but may be improved by administering a fractional, rather than full, booster dose at 12 months [Citation177]. The R21 vaccine, a variation to RTS,S (that displays a higher ratio of CSP to hepatitis B surface antigen), showed more than 70% protective efficacy against uncomplicated malaria in a phase 2b clinical trial [Citation178]. In an area of highly seasonal malaria, annual vaccination with RTS,S prior to the malaria season combined with seasonal chemoprophylaxis had a protective efficacy of 63% against clinical malaria, 70% against hospitalization with SM and 73% against death, when compared to seasonal chemoprophylaxis alone [Citation179]. The ongoing development of RTS,S/AS01, which initially showed modest protective efficacy, has arguably set precedence for continued development of vaccines that show low efficacy in phase 1 and 2 clinical trials.

Additionally, live-attenuated sporozoites or live sporozoites administered simultaneously with chemoprophylaxis can induce sterile protection from infection against homologous controlled human malaria infection (CHMI) challenge [Citation180]. The vaccines showed limited efficacy against heterologous parasite challenge [Citation181] but have been shown to invoke sterile protection when co-administered with prophylactic drugs [Citation182]. Efficacy was significantly lower in individuals from malaria endemic regions compared to US malaria naïve individuals [Citation180] however altered dosing regimens may overcome these differences [Citation183]. In a recent phase 2 clinical trial in children aged 5–12 months, the attenuated sporozoite vaccine gave no significant protection from infection, possibly due to poor T cell immunity, suggesting these vaccines may not be efficacious in this age group [Citation184].

Immunity to blood-stage antigens – merozoites

During the blood stage of infection, monocytes, neutrophils and dendritic cells recognize excess GPI on the merozoite via Toll like receptors (TLRs, a type of pattern recognition receptor), as well as haemozoin and parasite DNA released from the ruptured RBC. This initiates cell killing mechanisms such as phagocytosis and the production of proinflammatory cytokines that activate NK cells and T cells [Citation185]. The innate immune system is enhanced by circulating serum factors including complement components and mannose binding lectin, that lead to pore insertion in the pathogen cell membrane and osmotic lysis of merozoites, but not IEs [Citation186].

Antibodies to P. falciparum merozoites function to block erythrocyte invasion ligands [Citation187,Citation188] as well as activate the complement cascade and lead to phagocytosis [Citation189–191]. Studies have primarily focused on antibody responses to merozoite proteins involved in invasion or in high abundance on the merozoite surface, including merozoite surface proteins (MSPs), erythrocyte binding proteins (EBAs), reticulocyte binding protein homologues (RHs) and apical membrane antigen-1 (AMA1) [Citation192], although multiple other merozoite antigens exist [Citation193,Citation194]. Antibody responses to several merozoite antigens including P. falciparum glutamate-rich proteins (GLURPs), MSP1, MSP2, MSP3, MSP7, RH5, RIPR, and EBA175 have been associated with protection from experiencing an episode of symptomatic infection or protection from symptomatic compared to asymptomatic infection [Citation195–200]. Several vaccines containing merozoite antigens have reached phase 2 clinical trials and are under continued development, including AMA1 [Citation201], Rh5 [Citation202], and combined GLURP and MSP3 [Citation203]. Of note, recently, Rh5.1 vaccination was found to significantly reduce parasite growth rate following CHMI [Citation202].

However, other studies have reported a lack of association between antibody responses to merozoite antigens and protective immunity, including in regards to MSP1 or AMA1 [Citation204], or have found that protective associations are limited to older children [Citation205]. Antibody acquisition is antigen specific, with antibodies to some antigens acquired at a young age and to others later in life or after exposure to high parasitaemia [Citation206]. It has been suggested that antibodies to merozoite antigens may be more useful as markers of exposure [Citation207–209]. Similarly, antibodies against P. vivax merozoite antigens can be used to predict recent infection [Citation210,Citation211]. However, further research is required to understand which antigens are most appropriate to monitor for exposure in varying populations, such as those in high transmission rather than low transmission settings [Citation212].

Immunity to infected erythrocyte surface antigens

Innate immune cell scavenger receptors recognize surface markers on the IE membrane, such as lipid phosphatidylserines that are usually confined to the inner-erythrocyte membrane [Citation213], increased Band 3 protein [Citation214] and PfEMP1 [Citation215], and IE recognition can initiate phagocytosis of the IE and production of proinflammatory cytokines and activation of NK cells and T cells [Citation216]. Interestingly, PfEMP1 variants have been shown to directly contribute to NK cell activation and cytokine secretion [Citation217,Citation218] and other PfEMP1 variants may have an inhibitory effect [Citation219], as do some RIFIN variants [Citation220]. It is therefore possible that antibodies targeting specific PfEMP1 variants may either suppress or enhance antibody independent NK cell activation during the blood stage of infection.

PfEMP1 is the dominant IE VSA targeted by antibodies, since parasites that do not express PfEMP1 have markedly lower antibody recognition by immune sera [Citation221]. Several studies have found an association between antibodies to certain variants of PfEMP1 and protection from severe, cerebral or uncomplicated malaria, and will be summarized below. Almost all RIFINs, STEVORs and SURFINs are also immunogenic [Citation222], although less studied. Antibodies to specific variants have been associated with a reduced prospective risk of developing febrile malaria [Citation222] and a lack of antibodies to specific RIFINS and STEVORs was identified in Malian children with severe malaria compared to uncomplicated malaria [Citation223]. Other more conserved antigens have been identified on the IE surface [Citation224] although less is known about their immunogenicity.

Antibodies targeting trophozoite and schizont stage IEs have multiple functions, including inhibiting rosetting [Citation89]; blocking adhesion to endothelial/epithelial cell receptors [Citation119]; opsonising IE for phagocytosis by monocytes [Citation205,Citation225]; or initiating NK cell mediated lysis [Citation217,Citation226]. The contribution of each of these antibody mediated mechanisms to reduce blood-stage infection and protect from severe malaria requires further study. Opsonic phagocytosis of IE displaying PfEMP1 associated with severe malaria has been associated with protection from severe malaria [Citation227] and opsonic phagocytosis of IE expressing VAR2CSA with protection from malaria in pregnancy [Citation228] however it is not clear if some variants are better at inducing cytophilic, opsonising antibodies than others. Additionally, antibodies targeting late-trophozoite stage antigen glutamic-acid-rich protein (GARP) initiate programmed cell death in vitro and a lack of anti-GARP antibodies has been associated with risk of severe malaria in one study [Citation229]. In the late schizont stages, IEs become permeable and antibodies can access intracellular antigens [Citation230]. Antibodies that bind to intracellular Schizont Egress Antigen-1 (SEA-1) have been associated with protection from severe malaria [Citation231,Citation232] although the mechanism of protection are unclear [Citation233]. Unlike merozoites, it is thought that IEs have mechanisms to evade complement fixation and lysis, possibly by upregulating complement regulatory proteins such as CD59 [Citation186], or the orientation of PfEMP1 on knobs may prevent antibody hexamerization required for complement fixation [Citation234]. In a recent study, complement component-1 (C1s) cleaved PfEMP1 at semi-conserved interdomain sites, found in approximately 80% of PfEMP1, which suggests that this may be a mechanism to reduce endothelial cell binding when the host is in a hyperinflammatory state and at risk of dying, or to escape opsonic phagocytosis [Citation235].

Antibodies targeting specific PfEMP1 domains and associations with protection from severe malaria

Due to the key pathogenic role of PfEMP1 in mediating sequestration and rosetting during blood-stage P. falciparum infection and the clear associated of particular var genes or PfEMP1 receptor binding phenotypes with severe cerebral and placental malaria, several studies have hypothesized that antibodies targeting virulent PfEMP1 types may protect from severe malaria. Antibodies to severe forms of malaria are acquired early in life in high transmission settings and severe infections are most common in children under 5 [Citation236]. In line with a role of group A or B/A PfEMP1s in severe infections, antibodies to group A or B/A domains are more common in children under 5 and are generally acquired more rapidly than antibodies to group B, B/C, or C domains [Citation126,Citation237], although this pattern was only found for DBL and not CIDR domains in one study [Citation238]. Preferential acquisition of antibodies targeting group A PfEMP1 in children may be due to the greater sequence conservation of group A var genes than other var genes or may reflect preferential expression of group A var genes in the naïve human host compared to hosts with pre-existing immunity, as shown in a CHMI study [Citation239].

Antibody responses to VAR2CSA and malaria in pregnancy

In high transmission settings, women in their first pregnancy and men have low levels of VAR2CSA-specific antibodies and women acquire VAR2CSA-specific antibodies in a gravidity-dependent manner [Citation240]. Although antibodies to VAR2CSA block adhesion of IEs to CSA [Citation240], a systematic review found that levels of antibodies to VAR2CSA and VAR2CSA expressing IEs measured at delivery are markers of placental infection, rather than markers of protection [Citation241]. Rather than total levels of antibodies, a combination of IgG3 and functional antibody responses, including binding inhibition and phagocytosis, could collectively predict protection from placental infection in a recent study [Citation225]. The glycosylation state of antibodies to VAR2CSA may also be important for protection. Naturally acquired antibody to PfEMP1 is often afucosylated, which increases its ability to interact with FcγRIIIa, expressed on NK cells and others [Citation242].

The relationship between antibodies to pregnancy-specific IE and low birthweight is complex. Antibodies to VAR2CSA-expressing IE have been associated with reduced risk of low birthweight in some studies [Citation243–246], but antibodies to individual VAR2CSA domains were not associated with birthweight or gestational age in another study [Citation247]. In a recent meta-analysis, there were no significant associations between IgG responses to VAR2CSA domains and low birthweight [Citation241]. Two vaccines have been developed based on the minimum CSA binding domain of VAR2CSA, PRIMVAC [Citation248] and PAMVAC [Citation249]. Both vaccines were well tolerated and immunogenic in Phase 1a clinical trials, but gave limited antibody recognition of heterologous IE [Citation248]. PRIMVAC did not elicit antibodies that inhibit binding of IE to CSA in vitro [Citation248] and it will be interesting to know whether these vaccines induce other functional antibodies. Antibodies to full- length VAR2CSA proteins show greater cross reactivity and future vaccines might need to include multiple VAR2CSA proteins, or more extensive sections of the protein [Citation250]. While the role of VAR2CSA outside of placental malaria is still not clear, there is hope that vaccines against other PfEMP1 types will be developed to combat severe pathologies such as CM.

Antibody responses to CD36 binding phenotype associated with UM

IE that adhere to CD36 have generally been associated with infections causing uncomplicated rather than severe malaria and antibodies targeting CD36-binding CIDRα among other domains, are predictive of a reduced prospective risk of clinical malaria and severe malaria in some studies [Citation155,Citation251]. Malian children with cerebral malaria lack antibodies to both CD36 and non-CD36 binding PfEMP1 fragments compared to uncomplicated malaria [Citation223]. It seems presently unclear whether antibodies to CD36 binding PfEMP1 contribute to protection from severe malaria.

Antibody responses to EPCR binding IE associated with SM

In line with a role of EPCR binding CIDRα1 domains in severe malaria, antibodies against these domains or downstream domains from EPCR binding PfEMP1, are significantly elevated after an episode of severe malaria [Citation227,Citation252] and cerebral malaria [Citation223]. Antibodies targeting EPCR binding domains or downstream domains are associated with protection from severe malaria [Citation227,Citation251,Citation253] as well as reduced prospective risk of uncomplicated malaria in one study [Citation155] (). In contrast to these protective associations, a recent study showed that antibodies to CIDRα1.4 in young Malian children were predictive of prospective infection with higher parasite density [Citation255]. Further studies are needed that examine functional antibodies to EPCR-binding PfEMP1s and to IEs expressing these domains. Such studies can help determine whether naturally-acquired antibody to these domains is protective, and can help identify the exact targets and features of protective antibody responses.

Table 1. Key PfEMP1 types associated with clinical outcomes and evidence of antibody-mediated protection from any clinical outcome. Summarizes studies included in the text only.

Antibody responses to dual ICAM-1 and EPCR binding IE associated with CM

Antibodies against the DBLβ3 domains with the Lennartz motif have been associated with reduced risk of uncomplicated malaria [Citation155,Citation254] and non-cerebral severe malaria [Citation126], and are boosted after an episode of severe malarial anaemia [Citation223]. In other studies, there was no difference in antibodies between uncomplicated and severe non-cerebral malaria [Citation145], or uncomplicated and cerebral malaria [Citation258]. Although expression of dual ICAM-1-EPCR binding PfEMP1 is associated with cerebral malaria and antibodies targeting these domains can efficiently block adhesion of IE in vitro [Citation145], further studies are needed to confirm if naturally acquired antibodies to these parasites are associated with protection from cerebral malaria. In a recent study, Beninese children with uncomplicated malaria had higher antibodies to a recombinant Group A DBLβ3 and a CIDRα1.4-DBLβ3 couplet (predicted to be dual ICAM-1 and EPCR binding) than children with cerebral malaria. No difference between cerebral and uncomplicated malaria was observed in the subset of children confirmed to be infected with parasites expressing Group A DBLβ3 with the motif (presumed to be dual binders), although only a single recombinant DBLβ3 was tested [Citation139]. Interestingly, in another small cohort, total IgG targeting full length DC13 (which is dual ICAM-1 and EPCR binding) was boosted in individuals with uncomplicated malaria from presentation to convalescence, whereas IgG1 and IgG3 to this protein were boosted in convalescence from cerebral malaria [Citation259]. This suggests that the quality of the antibody response to dual ICAM-1 and EPCR binding PfEMP1 may be a more effective correlate of protection from cerebral malaria.

Antibody to domains associated with rosetting

Antibodies raised against recombinant DBLβ1 domains of rosetting parasite lines are able to bind and inhibit rosette formation [Citation256,Citation261] and induce phagocytosis of IEs [Citation262]. Antibodies to recombinant domains predicted to be from rosetting parasite lines are elevated in uncomplicated malaria compared to severe malaria and are associated with reduced prospective risk of severe malaria [Citation251,Citation257] and uncomplicated malaria [Citation237]. Furthermore, antibodies against rosetting parasite lines were elevated in children with uncomplicated malaria compared to severe malaria and to children with cerebral malaria [Citation89,Citation257].

Cytokine response

A strong cytokine response is triggered by Plasmodium-derived toxins that are released into circulation during cell lysis, including glycosylphosphatidylinositol (GPI), haemozoin, and cell-free DNA and RNA. Toxins are recognized by pattern recognition receptors, including TLR4 on the surface of innate immune cells, TLR7, TLR8, and TLR9 on endosomes and cytosolic cGAS and AIM2 to induce production of a type 1 interferon response and pro-inflammatory cytokines (reviewed in [Citation263]). During the liver stage of infection, hepatocytes detect sporozoite RNA via cytosolic MDA5 to produce a Type I IFN response that primes NK cells, NKT cells and DCs and recruits CD8+ T cells, although the contribution of this response to preventing subsequent disease is unclear [Citation263,Citation264]. Malaria is associated with an increase in circulating pro-inflammatory cytokines, including IL-1α, IL-1β, IL-6, IL-12, IL-18, and TNF-α, as well as anti-inflammatory cytokines, particularly IL-10, and chemokines including MIP1, IL-8, CCL5, and others [Citation185,Citation265]. Cytokines and chemokines recruit and activate innate immune cells including neutrophils, monocytes, macrophages, dendritic cells, and NK cells for innate immune killing mechanisms, such as phagocytosis [Citation185]. Interestingly, P. falciparum infection leads to epigenetic changes in monocytes that increase the level of secretion of IL-6 and TNF-α upon secondary stimulation one month after infection [Citation266], suggesting “trained immunity” affects cytokine responses and potentially contributes to long-term protection.

Alongside the importance of type 1 interferons and pro-inflammatory cytokines/chemokines in mounting an effective immune response against liver and blood-stage malaria, several studies have associated a dysregulated pro-inflammatory cytokine response with severe malaria, cerebral malaria, and death [Citation265,Citation267–269]. Larger cohort studies using consistent markers are required to evaluate whether a particular cytokine combination may have prognostic value for severe clinical outcomes [Citation265,Citation270]. Cytokines activate human endothelial cells in vitro [Citation271] and circulating cytokines have been proposed to contribute to localized brain inflammation in patients with cerebral malaria, ultimately leading to impaired blood–brain barrier integrity and brain swelling that is associated with death [Citation33,Citation265]. Endothelial cells (including human brain endothelial cells) also secrete cytokines when stimulated with IEs in vitro [Citation272] that may perpetuate localized inflammation in vivo. In a study of children with cerebral malaria, there were no differences in peripheral cytokine levels of children with severe brain swelling compared to no severe brain swelling [Citation273]. This may reflect differences in the peripheral cytokines compared to a local environment or suggest that secondary factors are involved in brain swelling [Citation273].

Malaria also disrupts the balance of pro-inflammatory and anti-inflammatory cytokines required for a healthy pregnancy and several maternal cytokines have been associated with poor pregnancy outcomes including preterm birth, small for gestational age babies, and pregnancy loss [Citation274–276]. Placental inflammation is strongly linked to poor pregnancy outcomes, due to impaired placental development or function (see section 9.4), and the local placental cytokine environment can differ significantly from the maternal periphery [Citation277], adding a layer of complexity to finding prognostic markers.

Molecular basis of severe disease phenotypes and symptoms

Cerebral malaria

Unarousable coma from malaria is known as cerebral malaria. Histologically, it is characterized by the sequestration of IE in the brain, typically in the capillaries, as well as pre-capillary arterioles and post-capillary venules. In the majority of cases, sequestration is accompanied by microvasculature pathologies, particularly haemorrhages in the brain white matter, thrombosis and accumulation of monocytes [Citation63].

Cerebral malaria in children with documented malaria infection is clinically diagnosed by the Blantyre coma score, based on motor, verbal and eye movement responses. This diagnostic method overestimates cerebral malaria cases by up to 30% in a high transmission settings and up to 54% in low or moderate transmission settings [Citation278,Citation279]. The retina reflects changes to the brain microvasculature and retinal examination for retinal whitening and blood vessel changes can drastically increase diagnosis sensitivity and specificity [Citation280]. A normal retinal examination in individuals with impaired consciousness and parasitaemia should lead to consideration of other causes of coma, but may also reflect an earlier stage of cerebral malaria with retinopathy [Citation281,Citation282]. An incorrect clinical diagnosis of cerebral malaria may have devastating consequences, resulting in delayed treatment of alternative, life threatening conditions, and may underpin discrepancies in surveillance and epidemiological research.

Increased brain volume due to swelling in the brain white matter [Citation283] is common to almost all children who die from cerebral malaria [Citation284] and the final cause of death is respiratory arrest due to pressure on the brain stem [Citation284]. In adults, swelling is more pronounced in the basal ganglia and fatal cerebral malaria is associated with severe hypoxia, rather than pressure on the brainstem [Citation283], suggesting potentially different mechanisms of fatality in adults and children. Mortality from cerebral malaria in Africa is between 15% and 25% [Citation285]. Amongst survivors, up to 30% experience neurological impairment, which often manifests as epilepsy, motor and language regression and greater risk of developing disruptive behavioural disorders [Citation286–288]. The mechanisms of long-term neurological impairment are unknown. Elevated levels of tau in cerebral spinal fluid and increased transcriptions of genes linked to Alzheimer’s disease may be involved [Citation289,Citation290]. In a recent study, examination of the retina revealed that leaks from endothelial cell tight junctions were associated with neurological disability, whereas larger haemorrhagic breaches in the endothelium were associated with brain swelling and death [Citation291].

The current understanding of the pathogenesis of cerebral malaria [Citation33] centres around activation of the endothelial cells leading to impaired blood–brain barrier integrity and ultimately neuroinflammation, as will be summarized below ().

Figure 2. Sequestration in Plasmodium falciparum. Reading left to right, the brain colour changes from light tan to a “slate grey” colour in cases of fatal cerebral malaria. Mature infected erythrocytes (IEs) sequester in the deep vasculature, including cerebral vessels. Receptors on endothelial cells include endothelial protein C receptor (EPCR), ICAM-1 and heparan sulphate proteoglycans. Platelets can clump IEs, and bridge between CD36 binding IEs and endothelium. IE can activate monocytes and neutrophils to release cytokines and immune mediators, which can upregulate ICAM-1 expression, and induce damage to the blood–brain barrier. Fluid leakage through and between cerebral endothelial cells can lead to neuronal activation and may result in potentially fatal brain swelling, associated with brain discolouration.

Figure 2. Sequestration in Plasmodium falciparum. Reading left to right, the brain colour changes from light tan to a “slate grey” colour in cases of fatal cerebral malaria. Mature infected erythrocytes (IEs) sequester in the deep vasculature, including cerebral vessels. Receptors on endothelial cells include endothelial protein C receptor (EPCR), ICAM-1 and heparan sulphate proteoglycans. Platelets can clump IEs, and bridge between CD36 binding IEs and endothelium. IE can activate monocytes and neutrophils to release cytokines and immune mediators, which can upregulate ICAM-1 expression, and induce damage to the blood–brain barrier. Fluid leakage through and between cerebral endothelial cells can lead to neuronal activation and may result in potentially fatal brain swelling, associated with brain discolouration.

Several factors have been proposed to contribute to activation and impaired integrity of brain blood vessel endothelial cells. Sequestration of IEs in the brain microvasculature is strongly associated with coma and it has been hypothesized that microvascular congestion due to sequestration of IEs, platelet mediated clumping or rosetting causes hypoperfusion and ischaemic damage to the endothelium [Citation292]. IE, particularly in the schizont stage, can directly activate endothelial cells and trigger apoptosis [Citation33]. EPCR binding IE compete for EPCR binding with activated protein C (APC), involved in regulation of blood coagulation, inflammation, and endothelial cell integrity and apoptosis [Citation293]. It has been proposed that reduced APC binding results in downregulation of EPCR, which may decrease adhesion of EPCR binding IE, but also upregulation of ICAM-1 that can preserve adhesion of ICAM-1-EPCR dual binding IE as well as increase platelet activation and endothelial barrier breakdown [Citation33]. A recent study showed that in vitro, human brain endothelial cells can phagocytose dual ICAM-1-EPCR binding IE in an ICAM-1 dependent manner, and parasites engulfed in brain endothelial cells were observed in ex vivo samples from two cerebral malaria patients [Citation294], highlighting a novel potential pathway of endothelial cell activation or defence.

In addition to IE, several parasite factors are thought to contribute to endothelial cell activation, including haemozoin and products bound to haemozoin [Citation295], HRP2 [Citation296], histones [Citation297], and kinins [Citation298] released from IE. Local inflammatory factors have also been shown to contribute, such as matrix metalloprotein 8 released from leukocytes, reduced endothelial nitric oxide bioavailability, and pro-inflammatory cytokines [Citation33,Citation265], although neither inhaled nitric oxide nor anti-TNFα therapy showed any potential as adjunctive therapies [Citation299,Citation300]. In the P. berghei ANKA mouse model of experimental cerebral malaria (ECM), CD3+CD8+ T cells in the brain vasculature are considered key drivers of intense inflammation and disruption of the blood–brain barrier [Citation301]. Recent studies suggest that similar T cells are also present in the brains of children with cerebral malaria [Citation302], although with potentially different compartmentalization [Citation303], therefore the role of T cell accumulation in the brain of children with cerebral malaria requires further elucidation.

In response to IEs, parasite factors and inflammatory mediators, the activated endothelium expresses local inflammatory cytokines to perpetuate inflammation and dysregulates factors that mediate endothelial cell growth and integrity, including angiopoietin 2 and vascular endothelial growth factor [Citation33]. The activated endothelium also creates a pro-coagulant stage that perpetuates endothelial barrier breakdown [Citation272,Citation304]. Platelets are activated, becoming capable of killing IEs by release of Platelet Factor 4 [Citation305], and additionally contribute to IE sequestration [Citation136,Citation306] and endothelial apoptosis [Citation307].

In summary, multiple factors contribute to endothelial cell activation, breakdown, and impaired blood–brain barrier integrity, and there is strong evidence for a role of sequestration and cytoadherence, particularly of dual EPCR-ICAM-1 binding IE. Further studies are needed to understand the relative importance of these factors and the mechanistic links to clinical outcomes of cerebral malaria, including coma, severe brain swelling that leads to death, and long-term neurological sequelae.

Severe malarial anaemia

Severe anaemia, defined as haemoglobin <7 g/dL in adults and <5 g/dL in children, is a common cause of death in Africa [Citation1], particularly in malnourished children [Citation308] and malaria is a significant risk factor for severe anaemia, particularly in children under five years of age [Citation309]. According to a large household survey conducted in 16 malaria endemic African countries, 79% of children under five years of age with malaria had anaemia (haemoglobin <12 g/dL) and 8% had severe anaemia [Citation1]. Parasitaemia in severe anaemia is generally low, and the loss of IE alone is not sufficient to account for the extremely low haemoglobin, although severe malaria anaemia is associated with parasitaemia [Citation310]. Anaemia has been associated with destruction of large numbers of non-IEs [Citation311] and evidence suggests this may be attributed to increased erythrocyte rigidity leading to increased splenic clearance [Citation312].

Several studies have suggested a key role of systemic inflammation in the pathogenesis of severe malaria anaemia. The release of parasite factors during IE rupture can induce complement deposition on nearby non-IEs leading to the latter’s phagocytic clearance [Citation313]. Additionally, complement regulatory proteins are reduced on the erythrocyte surface in severe malaria anaemia, most notably on uninfected cells, whereas IEs have higher levels of complement regulatory proteins that may be protective [Citation314].

IEs and secreted parasite factors may contribute to severe anaemia by the disruption of erythropoietic processes. In vitro, IEs directly alter transcription of globin mRNA [Citation315]. Secretion of parasite factor haemozoin may inhibit erythropoiesis directly [Citation316] or indirectly via factors released from macrophages following phagocytosis in the bone marrow [Citation317]. Additionally, systemic inflammatory factors [Citation269,Citation318] and pro-inflammatory cytokines have been associated with SMA [Citation319,Citation320] and may upset erythrocyte homoeostasis to reduce erythropoiesis [Citation321].

Delayed haemolytic anaemia following artemisinin treatment was first observed following infection in non-immune travellers [Citation322] and is thought to be due to the reduced lifespan of erythrocytes that are rapidly pitted by the spleen following artemisinin treatment [Citation322]. A significant drop in haemoglobin following treatment has been observed in up to 5% of patients [Citation323,Citation324] and can be severe [Citation325] but there is debate as to whether this is specifically associated with artemisinin treatment [Citation323,Citation326,Citation327]. Other studies in Africa have found no evidence of delayed haemolysis [Citation328] or rare incidence of post treatment anaemia [Citation329]. These discrepancies may be partially explained by differences in exposure. Post-artemisinin anaemia appears to be more common in non-immune travellers [Citation322] compared to individuals living in endemic settings [Citation327] and a recent report found it was more common in low transmission settings (Asia) compared to high transmission settings (Africa) [Citation310].

Acute kidney injury (AKI)

The WHO defines Acute Kidney Injury (AKI, renal impairment) as impaired glomerular filtration, based on serum creatine of >3 mg/dL or blood urea >20 mM [Citation330]. However, the Kidney Disease: Improving Global Outcomes (KDIGO) Acute Kidney Injury working group recommends using serum creatinine of <0.3 mg/dL in 28 h or urine volume below 0.5 mL/kg/h. Under the KDIGO definition, AKI is a manifestation of severe P. falciparum malaria in both adults [Citation331] and children [Citation332], including in conjunction with severe malarial anaemia and cerebral malaria [Citation332]. AKI has also been reported in P. vivax [Citation331,Citation333], P. knowlesi [Citation333] and occasionally P. malariae [Citation334]. AKI is associated with an increased duration of hospitalization and mortality [Citation332,Citation335] and potentially an increased risk of chronic kidney disease, particularly in children with cerebral malaria.

Multiple molecular mechanisms of AKI in malaria have been proposed. Renal ischaemia can directly cause tubular damage or necrosis, and it may be brought about by excessive fluid loss or impaired glomerular blood flow due to parasite obstruction. Histologically, IEs (P. falciparum, P. vivax and P. knowlesi) and haemozoin have been observed in the glomerular capillaries of some individuals with AKI [Citation333], although at autopsy there is less sequestration in the kidneys than in the brains of the same patients [Citation336]. Parasite risk factors may include high parasite biomass [Citation332], release of cell free haem from IE that directly damages renal tubules and initiates infiltrates of monocytes and neutrophils [Citation336–338], and generation of auto anti-DNA antibodies [Citation339]. Parasite sequestration, reduced renal blood flow and systemic and local inflammation can contribute to glomerular endothelial cell activation [Citation335], which perpetuates inflammation and endothelial cell damage. In mice (P. berghei ANKA) treatment with Angiopoietin 2 or Angiopoietin 2 receptor prevented pro-inflammatory cytokine production and mouse AKI [Citation340]. Understanding the mechanisms of AKI may lead to development of better therapeutics.

Malaria in pregnancy

Malaria in pregnancy causes adverse health outcomes for both mother and baby. It is strongly associated with an increased risk of maternal anaemia and P. falciparum at any point in pregnancy is associated with low birthweight (<2,500 g) due to foetal growth restriction or preterm delivery [Citation8]. Approximately 34% of pregnancies are exposed to malaria in Sub-Saharan Africa resulting in approximately 800,000 low birthweight deliveries each year [Citation1]. P. vivax malaria in pregnancy is less well studied but has also been associated with preterm birth and low birthweight [Citation341,Citation342]. Both P. falciparum and P. vivax malaria in the first trimester are associated with increased risk of miscarriage, and malaria during pregnancy is associated with increased risk of in utero still birth [Citation343,Citation344].

P. falciparum placental malaria occurs when IE cytoadhere in the intervillous space of the placenta via VAR2CSA [Citation114]. There are no reliable biomarkers of placental malaria before delivery, so it is diagnosed by histological examination of the delivered placenta. Placental malaria is characterized by the presence of IEs (active infection) or haemozoin without IEs (past infection) in the placenta intervillous space [Citation345]. Placental malaria is associated with low birthweight, pre-term delivery and maternal anaemia [Citation346]. Placental infiltrates of IEs are sometimes reported following P. vivax in pregnancy but seem to be uncommon [Citation347,Citation348]. However, evidence of placental damage is present in both P. falciparum and P. vivax during pregnancy, including syncytial knotting, fibrin deposits, increased placental barrier thickness, and presence of mononuclear cells [Citation347,Citation349–352].

There are multiple proposed mechanisms by which parasites in peripheral or placental blood may contribute to poor birth outcomes and inflammation is thought to be a major contributor. P. falciparum infection can lead to placental intervillous infiltrates of monocytes [Citation353], T cells [Citation354], and polymorphonuclear cells [Citation355]. If intense, these cell accumulations are termed intervillositis, and monocytes and macrophages are the dominant cell types. Intervillositis is more closely associated with poor outcomes than parasites alone [Citation353,Citation356], particularly low birthweight [Citation356–358]. The release of circulating and localized inflammatory cytokines and complement factors has been associated with poor pregnancy outcomes in P. falciparum [Citation359,Citation360], although other studies have found a protective association for some cytokines [Citation361]. In P. vivax malaria in pregnancy, cytokines are also skewed towards a proinflammatory response although further studies are needed to determine the associations with poor pregnancy outcomes [Citation362,Citation363]. Placental angiogenesis and vascularization are disrupted in placental malaria [Citation356,Citation360], possibly due to inflammation and increased asymmetric dimethylarginine that impairs the nitric oxide synthesis pathway [Citation364]. A clearer understanding of these mechanisms may lead to development of adjunctive therapies to reduce the impacts of malaria in pregnancy.

Therapeutic intervention

Treatment of malaria is continuously challenged by the emergence of drug resistant parasites. Delayed parasite clearance with artemisinins, the frontline anti-malarial drugs, has emerged in South East Asia [Citation365] and has been linked to several mutations in the propeller region of P. falciparum Kelch13 protein [Citation366]. To curb the spread of these mutations, malaria is treated with artemisinin derivatives in combination with a partner drug such as amodiaquine, mefloquine, piperaquine, sulphadoxine-pyrimethamine (SP) or lumefantrine (to clear any remaining parasites), known as Artemisinin Combination Therapy (ACT) [Citation4]. ACT partner drug resistance to SP, piperaquine, lumefantrine and mefloquine has been identified [Citation367–370] and likely contributes to delayed parasite clearance following ACT in South East Asia. In Africa, ACT remains highly effective for treatment of uncomplicated malaria [Citation371–374] and Kelch13 mutations are rare [Citation375]. However, genetic diversification of Kelch13 has been detected in Rwanda [Citation376] and high survival of rings and mutations associated with partner drug resistance have been reported in Uganda [Citation377]. Reduced efficacy of artemisinin would be particularly devastating in Africa due to high rates of severe malaria, which is treated with rectal or intravenous artesunate, to rapidly kill ring-stage parasites, followed by full course of ACT [Citation4]; therefore, continued active surveillance of mutations associated with reduced artemisinin efficacy and partner drug resistance is necessary. Local emergence of drug resistance is likely driven by drug pressure (overuse) or incomplete dosing regimes (underuse), and resistance is more likely to spread in low transmission settings [Citation378]. In Africa and South East Asia, substandard antimalarials [Citation379] and subtherapeutic dosing have been shown to promote growth of drug resistant parasites in vitro [Citation380].

Cerebral malaria is treated with parenteral artesunate [Citation1] and by management of associated morbidities, such as hypoglycaemia and convulsions, and few adjunctive therapies exist. Fluid bolus resuscitation was shown to increase mortality compared to use of maintenance fluids alone [Citation381]. Similarly, routine treatment of seizures in children with cerebral malaria with the anticonvulsant drug, phenobarbitone, was associated with increased mortality in a clinical trial meta-analysis [Citation382], although a recent small study suggested levetiracetam may be a safer alternative [Citation383], and management of brain swelling with mannitol was associated with increased length of coma [Citation384]. A deeper understanding of the molecular mechanisms of cerebral malaria may lead to more effective adjunctive therapies.

In areas where it is chloroquine sensitive, P. vivax can be treated with chloroquine or an ACT. Where chloroquine resistance occurs, an ACT is recommended [Citation385]. In both cases, follow-up treatment with primaquine is necessary to eliminate P. vivax and P. ovale hypnozoites and prevent relapse [Citation4]. Single dose primaquine is recommended following P. falciparum to reduce gametocyte burden and transmission [Citation386]. Individuals with Glucose 6 Phosphate Dehydrogenase deficiency (G6PDd) are susceptible to haemolytic anaemia when treated with primaquine but haemolysis is stopped when treatment is stopped. G6PDd is an X-linked genetic condition caused by a large variety of mutations and reaches high prevalence in malaria endemic populations as it has a protective effect against malaria [Citation387]. The recently approved single-dose alternative to primaquine, tafenoquine [Citation388], also induces G6PDd dependent haemolysis but, unlike primaquine, only requires a single dose, and thus treatment cannot be stopped to limit haemolysis. Pretreatment G6PD screening is recommended but is not widely available. Current RDTs detecting G6PDd may not detect heterozygous females who may also be vulnerable to haemolysis [Citation389,Citation390] and such tests are not always widely available [Citation391].

Current and future research, barriers to effective treatment, and models of disease

We face ongoing challenges to the improved control, elimination and eventual eradication of malaria. Improved diagnostics are needed, both to circumvent the problem of HRP2 mutant parasites that evade commonly used P. falciparum RDTs, and to improve detection of low-density infections. In pre-elimination settings, serology holds promise, especially for detection of recent exposures to P. vivax. Better, more widely available point of care G6PD testing is critical to the widespread implementation of primaquine or tafenoquine to eradicate P. vivax hypnozoites ().

Extending the implementation of proven effective preventives and improving vaccine efficacy are critical. The roll out of RTS,S vaccine to small children in Africa can potentially prevent thousands of deaths each year. If the efficacy of the R21 vaccine is confirmed in definitive trials, this could be a step-change in malaria prevention. Combining sporozoite proteins with gametocyte antigens has the potential to decrease infection and transmission and make inroads into the otherwise unchanging burden of malaria deaths. Similar vaccines are urgently needed for other species, most notably P. vivax.

Figure 3. Challenges in Plasmodium vivax. Vivax malaria presents a unique set of challenges for elimination. 1. Genetic mutations: CYP2D6 polymorphisms decrease effectiveness of radical cure, G6PD deficiency predisposes to 2. Drug-induced haemolysis. 3. Hypnozoite reactivation in the liver causes most new episodes of P. vivax malaria. 4. Reservoirs of hidden infection include the spleen (illustrated) and bone marrow. 5. P. vivax forms gametocytes early in infection, increasing transmissibility. 6. Asymptomatic carriers of vivax whose infection may be submicroscopic (figure in red) are common. 7. Low-density infections may not be detected by RDTs. 8. There are no imminent prospects for a P. vivax vaccine. 9. Robust in vitro culture systems are yet to be developed.

Figure 3. Challenges in Plasmodium vivax. Vivax malaria presents a unique set of challenges for elimination. 1. Genetic mutations: CYP2D6 polymorphisms decrease effectiveness of radical cure, G6PD deficiency predisposes to 2. Drug-induced haemolysis. 3. Hypnozoite reactivation in the liver causes most new episodes of P. vivax malaria. 4. Reservoirs of hidden infection include the spleen (illustrated) and bone marrow. 5. P. vivax forms gametocytes early in infection, increasing transmissibility. 6. Asymptomatic carriers of vivax whose infection may be submicroscopic (figure in red) are common. 7. Low-density infections may not be detected by RDTs. 8. There are no imminent prospects for a P. vivax vaccine. 9. Robust in vitro culture systems are yet to be developed.

At the same time, more widespread implementation of seasonal malaria chemoprevention (possibly combined with seasonal vaccination) could protect up to three times as many children from severe malaria and risk of dying each year. Increasing coverage with intermittent preventive treatment in pregnancy and determining the most effective drug combination will decrease the burden of placental malaria and associated low birthweight and neonatal and infant mortality.

New drugs and smarter ways of administering existing drugs will be key to combating the rise of drug resistance. ACT with 3 drugs offers hope in the short term, while new classes of agents are developed. Controlled human malaria infections are a powerful tool to speed up the development of new drugs, vaccines and alternative therapies such as monoclonal antibodies, which could provide sustained protection against infection [Citation392].

Models of sequestration using organoids [Citation294] or engineered microcapillaries will provide new insights into the mechanisms underlying sequestration [Citation393] and may reveal novel targets for intervention to block sequestration or to prevent severe disease. These may include members of the PfEMP1 family of proteins if these prove functionally conserved and are confirmed to be major targets of anti-disease immunity. VAR2CSA, the placental malaria associated PfEMP1, is a first example. The murine experimental malaria model lacks PfEMP1, but can give insights into pathogenic processes at the cellular and molecular level. The Aotus nancymaae monkey model of placental malaria caused by P. falciparum, however, does recapitulate many of the features of human placental malaria [Citation394].

A major difficulty with P. vivax elimination has been the lack of robust experimental challenge and in vitro culture systems. The liver humanized mouse model shows significant promise as a tool to study liver stage infection and development of hypnozoites [Citation395], and a P. vivax CHMI has been developed [Citation396]. More recently, bone marrow humanized mice that produce human CD71+ human red blood cells have been shown to maintain long-lasting P. vivax cultures [Citation397]. Other challenges are summarized in a recent series of reviews [Citation398]. Meanwhile the impact of P. knowlesi in risk groups in South East Asia remains a significant concern [Citation399].

The lack of progress in reducing global malaria deaths in recent years highlights the problems we face in controlling this disease. While better deployment of existing tools can make a significant impact we still lack the powerful tools, such as highly effective vaccines, that could significantly accelerate progress towards eradication.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

ISW is supported by a University of Melbourne Graduate Research Scholarship. Work in SJR’s laboratory is supported by the Australian Centre for Research Excellence in Malaria Elimination, National Health and Medical Research Council of Australia (APP1143946; APP1134989), the Bill & Melinda Gates Foundation (OPP159078) and the National Institute of Health (1 R01 AI165595-01A1).

References

  • World Health Organization. Word Malaria Report 2021. 2021. Available from: https://www.who.int/publications/i/item/9789240040496
  • Kojom Foko LP, Arya A, Sharma A, et al. Epidemiology and clinical outcomes of severe Plasmodium vivax malaria in India. J Infect. 2021;82(6):231–34.
  • Grigg MJ, William T, Barber BE, et al. Age-Related Clinical Spectrum of Plasmodium knowlesi Malaria and Predictors of Severity. Clin Infect Dis. 2018;67(3):350–359. DOI:10.1093/cid/ciy065
  • World Health Organization. WHO Guidelines for Malaria. 2021. Available from: https://www.who.int/publications/i/item/guidelines-for-malaria
  • Paton RS, Kamau A, Akech S, et al. Malaria infection and severe disease risks in Africa. Science. 2021;373(6557):926–931. https://doi.org/10.1126/science.abj0089
  • Kamau A, Paton RS, Akech S, et al. Malaria hospitalisation in East Africa: age, phenotype and transmission intensity. BMC Med. 2022;20(1):28. DOI:10.1186/s12916-021-02224-w
  • Kapesa A, Kweka EJ, Atieli H, et al. The current malaria morbidity and mortality in different transmission settings in Western Kenya. PLoS One. 2018;13(8):e0202031. https://doi.org/10.1371/journal.pone.0202031
  • Rogerson SJ, Desai M, Mayor A, et al. Burden, pathology, and costs of malaria in pregnancy: new developments for an old problem. Lancet Infect Dis. 2018;18(4):e107–118. DOI:10.1016/S1473-3099(18)30066-5
  • Tatem AJ, Gething PW, Smith DL, et al. Urbanization and the global malaria recession. Malar J. 2013;12(1):133.
  • Doumbe-Belisse P, Kopya E, Ngadjeu CS, et al. Urban malaria in sub-Saharan Africa: dynamic of the vectorial system and the entomological inoculation rate. Malar J. 2021;20(1):364. DOI:10.1186/s12936-021-03891-z
  • De Silva PM, Marshall JM. Factors contributing to urban malaria transmission in sub-saharan Africa: a systematic review. J Trop Med. 2012;2012:819563.
  • Multini LC, AL da S DS, Marrelli MT, et al. The influence of anthropogenic habitat fragmentation on the genetic structure and diversity of the malaria vector Anopheles cruzii (Diptera: culicidae). Sci Rep. 2020;10(1):18018.
  • Balkew M, Mumba P, Yohannes G, et al. An update on the distribution, bionomics, and insecticide susceptibility of Anopheles stephensi in Ethiopia, 2018–2020. Malar J. 2021;20(1):263. DOI:10.1186/s12936-021-03801-3
  • Antonio-Nkondjio C, Fossog BT, Ndo C, et al. Anopheles gambiae distribution and insecticide resistance in the cities of Douala and Yaoundé (Cameroon): influence of urban agriculture and pollution. Malar J. 2011;10(1):154. DOI:10.1186/1475-2875-10-154
  • Thomas S, Ravishankaran S, Justin NAJA, et al. Resting and feeding preferences of Anopheles stephensi in an urban setting, perennial for malaria. Malar J. 2017;16(1):111. DOI:10.1186/s12936-017-1764-5
  • Sinka ME, Pironon S, Massey NC, et al. A new malaria vector in Africa: predicting the expansion range of Anopheles stephensi and identifying the urban populations at risk. Proc Natl Acad Sci U S A. 2020;117(40):24900–24908. DOI:10.1073/pnas.2003976117
  • Worges M, Whitehurst N, Saye R, et al. Performance Outcomes from Africa-Based Malaria Diagnostic Competency Assessment Courses. Am J Trop Med Hyg. 2019;100(4):851–860. DOI:10.4269/ajtmh.18-0361
  • Kamaliddin C, Sutherland CJ, Houze S, et al. The Role of Ultrasensitive Molecular Methods for Detecting Malaria—The Broader Perspective. Clin Infect Dis. 2021;73(6):e1387–1390. DOI:10.1093/cid/ciab221
  • Britton S, Cheng Q, McCarthy JS. Novel molecular diagnostic tools for malaria elimination: a review of options from the point of view of high-throughput and applicability in resource limited settings. Malar J. 2016;15(1):88.
  • Zelman BW, Baral R, Zarlinda I, et al. Costs and cost-effectiveness of malaria reactive case detection using loop-mediated isothermal amplification compared to microscopy in the low transmission setting of Aceh Province, Indonesia. Malar J. 2018;17(1):220. DOI:10.1186/s12936-018-2361-y
  • Cunningham J, Jones S, Gatton ML, et al. A review of the WHO malaria rapid diagnostic test product testing programme (2008–2018): performance, procurement and policy. Malar J. 2019;18(1):387. DOI:10.1186/s12936-019-3028-z
  • Jejaw Zeleke A, Hailu A, Bayih AG, et al. Plasmodium falciparum histidine-rich protein 2 and 3 genes deletion in global settings (2010–2021): a systematic review and meta-analysis. Malar J. 2022;21(1):26. DOI:10.1186/s12936-022-04051-7
  • Gatton ML, Chaudhry A, Glenn J, et al. Impact of Plasmodium falciparum gene deletions on malaria rapid diagnostic test performance. Malar J. 2020;19(1):392. DOI:10.1186/s12936-020-03460-w
  • Amoah LE, Abankwa J, Oppong A. Plasmodium falciparum histidine rich protein-2 diversity and the implications for PfHRP 2: based malaria rapid diagnostic tests in Ghana. Malar J. 2016;15(1):101.
  • Mihreteab S, Anderson K, Pasay C, et al. Epidemiology of mutant Plasmodium falciparum parasites lacking histidine-rich protein 2/3 genes in Eritrea 2 years after switching from HRP2-based RDTs. Sci Rep. 2021;11(1):21082. DOI:10.1038/s41598-021-00714-8
  • Watson OJ, Slater HC, Verity R, et al. Modelling the drivers of the spread of Plasmodium falciparum hrp2 gene deletions in sub-Saharan Africa. Elife. 2017;6:e25008. DOI:10.7554/eLife.25008
  • Laveran A. A new parasite found in the blood of malarial patients. Parasitic origin of malarial attacks. Bull Mem Soc Med Hosp Paris. 1880;17:158–164.
  • Mota MM, Hafalla JCR, Rodriguez A. Migration through host cells activates Plasmodium sporozoites for infection. Nat Med. 2002;8(11):1318–1322.
  • Yahata K, Hart MN, Davies H, et al. Gliding motility of Plasmodium merozoites. Proc Natl Acad Sci. 2021;118(48):e2114442118. DOI:10.1073/pnas.2114442118
  • Cowman AF, Berry D, Baum J. The cellular and molecular basis for malaria parasite invasion of the human red blood cell. J Cell Biol. 2012;198(6):961–971.
  • Warncke JD, Beck H-P. Host Cytoskeleton Remodeling throughout the Blood Stages of Plasmodium falciparum. Microbiol Mol Biol Rev. 2019;83(4):e00013–19.
  • Pologe LG, Pavlovec A, Shio H, et al. Primary structure and subcellular localization of the knob-associated histidine-rich protein of Plasmodium falciparum. Proc Natl Acad Sci U S A. 1987;84(20):7139–7143.
  • Jensen AR, Adams Y, Hviid L. Cerebral Plasmodium falciparum malaria: the role of PfEMP1 in its pathogenesis and immunity, and PfEMP1-based vaccines to prevent it. Immunol Rev. 2020;293(1):230–252.
  • Francis SE, Sullivan DJ, Goldberg DE. HEMOGLOBIN METABOLISM in the MALARIA PARASITE Plasmodium FALCIPARUM. Annu Rev Microbiol. 1997;51(1):97–123.
  • Beri D, Balan B, Tatu U. Commit, hide and escape: the story of Plasmodium gametocytes. Parasitology. 2018;145(13):1772–1782.
  • Brancucci NMB, Gerdt JP, Wang C, et al. Lysophosphatidylcholine Regulates Sexual Stage Differentiation in the Human Malaria Parasite Plasmodium falciparum. Cell. 2017;171(7):1532–1544.e15. DOI:10.1016/j.cell.2017.10.020
  • Sinden RE. The cell biology of malaria infection of mosquito: advances and opportunities. Cell Microbiol. 2015;17(4):451–466.
  • Leopold SJ, Watson JA, Jeeyapant A, et al. Investigating causal pathways in severe falciparum malaria: a pooled retrospective analysis of clinical studies. PLoS Med. 2019;16(8):e1002858. DOI:10.1371/journal.pmed.1002858
  • Uyoga S, Wanjiku P, Rop JC, et al. Plasma Plasmodium falciparum Histidine-rich Protein 2 Concentrations in Children with Malaria Infections of Differing Severity in Kilifi, Kenya. Clin Infect Dis. 2021;73(7):e2415–2423. DOI:10.1093/cid/ciaa1141
  • Fox LL, Taylor TE, Pensulo P, et al. Histidine-rich protein 2 plasma levels predict progression to cerebral malaria in Malawian children with Plasmodium falciparum infection. J Infect Dis. 2013;208(3):500–503. DOI:10.1093/infdis/jit176
  • Park GS, Opoka RO, Shabani E, et al. Plasmodium falciparum Histidine-Rich Protein-2 Plasma Concentrations are Higher in Retinopathy-Negative Cerebral Malaria Than in Severe Malarial Anemia. Open Forum Infect Dis. 2017;4(3):ofx151. DOI:10.1093/ofid/ofx151
  • Manning L, Laman M, Stanisic D, et al. Plasma Plasmodium falciparum histidine-rich protein-2 concentrations do not reflect severity of malaria in Papua new Guinean children. Clin Infect Dis. 2011;52(4):440–446. DOI:10.1093/cid/ciq105
  • Silva-Filho JL, Dos-Santos JC, Judice C, et al. Total parasite biomass but not peripheral parasitaemia is associated with endothelial and haematological perturbations in Plasmodium vivax patients. Elife. 2021;10:e71351. DOI:10.7554/eLife.71351
  • Dollat M, Talla C, Sokhna C, et al. Measuring malaria morbidity in an area of seasonal transmission: pyrogenic parasitemia thresholds based on a 20-year follow-up study. PLoS One. 2019;14(6):e0217903. https://doi.org/10.1371/journal.pone.0217903
  • Hartley M-A, Hofmann N, Keitel K, et al. Clinical relevance of low-density Plasmodium falciparum parasitemia in untreated febrile children: a cohort study. PLoS Med. 2020;17(9):e1003318. DOI:10.1371/journal.pmed.1003318
  • Nguyen TN, von Seidlein L, Nguyen TV, et al. The persistence and oscillations of submicroscopic Plasmodium falciparum and Plasmodium vivax infections over time in Vietnam: an open cohort study. Lancet Infect Dis. 2018;18(5):565–572. DOI:10.1016/S1473-3099(18)30046-X
  • Katrak S, Nayebare P, Rek J, et al. Clinical consequences of submicroscopic malaria parasitaemia in Uganda. Malar J. 2018;17(1):67. DOI:10.1186/s12936-018-2221-9
  • Chen I, Clarke SE, Gosling R, et al. “Asymptomatic” Malaria: a Chronic and Debilitating Infection That Should Be Treated. PLoS Med. 2016;13(1):e1001942. DOI:10.1371/journal.pmed.1001942
  • Slater HC, Ross A, Felger I, et al. The temporal dynamics and infectiousness of subpatent Plasmodium falciparum infections in relation to parasite density. Nat Commun. 2019;10(1):1433. DOI:10.1038/s41467-019-09441-1
  • Rovira-Vallbona E, Moncunill G, Bassat Q, et al. Low antibodies against Plasmodium falciparum and imbalanced pro-inflammatory cytokines are associated with severe malaria in Mozambican children: a case–control study. Malar J. 2012;11(1):181. DOI:10.1186/1475-2875-11-181
  • Kariuki SN, Williams TN. Human genetics and malaria resistance. Hum Genet. 2020;139(6–7):801–811.
  • Goheen MM, Campino S, Cerami C. The role of the red blood cell in host defence against falciparum malaria: an expanding repertoire of evolutionary alterations. Br J Haematol. 2017;179(4):543–556.
  • Band G, Leffler EM, Jallow M, et al. Malaria protection due to sickle haemoglobin depends on parasite genotype. Nature. 2022;602(7895):106–111. https://doi.org/10.1038/s41586-021-04288-3
  • Ndila CM, Uyoga S, Macharia AW, et al. Human candidate gene polymorphisms and risk of severe malaria in children in Kilifi, Kenya: a case-control association study. Lancet Haematol. 2018;5(8):e333–345. DOI:10.1016/S2352-3026(18)30107-8
  • Timmann C, Thye T, Vens M, et al. Genome-wide association study indicates two novel resistance loci for severe malaria. Nature. 2012;489(7416):443–446. https://doi.org/10.1038/nature11334
  • Leffler EM, Band G, Busby GBJ, et al. Resistance to malaria through structural variation of red blood cell invasion receptors. Science. 2017;356(6343):eaam6393. DOI:10.1126/science.aam6393
  • Kariuki SN, Marin-Menendez A, Introini V, et al. Red blood cell tension protects against severe malaria in the Dantu blood group. Nature. 2020;585(7826):579–583. https://doi.org/10.1038/s41586-020-2726-6
  • Petersen JEV, Saelens JW, Freedman E, et al. Sickle-trait hemoglobin reduces adhesion to both CD36 and EPCR by Plasmodium falciparum-infected erythrocytes. PLoS Pathog. 2021;17(6):e1009659. DOI:10.1371/journal.ppat.1009659
  • Fairhurst RM, Baruch DI, Brittain NJ, et al. Abnormal display of PfEMP-1 on erythrocytes carrying haemoglobin C may protect against malaria. Nature. 2005;435(7045):1117–1121. https://doi.org/10.1038/nature03631
  • Nguetse CN, Purington N, Ebel ER, et al. A common polymorphism in the mechanosensitive ion channel PIEZO1 is associated with protection from severe malaria in humans. Proc Natl Acad Sci U S A. 2020;117(16):9074–9081. DOI:10.1073/pnas.1919843117
  • Cholera R, Brittain NJ, Gillrie MR, et al. Impaired cytoadherence of Plasmodium falciparum-infected erythrocytes containing sickle hemoglobin. Proc Natl Acad Sci U S A. 2008;105(3):991–996. DOI:10.1073/pnas.0711401105
  • McQuaid F, Rowe JA. Rosetting revisited: a critical look at the evidence for host erythrocyte receptors in Plasmodium falciparum rosetting. Parasitology. 2020;147(1):1–11.
  • Milner DA, Whitten RO, Kamiza S, et al. The systemic pathology of cerebral malaria in African children. Front Cell Infect Microbiol. 2014;4:1–13. DOI:10.3389/fcimb.2014.00104
  • Bachmann A, Esser C, Petter M, et al. Absence of erythrocyte sequestration and lack of multicopy gene family expression in Plasmodium falciparum from a splenectomized malaria patient. PLoS One. 2009;4(10):e7459. https://doi.org/10.1371/journal.pone.0007459
  • Mahamar A, Attaher O, Swihart B, et al. Host factors that modify Plasmodium falciparum adhesion to endothelial receptors. Sci Rep. 2017;7(1):13872. DOI:10.1038/s41598-017-14351-7
  • Langreth SG, Peterson E. Pathogenicity, stability, and immunogenicity of a knobless clone of Plasmodium falciparum in Colombian owl monkeys. Infect Immun. 1985;47(3):760–766.
  • Stanisic DI, Gerrard J, Fink J, et al. Infectivity of Plasmodium falciparum in Malaria-Naive Individuals is Related to Knob Expression and Cytoadherence of the Parasite. Infect Immun. 2016;84(9):2689–2696. DOI:10.1128/IAI.00414-16
  • Webster R, Sekuloski S, Odedra A, et al. Safety, infectivity and immunogenicity of a genetically attenuated blood-stage malaria vaccine. BMC Med. 2021;19(1):293. DOI:10.1186/s12916-021-02150-x
  • Arakawa C, Gunnarsson C, Howard C, et al. Biophysical and biomolecular interactions of malaria-infected erythrocytes in engineered human capillaries. Sci Adv. 2020;6(3):eaay7243. DOI:10.1126/sciadv.aay7243
  • Joice R, Nilsson SK, Montgomery J, et al. Plasmodium falciparum transmission stages accumulate in the human bone marrow. Sci Transl Med. 2014;6(244):244re5. DOI:10.1126/scitranslmed.3008882
  • De Niz M, Meibalan E, Mejia P, et al. Plasmodium gametocytes display homing and vascular transmigration in the host bone marrow. Sci Adv. 2018;4(5):eaat3775. DOI:10.1126/sciadv.aat3775
  • Rogers NJ, Hall BS, Obiero J, et al. A model for sequestration of the transmission stages of Plasmodium falciparum: adhesion of gametocyte-infected erythrocytes to human bone marrow cells. Infect Immun. 2000;68(6):3455–3462. DOI:10.1128/IAI.68.6.3455-3462.2000
  • Silvestrini F, Tibúrcio M, Bertuccini L, et al. Differential Adhesive Properties of Sequestered Asexual and Sexual Stages of Plasmodium falciparum on Human Endothelial Cells are Tissue Independent. PLoS One. 2012;7(2):e31567.
  • Messina V, Valtieri M, Rubio M, et al. Gametocytes of the Malaria Parasite Plasmodium falciparum Interact with and Stimulate Bone Marrow Mesenchymal Cells to Secrete Angiogenetic Factors. Front Cell Infect Microbiol. 2018;8:50. DOI:10.3389/fcimb.2018.00050
  • Camarda G, Jirawatcharadech P, Priestley RS, et al. Antimalarial activity of primaquine operates via a two-step biochemical relay. Nat Commun. 2019;10(1):3226. DOI:10.1038/s41467-019-11239-0
  • Carvalho BO, Lopes SCP, Nogueira PA, et al. On the Cytoadhesion of Plasmodium vivax –Infected Erythrocytes. J Infect Dis. 2010;202(4):638–647. DOI:10.1086/654815
  • Lacerda MVG, Fragoso SCP, Alecrim MGC, et al. Postmortem characterization of patients with clinical diagnosis of Plasmodium vivax malaria: to what extent does this parasite kill? Clin Infect Dis. 2012;55(8):e67–74. DOI:10.1093/cid/cis615
  • Obaldia IN, Meibalan E, JM S, et al. Bone Marrow is a Major Parasite Reservoir in Plasmodium vivax Infection. Infection MBio. 2018;9(3): e00625-18. doi:10.1128/mBio.00625-18.
  • Baro B, Deroost K, Raiol T, et al. Plasmodium vivax gametocytes in the bone marrow of an acute malaria patient and changes in the erythroid miRNA profile. PLoS Negl Trop Dis. 2017;11(4):e0005365. DOI:10.1371/journal.pntd.0005365
  • Kho S, Qotrunnada L, Leonardo L, et al. Hidden Biomass of Intact Malaria Parasites in the Human Spleen. N Engl J Med. 2021;384(21):2067–2069. DOI:10.1056/NEJMc2023884
  • Pongponratn E, Riganti M, Harinasuta T, et al. Electron microscopic study of phagocytosis in human spleen in falciparum malaria. Southeast Asian J Trop Med Public Health. 1989 20(1):31–39.
  • Ho M, Bannister LH, Looareesuwan S, et al. Cytoadherence and ultrastructure of Plasmodium falciparum-infected erythrocytes from a splenectomized patient. Infect Immun. 1992;60(6):2225–2228.
  • Safeukui I, Correas J-M, Brousse V, et al. Retention of Plasmodium falciparum ring-infected erythrocytes in the slow, open microcirculation of the human spleen. Blood. 2008;112(6):2520–2528. https://doi.org/10.1182/blood-2008-03-146779
  • Elizalde-Torrent A, Trejo-Soto C, Méndez-Mora L, et al. Pitting of malaria parasites in microfluidic devices mimicking spleen interendothelial slits. Sci Rep. 2021;11(1):22099. DOI:10.1038/s41598-021-01568-w
  • David PH, Hommel M, Miller LH, et al. Parasite sequestration in Plasmodium falciparum malaria: spleen and antibody modulation of cytoadherence of infected erythrocytes. Proc Natl Acad Sci U S A. 1983;80(16):5075–5079. DOI:10.1073/pnas.80.16.5075
  • Fernandez-Becerra C, Bernabeu M, Castellanos A, et al. Plasmodium vivax spleen-dependent genes encode antigens associated with cytoadhesion and clinical protection. Proc Natl Acad Sci U S A. 2020;117(23):13056–13065. DOI:10.1073/pnas.1920596117
  • Doumbo OK, Thera MA, Koné AK, et al. High levels of Plasmodium falciparum rosetting in all clinical forms of severe malaria in African children. Am J Trop Med Hyg. 2009;81(6):987–993. DOI:10.4269/ajtmh.2009.09-0406
  • Rowe A, Obeiro J, Newbold CI, et al. Plasmodium falciparum rosetting is associated with malaria severity in Kenya. Infect Immun. 1995;63(6):2323–2326.
  • Carlson J, Helmby H, Hill AV, et al. Human cerebral malaria: association with erythrocyte rosetting and lack of anti-rosetting antibodies. Lancet. 1990;336(8729):1457–1460. DOI:10.1016/0140-6736(90)93174-n
  • Goel S, Palmkvist M, Moll K, et al. RIFINs are adhesins implicated in severe Plasmodium falciparum malaria. Nat Med. 2015;21(4):314–317. DOI:10.1038/nm.3812
  • Jötten AM, Moll K, Wahlgren M, et al. Blood group and size dependent stability of P. falciparum infected red blood cell aggregates in capillaries. Biomicrofluidics. 2020;14(2):24104. https://doi.org/10.1063/1.5125038
  • Hedberg P, Sirel M, Moll K, et al. Red blood cell blood group a antigen level affects the ability of heparin and PfEMP1 antibodies to disrupt Plasmodium falciparum rosettes. Malar J. 2021;20(1):441. DOI:10.1186/s12936-021-03975-w
  • Stevenson L, Huda P, Jeppesen A, et al. Investigating the function of Fc-specific binding of IgM to Plasmodium falciparum erythrocyte membrane protein 1 mediating erythrocyte rosetting. Cell Microbiol. 2015;17(6):819–831. DOI:10.1111/cmi.12403
  • Stevenson L, Laursen E, Cowan GJ, et al. α2-Macroglobulin Can Crosslink Multiple Plasmodium falciparum Erythrocyte Membrane Protein 1 (PfEMP1) Molecules and May Facilitate Adhesion of Parasitized Erythrocytes. PLoS Pathog. 2015;11(7):e1005022. DOI:10.1371/journal.ppat.1005022
  • Al-Yaman F, Genton B, Mokela D, et al. Human cerebral malaria: lack of significant association between erythrocyte rosetting and disease severity. Trans R Soc Trop Med Hyg. 1995;89(1):55–58. DOI:10.1016/0035-9203(95)90658-4
  • Degarege A, Gebrezgi MT, Ibanez G, et al. Effect of the ABO blood group on susceptibility to severe malaria: a systematic review and meta-analysis. Blood Rev. 2019;33:53–62. DOI:10.1016/j.blre.2018.07.002
  • Wang CW, Hviid L. Rifins, rosetting, and red blood cells. Trends Parasitol. 2015;31(7):285–286.
  • Moll K, Palmkvist M, Ch’Ng J, et al. Evasion of Immunity to Plasmodium falciparum: rosettes of Blood Group a Impair Recognition of PfEMP1. PLoS One. 2015;10(12):e0145120. https://doi.org/10.1371/journal.pone.0145120
  • Albrecht L, Lopes SCP, da Silva ABIE, et al. Rosettes integrity protects Plasmodium vivax of being phagocytized. Sci Rep. 2020;10(1):16706. DOI:10.1038/s41598-020-73713-w
  • Lee W-C, Russell B, Lee B, et al. Plasmodium falciparum rosetting protects schizonts against artemisinin. EBioMedicine. 2021;73:103680. DOI:10.1016/j.ebiom.2021.103680
  • Lowe BS, Mosobo M, Bull PC. All four species of human malaria parasites form rosettes. Trans R Soc Trop Med Hyg. 1998;92(5):526.
  • Zhang R, Lee W-C, Lau Y-L, et al. Rheopathologic Consequence of Plasmodium vivax Rosette Formation. PLoS Negl Trop Dis. 2016;10(8):e0004912. DOI:10.1371/journal.pntd.0004912
  • Marín-Menéndez A, Bardají A, Martínez-Espinosa FE, et al. Rosetting in Plasmodium vivax: a cytoadhesion phenotype associated with anaemia. PLoS Negl Trop Dis. 2013;7(4):e2155. DOI:10.1371/journal.pntd.0002155
  • Pain A, Ferguson DJ, Kai O, et al. Platelet-mediated clumping of Plasmodium falciparum-infected erythrocytes is a common adhesive phenotype and is associated with severe malaria. Proc Natl Acad Sci U S A. 2001;98(4):1805–1810. DOI:10.1073/pnas.98.4.1805
  • Biswas AK, Hafiz A, Banerjee B, et al. Plasmodium falciparum Uses gC1qr/HABP1/p32 as a Receptor to Bind to Vascular Endothelium and for Platelet-Mediated Clumping. PLoS Pathog. 2007;3(9):e130. DOI:10.1371/journal.ppat.0030130
  • Mayor A, Hafiz A, Bassat Q, et al. Association of severe malaria outcomes with platelet-mediated clumping and adhesion to a novel host receptor. PLoS One. 2011;6(4):e19422. https://doi.org/10.1371/journal.pone.0019422
  • Arman M, Raza A, Tempest LJ, et al. Platelet-Mediated Clumping of Plasmodium falciparum–infected Erythrocytes is Associated with High Parasitemia but Not Severe Clinical Manifestations of Malaria in African Children. Am J Trop Med Hyg. 2007;77(5):943–946. DOI:10.4269/ajtmh.2007.77.943
  • Arman M, Rowe JA. Experimental conditions affect the outcome of Plasmodium falciparum platelet-mediated clumping assays. Malar J. 2008;7(1):243.
  • Chan JA, Fowkes FJI, Beeson JG. Surface antigens of Plasmodium falciparum-infected erythrocytes as immune targets and malaria vaccine candidates. Cell Mol Life Sci. 2014;71(19):3633–3657.
  • Nielsen MA, Staalsoe T, Kurtzhals JAL, et al. Plasmodium falciparum Variant Surface Antigen Expression Varies Between Isolates Causing Severe and Nonsevere Malaria and is Modified by Acquired Immunity. J Immunol. 2002;168(7):3444–3450. DOI:10.4049/jimmunol.168.7.3444
  • Warimwe GM, Keane TM, Fegan G, et al. Plasmodium falciparum var gene expression is modified by host immunity. Proc Natl Acad Sci. 2009;106(51):21801–21806. DOI:10.1073/pnas.0907590106
  • Bull PC, Lowe BS, Kortok M, et al. Parasite antigens on the infected red cell surface are targets for naturally acquired immunity to malaria. Nat Med. 1998;4(3):358–360. DOI:10.1038/nm0398-358
  • Smith JD, Craig AG, Kriek N, et al. Identification of a Plasmodium falciparum intercellular adhesion molecule-1 binding domain: a parasite adhesion trait implicated in cerebral malaria. Proc Natl Acad Sci U S A. 2000;97(4):1766–1771. DOI:10.1073/pnas.040545897
  • Salanti A, Staalsoe T, Lavstsen T, et al. Selective upregulation of a single distinctly structured var gene in chondroitin sulphate A-adhering Plasmodium falciparum involved in pregnancy-associated malaria. Mol Microbiol. 2003;49(1):179–191. DOI:10.1046/j.1365-2958.2003.03570.x
  • Roberts DJ, Craig AG, Berendt AR, et al. Rapid switching to multiple antigenic and adhesive phenotypes in malaria. Nature. 1992;357(6380):689–692. DOI:10.1038/357689a0
  • Bernabeu M, Lopez FJ, Ferrer M, et al. Functional analysis of Plasmodium vivax VIR proteins reveals different subcellular localizations and cytoadherence to the ICAM-1 endothelial receptor. Cell Microbiol. 2012;14(3):386–400. DOI:10.1111/j.1462-5822.2011.01726.x
  • Requena P, Rui E, Padilla N, et al. Plasmodium vivax VIR Proteins are Targets of Naturally-Acquired Antibody and T Cell Immune Responses to Malaria in Pregnant Women. PLoS Negl Trop Dis. 2016;10(10):e0005009. DOI:10.1371/journal.pntd.0005009
  • Rask TS, Hansen DA, Theander TG, et al. Plasmodium falciparum Erythrocyte Membrane Protein 1 Diversity in Seven Genomes – Divide and Conquer. PLoS Comput Biol. 2010;6(9):e1000933. DOI:10.1371/journal.pcbi.1000933
  • Jensen ATR, Theander TG, Nielsen MA, et al. Plasmodium falciparum Associated with Severe Childhood Malaria Preferentially Expresses PfEMP1 Encoded by Group a var Genes. J Exp Med. 2004;199(9):1179–1190. DOI:10.1084/jem.20040274
  • Rottmann M, Muller D, Jensen ATR, et al. Differential Expression of var Gene Groups is Associated with Morbidity Caused by Plasmodium falciparum Infection in Tanzanian Children. Infect Immun. 2006;74(7):3904–3911. DOI:10.1128/iai.02073-05
  • Bertin GI, Alao MJ, Ndam NT, et al. Expression of the Domain Cassette 8 Plasmodium falciparum Erythrocyte Membrane Protein 1 is Associated with Cerebral Malaria in Benin. PLoS One. 2013;8(7):e68368. DOI:10.1371/journal.pone.0068368
  • Kyriacou HM, Doumbo OK, Challis RJ, et al. Differential var gene transcription in Plasmodium falciparum isolates from patients with cerebral malaria compared to hyperparasitaemia. Mol Biochem Parasitol. 2006;150(2):211–218. DOI:10.1016/j.molbiopara.2006.08.005
  • Jespersen JS, Wang CW, Mkumbaye SI, et al. Plasmodium falciparum var genes expressed in children with severe malaria encode CIDRα1 domains. EMBO Mol Med. 2016;8(8):839–850. DOI:10.15252/emmm.201606188
  • Lennartz F, Adams Y, Bengtsson A, et al. Structure-Guided Identification of a Family of Dual Receptor-Binding PfEMP1 that is Associated with Cerebral Malaria. Cell Host Microbe. 2017;21(3):403–414. DOI:10.1016/j.chom.2017.02.009
  • Kaestli M, Cockburn IA, Cortés A, et al. Virulence of malaria is associated with differential expression of Plasmodium falciparum var gene subgroups in a case-control study. J Infect Dis. 2006;193(11):1567–1574. DOI:10.1086/503776
  • AV O, Morrison R, Amos E, et al. A Plasma Survey Using 38 PfEMP1 Domains Reveals Frequent Recognition of the Plasmodium falciparum Antigen VAR2CSA among Young Tanzanian Children. PLoS One. 2012;7(1):e31011. DOI:10.1371/journal.pone.0031011
  • Smith JD, Subramanian G, Gamain B, et al. Classification of adhesive domains in the Plasmodium falciparum Erythrocyte Membrane Protein 1 family. Mol Biochem Parasitol. 2000;110(2):293–310. DOI:10.1016/S0166-6851(00)00279-6
  • Janes JH, Wang CP, Levin-Edens E, et al. Investigating the host binding signature on the Plasmodium falciparum PfEMP1 protein family. PLoS Pathog. 2011;7(5):e1002032. DOI:10.1371/journal.ppat.1002032
  • Robinson BA, Welch TL, Smith JD. Widespread functional specialization of Plasmodium falciparum erythrocyte membrane protein 1 family members to bind CD36 analysed across a parasite genome. Mol Microbiol. 2003;47(5):1265–1278.
  • Mkumbaye SI, Wang CW, Lyimo E, et al. The severity of Plasmodium falciparum infection is associated with transcript levels of var genes encoding endothelial protein C receptor-binding P. falciparum erythrocyte membrane protein 1. Infect Immun. 2017;85(4):1–14. DOI:10.1128/IAI.00841-16
  • Rogerson SJ, Plitt S, Taylor TE, et al. Cytoadherence characteristics of Plasmodium falciparum-infected erythrocytes from Malawian children with severe and uncomplicated malaria. Am J Trop Med Hyg. 1999;61(3):467–472. DOI:10.4269/ajtmh.1999.61.467
  • Tuikue Ndam N, Moussiliou A, Lavstsen T, et al. Parasites Causing Cerebral Falciparum Malaria Bind Multiple Endothelial Receptors and Express EPCR and ICAM-1-Binding PfEMP1. J Infect Dis. 2017;215(12):1918–1925. DOI:10.1093/infdis/jix230
  • Wichers JS, Tonkin-Hill G, Thye T, et al. Common virulence gene expression in adult first-time infected malaria patients and severe cases. Elife. 2021;10:e69040. DOI:10.7554/eLife.69040
  • Turner GDH, Morrison H, Jones M, et al. An immunohistochemical study of the pathology of fatal malaria. Evidence for widespread endothelial activation and a potential role for intercellular adhesion molecule-1 in cerebral sequestration. Am J Pathol. 1994;145(5):1057–1069.
  • Avril M, Bernabeu M, Benjamin M, et al. Interaction between endothelial protein C receptor and intercellular adhesion molecule 1 to mediate binding of Plasmodium falciparum-infected erythrocytes to endothelial cells. MBio. 2016;7(4):1–10. https://doi.org/10.1128/mBio.00615-16
  • Wassmer SC, Lépolard C, Traoré B, et al. Platelets Reorient Plasmodium falciparum –Infected Erythrocyte Cytoadhesion to Activated Endothelial Cells. J Infect Dis. 2004;189(2):180–189. DOI:10.1086/380761
  • Turner L, Lavstsen T, Berger SS, et al. Severe malaria is associated with parasite binding to endothelial protein C receptor. Nature. 2013;498(7455):502–505. https://doi.org/10.1038/nature12216
  • Avril M, Tripathi AK, Brazier AJ, et al. A restricted subset of var genes mediates adherence of Plasmodium falciparum-infected erythrocytes to brain endothelial cells. Proc Natl Acad Sci. 2012;109(26):e1782–90. DOI:10.1073/pnas.1120534109
  • Joste V, Guillochon E, Fraering J, et al. PfEMP1 A-Type ICAM-1-Binding Domains are Not Associated with Cerebral Malaria in Beninese Children. MBio. 2020;11(6):e02103–20. https://doi.org/10.1128/mBio.02103-20
  • Kessler A, Dankwa S, Bernabeu M, et al. Linking EPCR-Binding PfEMP1 to Brain Swelling in Pediatric Cerebral Malaria. Cell Host Microbe. 2017;22(5):601–614.e5. DOI:10.1016/j.chom.2017.09.009
  • Storm J, Jespersen JS, Seydel KB, et al. Cerebral malaria is associated with differential cytoadherence to brain endothelial cells. EMBO Mol Med. 2019;11(2):1–15. DOI:10.15252/emmm.201809164
  • Shabani E, Hanisch B, Opoka RO, et al. Plasmodium falciparum EPCR-binding PfEMP1 expression increases with malaria disease severity and is elevated in retinopathy negative cerebral malaria. BMC Med. 2017;15(1):183. DOI:10.1186/s12916-017-0945-y
  • Lavstsen T, Gilbert MTP, Willerslev E, et al. Plasmodium falciparum erythrocyte membrane protein 1 domain cassettes 8 and 13 are associated with severe malaria in children. Proc Natl Acad Sci. 2012;109(26):e1791–1800. DOI:10.1073/pnas.1120455109
  • Bengtsson A, Joergensen L, Rask TS, et al. A Novel Domain Cassette Identifies Plasmodium falciparum PfEMP1 Proteins Binding ICAM-1 and is a Target of Cross-Reactive, Adhesion-Inhibitory Antibodies. J Immunol. 2013;190(1):240–249. DOI:10.4049/jimmunol.1202578
  • Olsen RW, Ecklu-Mensah G, Bengtsson A, et al. Natural and Vaccine-Induced Acquisition of Cross-Reactive IgG-Inhibiting ICAM-1-Specific Binding of a Plasmodium falciparum PfEMP1 Subtype Associated Specifically with Cerebral Malaria. Infect Immun. 2018;86(4):1–17. DOI:10.1128/IAI.00622-17
  • Tonkin-Hill GQ, Trianty L, Noviyanti R, et al. The Plasmodium falciparum transcriptome in severe malaria reveals altered expression of genes involved in important processes including surface antigen–encoding var genes. PLoS Biol. 2018;16(3):e2004328. DOI:10.1371/journal.pbio.2004328
  • Duffy MF, Caragounis A, Noviyanti R, et al. Transcribed var Genes Associated with Placental Malaria in Malawian Women. Infect Immun. 2006;74(8):4875–4883. DOI:10.1128/IAI.01978-05
  • Tuikue Ndam NG, Salanti A, Bertin G, et al. High Level of var2csa Transcription by Plasmodium falciparum Isolated from the Placenta. J Infect Dis. 2005;192(2):331–335. DOI:10.1086/430933
  • Clausen TM, Christoffersen S, Dahlbäck M, et al. Structural and functional insight into how the Plasmodium falciparum VAR2CSA protein mediates binding to chondroitin sulfate a in placental malaria. J Biol Chem. 2012;287(28):23332–23345. DOI:10.1074/jbc.M112.348839
  • Doolan DL, Dobaño C, Baird JK. Acquired immunity to malaria. Clin Microbiol Rev. 2009;22(1):13–36.
  • Griffin JT, Déirdre Hollingsworth T, Reyburn H, et al. Gradual acquisition of immunity to severe malaria with increasing exposure. Proc R Soc B Biol Sci. 2015;282(1801):20142657. DOI:10.1098/rspb.2014.2657
  • Gupta S, Snow RW, Donnelly CA, et al. Immunity to non-cerebral severe malaria is acquired after one or two infections. Nat Med. 1999;5(3):340–343. DOI:10.1038/6560
  • Fowkes FJI, Boeuf P, Bees JG. Immunity to malaria in an era of declining malaria transmission. Parasitology. 2016;143(2):139–153.
  • Cohen S, McGregor IA, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature. 1961;192(4804):733–737.
  • Kanoi BN, Nagaoka H, Morita M, et al. Comprehensive analysis of antibody responses to Plasmodium falciparum erythrocyte membrane protein 1 domains. Vaccine. 2018;36(45):6826–6833. https://doi.org/10.1016/j.vaccine.2018.08.058
  • Smith TG, Serghides L, Patel SN, et al. CD36-mediated nonopsonic phagocytosis of erythrocytes infected with stage I and IIA gametocytes of Plasmodium falciparum. Infect Immun. 2003;71(1):393–400. DOI:10.1128/IAI.71.1.393-400.2003
  • Saeed M, Roeffen W, Alexander N, et al. Plasmodium falciparum Antigens on the Surface of the Gametocyte-Infected Erythrocyte. PLoS One. 2008;3(5):e2280. DOI:10.1371/journal.pone.0002280
  • Chan JA, Drew DR, Reiling L, et al. Low levels of human antibodies to gametocyte-infected erythrocytes contrasts the PfEMP1-dominant response to asexual stages in P. falciparum malaria. Front Immunol. 2019;10:1–8. DOI:10.3389/fimmu.2018.03126
  • Drakeley CJ, Bousema JT, Akim NIJ, et al. Transmission-reducing immunity is inversely related to age in Plasmodium falciparum gametocyte carriers. Parasite Immunol. 2006;28(5):185–190. DOI:10.1111/j.1365-3024.2005.00818.x
  • Stone WJR, Campo JJ, Ouédraogo AL, et al. Unravelling the immune signature of Plasmodium falciparum transmission-reducing immunity. Nat Commun. 2018;9(1):558. DOI:10.1038/s41467-017-02646-2
  • Mendis KN, Munesinghe YD, de Silva YN, et al. Malaria transmission-blocking immunity induced by natural infections of Plasmodium vivax in humans. Infect Immun. 1987;55(2):369–372. DOI:10.1128/iai.55.2.369-372.1987
  • Acquah FK, Adjah J, Williamson KC, et al. Transmission-Blocking Vaccines: old Friends and New Prospects. Infect Immun. 2019;87(6). e00775-18. DOI:10.1128/IAI.00775-18.
  • RM DJ, Meerstein-Kessel L, Da DF, et al. Monoclonal antibodies block transmission of genetically diverse Plasmodium falciparum strains to mosquitoes. NPJ Vaccines. 2021;6(1):101. DOI:10.1038/s41541-021-00366-9
  • Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol. 2018;19(11):1199–1211.
  • Ishizuka AS, Lyke KE, DeZure A, et al. Protection against malaria at 1 year and immune correlates following PfSPZ vaccination. Nat Med. 2016;22(6):614–623. DOI:10.1038/nm.4110
  • Cockburn IA, Amino R, Kelemen RK, et al. In vivo imaging of CD8+ T cell-mediated elimination of malaria liver stages. Proc Natl Acad Sci U S A. 2013;110(22):9090–9095. DOI:10.1073/pnas.1303858110
  • Feng G, Wines BD, Kurtovic L, et al. Mechanisms and targets of Fcγ-receptor mediated immunity to malaria sporozoites. Nat Commun. 2021;12(1):1742. DOI:10.1038/s41467-021-21998-4
  • Bousema T, Barry A, Behet MC, et al. Functional antibodies against Plasmodium falciparum sporozoites are associated with a longer time to qPCR-detected infection among schoolchildren in Burkina Faso. Wellcome Open Res. 2019;3:1–40. DOI:10.12688/wellcomeopenres.14932.2
  • Mishra S, Nussenzweig RS, Nussenzweig V. Antibodies to Plasmodium circumsporozoite protein (CSP) inhibit sporozoite’s cell traversal activity. J Immunol Methods. 2012;377(1–2):47–52.
  • Longley JR, Reyes-Sandoval A, Eduardo M-D, et al. Acquisition and Longevity of Antibodies to Plasmodium vivax Preerythrocytic Antigens in Western Thailand. Clin Vaccine Immunol. 2016;23(2):117–124. DOI:10.1128/CVI.00501-15
  • Daou M, Kouriba B, Ouédraogo N, et al. Protection of Malian children from clinical malaria is associated with recognition of multiple antigens. Malar J. 2015;14(1):56. DOI:10.1186/s12936-015-0567-9
  • Crompton PD, Kayala MA, Traore B, et al. A prospective analysis of the Ab response to Plasmodium falciparum before and after a malaria season by protein microarray. Proc Natl Acad Sci U S A. 2010;107(15):6958–6963. DOI:10.1073/pnas.1001323107
  • Offeddu V, Olotu A, Osier F, et al. High Sporozoite Antibody Titers in Conjunction with Microscopically Detectable Blood Infection Display Signatures of Protection from Clinical Malaria. Front Immunol. 2017;8:488. DOI:10.3389/fimmu.2017.00488
  • Adepoju P. RTS,S malaria vaccine pilots in three African countries. Lancet. 2019;393(10182):1685.
  • Kurtovic L, Atre T, Feng G, et al. Multifunctional Antibodies are Induced by the RTS,S Malaria Vaccine and Associated with Protection in a Phase 1/2a Trial. J Infect Dis. 2021;224(7):1128–1138. DOI:10.1093/infdis/jiaa144
  • RTS, S Clinical Trials Partnership GBS. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet. 2015;386(9988):31–45. doi: 10.1016/S0140-6736(15)60721-8.
  • Moon JE, Greenleaf ME, Regules JA, et al. A phase IIA extension study evaluating the effect of booster vaccination with a fractional dose of RTS,S/AS01(E) in a controlled human malaria infection challenge. Vaccine. 2021;39(43):6398–6406. DOI:10.1016/j.vaccine.2021.09.024
  • Datoo MS, Natama MH, Somé A, et al. Efficacy of a low-dose candidate malaria vaccine, R21 in adjuvant Matrix-M, with seasonal administration to children in Burkina Faso: a randomised controlled trial. Lancet. 2021;397(10287):1809–1818. DOI:10.1016/S0140-6736(21)00943-0
  • Chandramohan D, Zongo I, Sagara I, et al. Seasonal Malaria Vaccination with or without Seasonal Malaria Chemoprevention. N Engl J Med. 2021;385(11):1005–1017. DOI:10.1056/nejmoa2026330
  • Jongo SA, Urbano V, Church LWP, et al. Immunogenicity and Protective Efficacy of Radiation-Attenuated and Chemo-Attenuated PfSPZ Vaccines in Equatoguinean Adults. Am J Trop Med Hyg. 2021;104(1):283–293. DOI:10.4269/ajtmh.20-0435
  • Walk J, Reuling IJ, Behet MC, et al. Modest heterologous protection after Plasmodium falciparum sporozoite immunization: a double-blind randomized controlled clinical trial. BMC Med. 2017;15(1):168. DOI:10.1186/s12916-017-0923-4
  • Sulyok Z, Fendel R, Eder B, et al. Heterologous protection against malaria by a simple chemoattenuated PfSPZ vaccine regimen in a randomized trial. Nat Commun. 2021;12(1):2518. DOI:10.1038/s41467-021-22740-w
  • Murphy SC, Deye GA, Sim BKL, et al. PfSPZ-CVac efficacy against malaria increases from 0% to 75% when administered in the absence of erythrocyte stage parasitemia: a randomized, placebo-controlled trial with controlled human malaria infection. PLoS Pathog. 2021;17(5):e1009594. DOI:10.1371/journal.ppat.1009594
  • Oneko M, Steinhardt LC, Yego R, et al. Safety, immunogenicity and efficacy of PfSPZ Vaccine against malaria in infants in western Kenya: a double-blind, randomized, placebo-controlled phase 2 trial. Nat Med. 2021;27(9):1636–1645. DOI:10.1038/s41591-021-01470-y
  • Bucşan AN, Williamson KC. Setting the stage: the initial immune response to blood-stage parasites. Virulence. 2020;11(1):88–103.
  • Rathnayake D, Aitken EH, Rogerson SJ. Beyond Binding: the Outcomes of Antibody-Dependent Complement Activation in Human Malaria. Front Immunol. 2021;12:683404.
  • Paul G, Deshmukh A, Kumar Chourasia B, et al. Protein–protein interaction studies reveal the Plasmodium falciparum merozoite surface protein-1 region involved in a complex formation that binds to human erythrocytes. Biochem J. 2018;475(6):1197–1209. DOI:10.1042/BCJ20180017
  • Chen L, Xu Y, Wong W, et al. Structural basis for inhibition of erythrocyte invasion by antibodies to Plasmodium falciparum protein CyRPA. Elife. 2017;6:e21347. DOI:10.7554/eLife.21347
  • Feng G, Boyle MJ, Cross N, et al. Human Immunization with a Polymorphic Malaria Vaccine Candidate Induced Antibodies to Conserved Epitopes That Promote Functional Antibodies to Multiple Parasite Strains. J Infect Dis. 2018;218(1):35–43. DOI:10.1093/infdis/jiy170
  • Garcia-Senosiain A, Kana IH, Singh S, et al. Neutrophils dominate in opsonic phagocytosis of P. falciparum blood-stage merozoites and protect against febrile malaria. Commun Biol. 2021;4:984. DOI:10.1038/s42003-021-02511-5
  • Boyle MJ, Reiling L, Feng G, et al. Human antibodies fix complement to inhibit Plasmodium falciparum invasion of erythrocytes and are associated with protection against malaria. Immunity. 2015;42(3):580–590. https://doi.org/10.1016/j.immuni.2015.02.012
  • Cowman AF, Crabb BS. Invasion of red blood cells by malaria parasites. Cell. 2006;124(4):755–766.
  • Aniweh Y, Nyarko PB, Charles-Chess E, et al. Plasmodium falciparum Merozoite Associated Armadillo Protein (PfMAAP) is Apically Localized in Free Merozoites and Antibodies are Associated with Reduced Risk of Malaria. Front Immunol. 2020;11:505. DOI:10.3389/fimmu.2020.00505
  • Michelow IC, Park S, Tsai S-W, et al. A newly characterized malaria antigen on erythrocyte and merozoite surfaces induces parasite inhibitory antibodies. J Exp Med. 2021;218(9):e20200170. DOI:10.1084/jem.20200170
  • Hamre KES, Ondigo BN, Hodges JS, et al. Antibody Correlates of Protection from Clinical Plasmodium falciparum Malaria in an Area of Low and Unstable Malaria Transmission. Am J Trop Med Hyg. 2020;103(6):2174–2182. DOI:10.4269/ajtmh.18-0805
  • Sakamoto H, Takeo S, Takashima E, et al. Identification of target proteins of clinical immunity to Plasmodium falciparum in a region of low malaria transmission. Parasitol Int. 2018;67(2):203–208. DOI:10.1016/j.parint.2017.12.002
  • Weaver R, Reiling L, Feng G, et al. The association between naturally acquired IgG subclass specific antibodies to the PfRH5 invasion complex and protection from Plasmodium falciparum malaria. Sci Rep. 2016;6(1):1–10. DOI:10.1038/srep33094
  • Richards JS, Stanisic DI, Fowkes FJI, et al. Association between naturally acquired antibodies to erythrocyte-binding antigens of Plasmodium falciparum and protection from malaria and high-density parasitemia. Clin Infect Dis. 2010;51(8):e50–60. DOI:10.1086/656413
  • Kana IH, Garcia-Senosiain A, Singh SK, 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(6):956–965. DOI:10.1093/infdis/jiy258
  • Kana IH, Singh SK, Garcia-Senosiain A, et al. Breadth of Functional Antibodies is Associated with Plasmodium falciparum Merozoite Phagocytosis and Protection Against Febrile Malaria. J Infect Dis. 2019;220(2):275–284. DOI:10.1093/infdis/jiz088
  • Duncan CJA, Sheehy SH, Ewer KJ, 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(7):e22271. DOI:10.1371/journal.pone.0022271
  • Minassian AM, Silk SE, Barrett JR, et al. Reduced blood-stage malaria growth and immune correlates in humans following RH5 vaccination. Med. 2021;2(6):701–719.e19. https://doi.org/10.1016/j.medj.2021.03.014
  • Dassah S, Adu B, Sirima SB, et al. Extended follow-up of children in a phase 2b trial of the GMZ2 malaria vaccine. Vaccine. 2021;39(31):4314–4319. https://doi.org/10.1016/j.vaccine.2021.06.024
  • Nash SD, Prevots DR, Kabyemela E, et al. A Malaria-Resistant Phenotype with Immunological Correlates in a Tanzanian Birth Cohort Exposed to Intense Malaria Transmission. Am J Trop Med Hyg. 2017;96(5):1190–1196. DOI:10.4269/ajtmh.16-0554
  • Chan JA, Stanisic DI, Duffy MF, et al. Patterns of protective associations differ for antibodies to P. falciparum-infected erythrocytes and merozoites in immunity against malaria in children. Eur J Immunol. 2017;47(12):2124–2136. DOI:10.1002/eji.201747032
  • McCallum FJ, Persson KEM, Fowkes FJI, et al. Differing rates of antibody acquisition to merozoite antigens in malaria: implications for immunity and surveillance. J Leukoc Biol. 2017;101(4):913–925. DOI:10.1189/jlb.5MA0716-294R
  • Adamou R, Dechavanne C, Sadissou I, et al. Plasmodium falciparum merozoite surface antigen-specific cytophilic IgG and control of malaria infection in a Beninese birth cohort. Malar J. 2019;18(1):194. DOI:10.1186/s12936-019-2831-x
  • Yman V, Tuju J, White MT, et al. Distinct kinetics of antibodies to 111 Plasmodium falciparum proteins identifies markers of recent malaria exposure. Nat Commun. 2022;13(1):331. DOI:10.1038/s41467-021-27863-8
  • Helb DA, Tetteh KKA, Felgner PL, et al. Novel serologic biomarkers provide accurate estimates of recent Plasmodium falciparum exposure for individuals and communities. Proc Natl Acad Sci. 2015;112(32):e4438–47. DOI:10.1073/pnas.1501705112
  • CV P, Alves JRS, Lima BAS, et al. Blood-stage Plasmodium vivax antibody dynamics in a low transmission setting: a nine year follow-up study in the Amazon region. PLoS One. 2018;13(11):e0207244. DOI:10.1371/journal.pone.0207244
  • Longley RJ, White MT, Takashima E, et al. Development and validation of serological markers for detecting recent Plasmodium vivax infection. Nat Med. 2020;26(5):741–749. DOI:10.1038/s41591-020-0841-4
  • Tayipto Y, Liu Z, Mueller I, et al. Serology for Plasmodium vivax surveillance: a novel approach to accelerate towards elimination. Parasitol Int. 2022;87:102492.
  • Fraser M, Jing W, Bröer S, et al. Breakdown in membrane asymmetry regulation leads to monocyte recognition of P. falciparum-infected red blood cells. PLoS Pathog. 2021;17(2):e1009259. DOI:10.1371/journal.ppat.1009259
  • Giribaldi G, Ulliers D, Mannu F, et al. Growth of Plasmodium falciparum induces stage-dependent haemichrome formation, oxidative aggregation of band 3, membrane deposition of complement and antibodies, and phagocytosis of parasitized erythrocytes. Br J Haematol. 2001;113(2):492–499. DOI:10.1046/j.1365-2141.2001.02707.x
  • Zelter T, Strahilevitz J, Simantov K, et al. Neutrophils impose strong immune pressure against PfEMP1 variants implicated in cerebral malaria. EMBO Rep. 2022:e53641. DOI:10.15252/embr.202153641
  • Gowda NM, Wu X, Kumar S, et al. CD36 Contributes to Malaria Parasite-Induced Pro-Inflammatory Cytokine Production and NK and T Cell Activation by Dendritic Cells. PLoS One. 2013;8(10):e77604. DOI:10.1371/journal.pone.0077604
  • Mavoungou E, Luty AJF, Kremsner PG Natural killer (NK) cell-mediated cytolysis of Plasmodium falciparum-infected human red blood cells in vitro. Eur Cytokine Netw. 2003 14(3):134–142.
  • Baratin M, Roetynck S, Pouvelle B, et al. Dissection of the role of PfEMP1 and ICAM-1 in the sensing of Plasmodium-falciparum-infected erythrocytes by natural killer cells. PLoS One. 2007;2(2):e228. DOI:10.1371/journal.pone.0000228
  • D’Ombrain MC, Voss TS, Maier AG, et al. Plasmodium falciparum Erythrocyte Membrane Protein-1 Specifically Suppresses Early Production of Host Interferon-γ. Cell Host Microbe. 2007;2(2):130–138. DOI:10.1016/j.chom.2007.06.012
  • Saito F, Hirayasu K, Satoh T, et al. Immune evasion of Plasmodium falciparum by RIFIN via inhibitory receptors. Nature. 2017;552(7683):101–105. https://doi.org/10.1038/nature24994
  • Chan J-A, Howell KB, Reiling L, et al. Targets of antibodies against Plasmodium falciparum–infected erythrocytes in malaria immunity. J Clin Invest. 2012;122(9):3227–3238. DOI:10.1172/JCI62182
  • Kanoi BN, Nagaoka H, White MT, et al. Global Repertoire of Human Antibodies Against Plasmodium falciparum RIFINs, SURFINs, and STEVORs in a Malaria Exposed Population. Front Immunol. 2020;11:893. DOI:10.3389/fimmu.2020.00893
  • Travassos MA, Niangaly A, Bailey JA, et al. Children with cerebral malaria or severe malarial anaemia lack immunity to distinct variant surface antigen subsets. Sci Rep. 2018;8(1):6281. DOI:10.1038/s41598-018-24462-4
  • Nilsson Bark SK, Ahmad R, Dantzler K, et al. Quantitative Proteomic Profiling Reveals Novel Plasmodium falciparum Surface Antigens and Possible Vaccine Candidates. Mol Cell Proteomics. 2018;17(1):43–60. DOI:10.1074/mcp.RA117.000076
  • Aitken EH, Damelang T, Ortega-Pajares A, et al. Developing a multivariate prediction model of antibody features associated with protection of malaria-infected pregnant women from placental malaria. Elife. 2021;10:1–30. DOI:10.7554/eLife.65776
  • Arora G, Hart GT, Manzella-Lapeira J, et al. NK cells inhibit Plasmodium falciparum growth in red blood cells via antibody-dependent cellular cytotoxicity. Elife. 2018;7:1–20. DOI:10.7554/eLife.36806
  • Chan J-A, Boyle MJ, Moore KA, et al. Antibody Targets on the Surface of Plasmodium falciparum– Infected Erythrocytes That are Associated with Immunity to Severe Malaria in Young Children. J Infect Dis. 2019;219(5):819–828. DOI:10.1093/infdis/jiy580
  • Aitken EH, Mahanty S, Rogerson SJ. Antibody effector functions in malaria and other parasitic diseases: a few needles and many haystacks. Immunol Cell Biol. 2020;98(4):264–275.
  • Akhouri RR, Goel S, Furusho H, et al. Architecture of Human IgM in Complex with P. falciparum Erythrocyte Membrane Protein 1. Cell Rep. 2016;14(4):723–736. DOI:10.1016/j.celrep.2015.12.067
  • Goodyer ID, Pouvelle B, Schneider TG, et al. Characterization of macromolecular transport pathways in malaria-infected erythrocytes. Mol Biochem Parasitol. 1997;87(1):13–28. https://doi.org/10.1016/S0166-6851(97)00039-X
  • Kurtis JD, Raj DK, Michelow IC, et al. Maternally-derived Antibodies to Schizont Egress Antigen-1 and Protection of Infants from Severe Malaria. Clin Infect Dis. 2019;68(10):1718–1724. DOI:10.1093/cid/ciy728
  • Raj DK, Nixon CP, Nixon CE, et al. Antibodies to PfSEA-1 block parasite egress from RBCs and protect against malaria infection. Science. 2014;344(6186):871–877. https://doi.org/10.1126/science.1254417
  • Perrin AJ, Bisson C, Faull PA, et al. Malaria Parasite Schizont Egress Antigen-1 Plays an Essential Role in Nuclear Segregation during Schizogony. MBio. 2021;12(2): e03377-20. https://doi.org/10.1128/mBio.03377-20.
  • Larsen MD, Del Pilar Quintana M, Ditlev SB, et al. Evasion of classical complement pathway activation on Plasmodium falciparum-infected erythrocytes opsonized by PfEMP1-Specific IgG. Front Immunol. 2019;10:1–10. DOI:10.3389/fimmu.2018.03088
  • Azasi Y, Low LM, Just AN, et al. Complement C1s cleaves PfEMP1 at interdomain conserved sites inhibiting Plasmodium falciparum cytoadherence. Proc Natl Acad Sci U S A. 2021;118(22):1–10. DOI:10.1073/pnas.2104166118
  • Gonçalves BP, Huang CY, Morrison R, et al. Parasite burden and severity of malaria in Tanzanian children. N Engl J Med. 2014;370(19):1799–1808. DOI:10.1056/NEJMoa1303944
  • Obeng-Adjei N, Larremore DB, Turner L, et al. Longitudinal analysis of naturally acquired PfEMP1 CIDR domain variant antibodies identifies associations with malaria protection. JCI Insight. 2020;5(12):e137262. https://doi.org/10.1172/jci.insight.137262
  • Cham GKK, Turner L, Lusingu J, et al. Sequential, Ordered Acquisition of Antibodies to Plasmodium falciparum Erythrocyte Membrane Protein 1 Domains. J Immunol. 2009;183(5):3356–3363. DOI:10.4049/jimmunol.0901331
  • Bachmann A, Bruske E, Krumkamp R, et al. Controlled human malaria infection with Plasmodium falciparum demonstrates impact of naturally acquired immunity on virulence gene expression. PLoS Pathog. 2019;15(7):e1007906. DOI:10.1371/journal.ppat.1007906
  • Ricke CH, Staalsoe T, Koram K, et al. Plasma Antibodies from Malaria-Exposed Pregnant Women Recognize Variant Surface Antigens on Plasmodium falciparum -Infected Erythrocytes in a Parity-Dependent Manner and Block Parasite Adhesion to Chondroitin Sulfate a. J Immunol. 2000;165(6):3309–3316. DOI:10.4049/jimmunol.165.6.3309
  • Cutts JC, Agius PA, Lin Z, et al. Pregnancy-specific malarial immunity and risk of malaria in pregnancy and adverse birth outcomes: a systematic review. BMC Med. 2020;18(1):1–21. DOI:10.1186/s12916-019-1467-6
  • Larsen MD, Lopez-Perez M, Dickson EK, et al. Afucosylated Plasmodium falciparum-specific IgG is induced by infection but not by subunit vaccination. Nat Commun. 2021;12(1):5838. DOI:10.1038/s41467-021-26118-w
  • Mayor A, Kumar U, Bardají A, et al. Improved pregnancy outcomes in women exposed to malaria with high antibody levels against Plasmodium falciparum. J Infect Dis. 2013;207(11):1664–1674. DOI:10.1093/infdis/jit083
  • Staalsoe T, Shulman CE, Bulmer JN, et al. Variant surface antigen-specific IgG and protection against clinical consequences of pregnancy-associated Plasmodium falciparum malaria. Lancet. 2004;363(9405):283–289. DOI:10.1016/S0140-6736(03)15386-X
  • Duffy PE, Fried M. Antibodies that inhibit Plasmodium falciparum adhesion to chondroitin sulfate a are associated with increased birth weight and the gestational age of newborns. Infect Immun. 2003;71(11):6620–6623.
  • Teo A, Hasang W, Randall LM, et al. Decreasing malaria prevalence and its potential consequences for immunity in pregnant women. J Infect Dis. 2014;210(9):1444–1455. DOI:10.1093/infdis/jiu264
  • Fried M, Kurtis JD, Swihart B, et al. Antibody levels to recombinant VAR2CSA domains vary with Plasmodium falciparum parasitaemia, gestational age, and gravidity, but do not predict pregnancy outcomes. Malar J. 2018;17(1):106. DOI:10.1186/s12936-018-2258-9
  • Sirima SB, Richert L, Chêne A, 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(5):585–597. DOI:10.1016/S1473-3099(19)30739-X
  • Mordmüller B, Sulyok M, Egger-Adam D, et al. First-in-human, Randomized, Double-blind Clinical Trial of Differentially Adjuvanted PAMVAC, a Vaccine Candidate to Prevent Pregnancy-associated Malaria. Clin Infect Dis. 2019;69(9):1509–1516. DOI:10.1093/cid/ciy1140
  • Doritchamou JYA, Renn JP, Jenkins B, et al. A single full-length VAR2CSA ectodomain variant purifies broadly neutralizing antibodies against placental malaria isolates. Elife. 2022;11:e76264. DOI:10.7554/eLife.76264
  • Tessema SK, Nakajima R, Jasinskas A, et al. Protective Immunity against Severe Malaria in Children is Associated with a Limited Repertoire of Antibodies to Conserved PfEMP1 Variants. Cell Host & Microbe. 2019;26(5):579–590.e5. https://doi.org/10.1016/j.chom.2019.10.012
  • Rambhatla JS, Davis TME, Warrel J, et al. Acquisition of Antibodies Against Endothelial Protein C Receptor–Binding Domains of Plasmodium falciparum Erythrocyte Membrane Protein 1 in Children with Severe Malaria. J Infect Dis. 2019;219(5):808–818. DOI:10.1093/infdis/jiy564
  • Rambhatla JS, Tonkin-Hill GQ, Takashima E, et al. Identifying Targets of Protective Antibodies against Severe Malaria in Papua, Indonesia, Using Locally Expressed Domains. Infect Immun. 2022;90(2):e0043521. DOI:10.1128/iai.00435-21
  • Tessema SK, Utama D, Chesnokov O, et al. Antibodies to Intercellular Adhesion Molecule 1-Binding Plasmodium falciparum Erythrocyte Membrane Protein 1-DBLβ are Biomarkers of Protective Immunity to Malaria in a Cohort of Young Children from Papua New Guinea. Infect Immun. 2018;86(8):1–14. DOI:10.1128/iai.00485-17
  • Araj BN, Swihart B, Morrison R, et al. Antibody Levels to Plasmodium falciparum Erythrocyte Membrane Protein 1-DBLγ11 and DBLδ-1 Predict Reduction in Parasite Density. mSystems. 2021;6(3):1–10. DOI:10.1128/mSystems.00347-21
  • Angeletti D, Albrecht L, Wahlgren M, et al. Analysis of antibody induction upon immunization with distinct NTS-DBL1α-domains of PfEMP1 from rosetting Plasmodium falciparum parasites. Malar J. 2013;12(1):32.
  • Albrecht L, Angeletti D, Moll K, et al. B-cell epitopes in NTS-DBL1α of PfEMP1 recognized by human antibodies in rosetting Plasmodium falciparum. PLoS One. 2014;9(12):e113248. https://doi.org/10.1371/journal.pone.0113248
  • Kessler A, Campo JJ, Harawa V, et al. Convalescent Plasmodium falciparum-specific seroreactivity does not correlate with paediatric malaria severity or Plasmodium antigen exposure. Malar J. 2018;17(1):178. DOI:10.1186/s12936-018-2323-4
  • Badaut C, Visitdesotrakul P, Chabry A, et al. Author Correction: IgG acquisition against PfEMP1 PF11_0521 domain cassette DC13, DBLβ3_D4 domain, and peptides located within these constructs in children with cerebral malaria. Sci Rep. 2021;11(1):1–12. DOI:10.1038/s41598-021-82444-5
  • Lusingu JPA, Jensen ATR, Vestergaard LS, et al. Levels of plasma immunoglobulin G with specificity against the cysteine-rich interdomain regions of a semiconserved Plasmodium falciparum erythrocyte membrane protein 1, VAR4, predict protection against malarial anemia and febrile episodes. Infect Immun. 2006;74(5):2867–2875. DOI:10.1128/IAI.74.5.2867-2875.2006
  • Ghumra A, Khunrae P, Ataide R, et al. Immunisation with recombinant PfEMP1 domains elicits functional rosette-inhibiting and phagocytosis-inducing antibodies to Plasmodium falciparum. PLoS One. 2011;6(1):e16414. https://doi.org/10.1371/journal.pone.0016414
  • Quintana MDP, Angeletti D, Moll K, et al. Phagocytosis-inducing antibodies to Plasmodium falciparum upon immunization with a recombinant PfEMP1 NTS-DBL1α domain. Malar J. 2016;15(1):1–9. DOI:10.1186/s12936-016-1459-3
  • He X, Xia L, Tumas KC, et al. Type I Interferons and Malaria: a Double-Edge Sword Against a Complex Parasitic Disease. Front Cell Infect Microbiol. 2020;10:594621. DOI:10.3389/fcimb.2020.594621
  • Du Y, Hertoghs N, Duffy FJ, et al. Systems analysis of immune responses to attenuated P. falciparum malaria sporozoite vaccination reveals excessive inflammatory signatures correlating with impaired immunity. PLoS Pathog. 2022;18:e1010282.
  • Dunst J, Kamena F, Matuschewski K. Cytokines and Chemokines in Cerebral Malaria Pathogenesis. Front Cell Infect Microbiol. 2017;7:324.
  • Walk J, Keramati F, de Bree LCJ, et al. Controlled Human Malaria Infection Induces Long-Term Functional Changes in Monocytes. Front Mol Biosci. 2020;7:604553. DOI:10.3389/fmolb.2020.604553
  • Mandala WL, Msefula CL, Gondwe EN, et al. Cytokine Profiles in Malawian Children Presenting with Uncomplicated Malaria, Severe Malarial Anemia, and Cerebral Malaria. Clin Vaccine Immunol. 2017;24(4): e00533-16. doi:10.1128/CVI.00533-16.
  • Royo J, Rahabi M, Kamaliddin C, et al. Changes in monocyte subsets are associated with clinical outcomes in severe malarial anaemia and cerebral malaria. Sci Rep. 2019;9(1):17545. DOI:10.1038/s41598-019-52579-7
  • Achieng AO, Guyah B, Cheng Q, et al. Molecular basis of reduced LAIR1 expression in childhood severe malarial anaemia: implications for leukocyte inhibitory signalling. EBioMedicine. 2019;45:278–289. DOI:10.1016/j.ebiom.2019.06.040
  • Leão L, Puty B, Dolabela MF, et al. Association of cerebral malaria and TNF-α levels: a systematic review. BMC Infect Dis. 2020;20:442. DOI:10.1186/s12879-020-05107-2
  • Raacke M, Kerr A, Dörpinghaus M, et al. Altered Cytokine Response of Human Brain Endothelial Cells after Stimulation with Malaria Patient Plasma. Cells. 2021;10(7):1656. https://doi.org/10.3390/cells10071656
  • Avril M, Benjamin M, Dols M-M, et al. Interplay of Plasmodium falciparum and thrombin in brain endothelial barrier disruption. Sci Rep. 2019;9(1):13142.
  • Harawa V, Njie M, Kessler A, et al. Brain swelling is independent of peripheral plasma cytokine levels in Malawian children with cerebral malaria. Malar J. 2018;17(1):435. DOI:10.1186/s12936-018-2590-0
  • Quanquin NM, Barres LG, Aliyari SR, et al. Gravidity-dependent associations between interferon response and birth weight in placental malaria. Malar J. 2020;19(1):280. DOI:10.1186/s12936-020-03351-0
  • Hountohotegbe T, Gbedande K, Agbota G, et al. Circulating Cytokines Associated with Poor Pregnancy Outcomes in Beninese Exposed to Infection with Plasmodium falciparum. Infect Immun. 2020;88(8). DOI:10.1128/IAI.00042-20.
  • Fried M, Kurtis JD, Swihart B, et al. Systemic Inflammatory Response to Malaria During Pregnancy is Associated with Pregnancy Loss and Preterm Delivery. Clin Infect Dis. 2017;65(10):1729–1735. DOI:10.1093/cid/cix623
  • Okamgba OC, Ifeanyichukwu MO, Ilesanmi AO, et al. Variations in the leukocyte and cytokine profiles between placental and maternal circulation in pregnancy-associated malaria. Res Rep Trop Med. 2018;9:1–8.
  • Taylor TE, Fu WJ, Carr RA, et al. Differentiating the pathologies of cerebral malaria by postmortem parasite counts. Nat Med. 2004;10(2):143–145. DOI:10.1038/nm986
  • Reyburn H, Mbatia R, Drakeley C, et al. Association of Transmission Intensity and Age with Clinical Manifestations and Case Fatality of Severe Plasmodium falciparum Malaria. JAMA. 2005;293(12):1461–1470. DOI:10.1001/jama.293.12.1461
  • NAV B, Taylor TE, Harding SP, et al. Malarial retinopathy: a newly established diagnostic sign in severe malaria. Am J Trop Med Hyg. 2006;75(5):790–797. DOI:10.4269/ajtmh.2006.75.790
  • Villaverde C, Namazzi R, Shabani E, et al. Retinopathy-Positive Cerebral Malaria is Associated with Greater Inflammation, Blood-Brain Barrier Breakdown, and Neuronal Damage Than Retinopathy-Negative Cerebral Malaria. J Pediatric Infect Dis Soc. 2020;9(5):580–586. DOI:10.1093/jpids/piz082
  • Villaverde C, Namazzi R, Shabani E, et al. Clinical Comparison of Retinopathy-Positive and Retinopathy-Negative Cerebral Malaria. Am J Trop Med Hyg. 2017;96:1176–1184. DOI:10.4269/ajtmh.16-0315
  • Sahu PK, Duffy FJ, Dankwa S, et al. Determinants of brain swelling in pediatric and adult cerebral malaria. JCI Insight. 2021;6(18):e145823. https://doi.org/10.1172/jci.insight.145823
  • Seydel KB, Kampondeni SD, Molyneux ME, et al. Brain Swelling and Death in Children with Cerebral Malaria. N Engl J Med. 2015;372(12):1126–1137. DOI:10.1056/nejmoa1400116
  • Dondorp AM, Fanello CI, Hendriksen IC, et al. Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial. Lancet. 2010;376(9753):1647–1657. DOI:10.1016/S0140-6736(10)61924-1
  • Idro R, Kakooza-Mwesige A, Asea B, et al. Cerebral malaria is associated with long-term mental health disorders: a cross sectional survey of a long-term cohort. Malar J. 2016;15(1):184. DOI:10.1186/s12936-016-1233-6
  • Boivin MJ, Mohanty A, Sikorskii A, et al. Early and middle childhood developmental, cognitive, and psychiatric outcomes of Malawian children affected by retinopathy positive cerebral malaria. Child Neuropsychol a J Norm Abnorm Dev Child Adolesc. 2019;25(1):81–102. DOI:10.1080/09297049.2018.1451497
  • Brim R, Mboma S, Semrud-Clikeman M, et al. Cognitive Outcomes and Psychiatric Symptoms of Retinopathy-Positive Cerebral Malaria: cohort Description and Baseline Results. Am J Trop Med Hyg. 2017;97(1):225–231. DOI:10.4269/ajtmh.17-0020
  • Datta D, Bangirana P, Opoka RO, et al. Association of Plasma Tau with Mortality and Long-term Neurocognitive Impairment in Survivors of Pediatric Cerebral Malaria and Severe Malarial Anemia. JAMA Network Open. 2021;4(12):e2138515. DOI:10.1001/jamanetworkopen.2021.38515
  • Cabantous S, Poudiougou B, Bergon A, et al. Understanding Human Cerebral Malaria through a Blood Transcriptomic Signature: evidences for Erythrocyte Alteration, Immune/Inflammatory Dysregulation, and Brain Dysfunction. Mediators Inflamm. 2020;2020:3280689. DOI:10.1155/2020/3280689
  • MacCormick IJC, Barrera V, Beare NAV, et al. How Does Blood-Retinal Barrier Breakdown Relate to Death and Disability in Pediatric Cerebral Malaria? J Infect Dis. 2022;225(6):1070–1080. DOI:10.1093/infdis/jiaa541
  • Ponsford MJ, Medana IM, Prapansilp P, et al. Sequestration and microvascular congestion are associated with coma in human cerebral malaria. J Infect Dis. 2012;205(4):663–671. DOI:10.1093/infdis/jir812
  • Mohan Rao LV, Esmon CT, Pendurthi UR. Endothelial cell protein C receptor: a multiliganded and multifunctional receptor. Blood. 2014;124(10):1553–1562.
  • Adams Y, Olsen RW, Bengtsson A, et al. Plasmodium falciparum erythrocyte membrane protein 1 variants induce cell swelling and disrupt the blood–brain barrier in cerebral malaria. J Exp Med. 2021;218(3):e20201266. DOI:10.1084/JEM.20201266
  • Eugenin EA, Martiney JA, Berman JW. The malaria toxin hemozoin induces apoptosis in human neurons and astrocytes: potential role in the pathogenesis of cerebral malaria. Brain Res. 2019;1720:146317.
  • Pal P, Daniels BP, Oskman A, et al. Plasmodium falciparum Histidine-Rich Protein II Compromises Brain Endothelial Barriers and May Promote Cerebral Malaria Pathogenesis. MBio. 2016;7(3). https://doi.org/10.1128/mBio.00617-16.
  • Moxon CA, Alhamdi Y, Storm J, et al. Parasite histones are toxic to brain endothelium and link blood barrier breakdown and thrombosis in cerebral malaria. Blood Adv. 2020;4(13):2851–2864. DOI:10.1182/bloodadvances.2019001258
  • Silva LS, Pinheiro AS, Teixeira DE, et al. Kinins Released by Erythrocytic Stages of Plasmodium falciparum Enhance Adhesion of Infected Erythrocytes to Endothelial Cells and Increase Blood Brain Barrier Permeability via Activation of Bradykinin Receptors. Front Med. 2019;6:75. DOI:10.3389/fmed.2019.00075
  • MB VH, Palmer A, Onyiorah E, et al. The effect of a monoclonal antibody to tumor necrosis factor on survival from childhood cerebral malaria. J Infect Dis. 1996;174(5):1091–1097. DOI:10.1093/infdis/174.5.1091
  • Bangirana P, Conroy AL, Opoka RO, et al. Inhaled nitric oxide and cognition in pediatric severe malaria: a randomized double-blind placebo controlled trial. PLoS One. 2018;13(1):e0191550. https://doi.org/10.1371/journal.pone.0191550
  • Ghazanfari N, Mueller SN, Heath WR. Cerebral Malaria in Mouse and Man. Front Immunol. 2018;9:2016.
  • Riggle BA, Manglani M, Maric D, et al. CD8+ T cells target cerebrovasculature in children with cerebral malaria. J Clin Invest. 2020;130(3):1128–1138. DOI:10.1172/JCI133474
  • Barrera V, Haley MJ, Strangward P, et al. Comparison of CD8(+) T Cell Accumulation in the Brain During Human and Murine Cerebral Malaria. Front Immunol. 2019;10:1747. DOI:10.3389/fimmu.2019.01747
  • Moxon CA, Wassmer SC, Milner DAJ, et al. Loss of endothelial protein C receptors links coagulation and inflammation to parasite sequestration in cerebral malaria in African children. Blood. 2013;122(5):842–851. https://doi.org/10.1182/blood-2013-03-490219
  • Kho S, Barber BE, Johar E, et al. Platelets kill circulating parasites of all major Plasmodium species in human malaria. Blood. 2018;132(12):1332–1344. https://doi.org/10.1182/blood-2018-05-849307
  • Bridges DJ, Bunn J, van Mourik JA, et al. Rapid activation of endothelial cells enables Plasmodium falciparum adhesion to platelet-decorated von Willebrand factor strings. Blood. 2010;115(7):1472–1474. https://doi.org/10.1182/blood-2009-07-235150
  • Wassmer SC, de Souza JB, Frère C, et al. TGF-β1 Released from Activated Platelets Can Induce TNF-Stimulated Human Brain Endothelium Apoptosis: a New Mechanism for Microvascular Lesion during Cerebral Malaria. J Immunol. 2006;176(2):1180 LP–1184. DOI:10.4049/jimmunol.176.2.1180
  • Nkosi-Gondwe T, Calis J, Boele van Hensbroek M, et al. A cohort analysis of survival and outcomes in severely anaemic children with moderate to severe acute malnutrition in Malawi. PLoS One. 2021;16(2):e0246267. https://doi.org/10.1371/journal.pone.0246267
  • Green HK, Sousa-Figueiredo JC, Basáñez MG, et al. Anaemia in Ugandan preschool-aged children: the relative contribution of intestinal parasites and malaria. Parasitology. 2011;138(12):1534–1545. DOI:10.1017/S0031182011001016
  • The WorldWide Antimalarial Resistance Network Falciparum Haematology Study Group, Mansoor R, Commons RJ, et al. Haematological consequences of acute uncomplicated falciparum malaria: a WorldWide Antimalarial Resistance Network pooled analysis of individual patient data. BMC Med. 2022;20(1):85.
  • Jakeman GN, Saul A, Hogarth WL, et al. Anaemia of acute malaria infections in non-immune patients primarily results from destruction of uninfected erythrocytes. Parasitology. 1999;119(Pt 2):127–133.
  • Dondorp AM, Angus BJ, Chotivanich K, et al. Red blood cell deformability as a predictor of anemia in severe falciparum malaria. Am J Trop Med Hyg. 1999;60(5):733–737. DOI:10.4269/ajtmh.1999.60.733
  • Dasari P, Fries A, Heber SD, et al. Malarial anemia: digestive vacuole of Plasmodium falciparum mediates complement deposition on bystander cells to provoke hemophagocytosis. Med Microbiol Immunol. 2014;203(6):383–393. DOI:10.1007/s00430-014-0347-0
  • Oyong DA, Kenangalem E, Poespoprodjo JR, et al. Loss of complement regulatory proteins on uninfected erythrocytes in vivax and falciparum malaria anemia. JCI Insight. 2018;3(22):e124854. https://doi.org/10.1172/jci.insight.124854
  • Pathak V, Colah R, Ghosh K. Plasmodium falciparum malaria skews globin gene expression balance in in-vitro haematopoietic stem cell culture system: its implications in malaria associated anemia. Exp Parasitol. 2018;185:29–38.
  • Thawani N, Tam M, Bellemare M-J, et al. Plasmodium products contribute to severe malarial anemia by inhibiting erythropoietin-induced proliferation of erythroid precursors. J Infect Dis. 2014;209(1):140–149. DOI:10.1093/infdis/jit417
  • Skorokhod OA, Caione L, Marrocco T, et al. Inhibition of erythropoiesis in malaria anemia: role of hemozoin and hemozoin-generated 4-hydroxynonenal. Blood. 2010;116(20):4328–4337. https://doi.org/10.1182/blood-2010-03-272781
  • Opoka RO, Conroy AL, Waiswa A, et al. Severe Anemia is Associated with Systemic Inflammation in Young Children Presenting to a Tertiary Hospital in Uganda. Am J Trop Med Hyg. 2020;103(6):2574–2580. DOI:10.4269/ajtmh.20-0199
  • Punnath K, Dayanand KK, Chandrashekhar VN, et al. Association between inflammatory cytokine levels and anemia during Plasmodium falciparum and Plasmodium vivax infections in Mangaluru: a Southwestern Coastal Region of India. Trop Parasitol. 2019;9(2):98–107. DOI:10.4103/tp.TP_66_18
  • AMR da S V, Fernandes AAM, Zanini GM, et al. Clinical and immunological profiles of anaemia in children and adolescents with Plasmodium vivax malaria in the Pará state, Brazilian Amazon. Acta Trop. 2018;181:122–131. DOI:10.1016/j.actatropica.2018.01.022
  • Lamikanra AA, Merryweather-Clarke AT, Tipping AJ, et al. Distinct mechanisms of inadequate erythropoiesis induced by tumor necrosis factor alpha or malarial pigment. PLoS One. 2015;10(3):e0119836.
  • Jauréguiberry S, Ndour PA, Roussel C, et al. Postartesunate delayed hemolysis is a predictable event related to the lifesaving effect of artemisinins. Blood. 2014;124(2):167–175. https://doi.org/10.1182/blood-2014-02-555953
  • Fanello C, Onyamboko M, Lee SJ, et al. Post-treatment haemolysis in African children with hyperparasitaemic falciparum malaria; a randomized comparison of artesunate and quinine. BMC Infect Dis. 2017;17(1):575. DOI:10.1186/s12879-017-2678-0
  • Scheu K, Adegnika AA, Addo MM, et al. Determinants of post-malarial anemia in African children treated with parenteral artesunate. Sci Rep. 2019;9(1):18134. DOI:10.1038/s41598-019-54639-4
  • Rehman K, Lötsch F, Kremsner PG, et al. Haemolysis associated with the treatment of malaria with artemisinin derivatives: a systematic review of current evidence. Int J Infect Dis IJID off Publ Int Soc Infect Dis. 2014;29:268–273.
  • Sagara I, Piarroux R, Djimde A, et al. Delayed anemia assessment in patients treated with oral artemisinin derivatives for uncomplicated malaria: a pooled analysis of clinical trials data from Mali. Malar J. 2014;13(1):358. DOI:10.1186/1475-2875-13-358
  • Rolling T, Wichmann D, Schmiedel S, et al. Artesunate versus quinine in the treatment of severe imported malaria: comparative analysis of adverse events focussing on delayed haemolysis. Malar J. 2013;12(1):241. DOI:10.1186/1475-2875-12-241
  • Hawkes MT, Opoka RO, Conroy AL, et al. Anemia and transfusion requirements among Ugandan children with severe malaria treated with intravenous artesunate. Pediatr Hematol Oncol. 2020;37(2):140–152. DOI:10.1080/08880018.2019.1701161
  • Guenther G, Saidi AM, Izem R, et al. Post-Malaria Anemia is Rare in Malawian Children with Cerebral Malaria. Am J Trop Med Hyg. 2021;104(6):2146–2151. DOI:10.4269/ajtmh.20-1668
  • World Health Organization. WHO Severe malaria 2014. Trop Med Int Heal. 2014;19:7–131. DOI:10.1111/tmi.12313_2
  • Anghan H, Sethi P, Soneja M, et al. Clinical and Laboratory Features Associated with Acute Kidney Injury in Severe Malaria. Indian J Crit Care Med. 2018;22(10):718–722. DOI:10.4103/ijccm.IJCCM_468_17
  • Conroy AL, Opoka RO, Bangirana P, et al. Acute kidney injury is associated with impaired cognition and chronic kidney disease in a prospective cohort of children with severe malaria. BMC Med. 2019;17(1):98. DOI:10.1186/s12916-019-1332-7
  • Kaur C, Pramanik A, Kumari K, et al. Renal detection of Plasmodium falciparum, Plasmodium vivax and Plasmodium knowlesi in malaria associated acute kidney injury: a retrospective case–control study. BMC Res Notes. 2020;13(1):37. DOI:10.1186/s13104-020-4900-1
  • Alexandru S, Ortiz Arduan A, López Picasso M, et al. Fracaso renal agudo anúrico persistente en paciente infectado con Plasmodium malariae: la importancia de la biopsia renal. Nefrología. 2020;40(5):571–573. DOI:10.1016/j.nefro.2019.10.002
  • Hawkes MT, Leligdowicz A, Batte A, et al. Pathophysiology of Acute Kidney Injury in Malaria and Non-Malarial Febrile Illness: a Prospective Cohort Study. Pathogens. 2022;11(4):436. https://doi.org/10.3390/pathogens11040436
  • Nguansangiam S, Day NPJ, Hien TT, et al. A quantitative ultrastructural study of renal pathology in fatal Plasmodium falciparum malaria. Trop Med Int Health. 2007;12(9):1037–1050. DOI:10.1111/j.1365-3156.2007.01881.x
  • Katsoulis O, Georgiadou A, Cunnington AJ. Immunopathology of Acute Kidney Injury in Severe Malaria. Front Immunol. 2021;12:651739.
  • Plewes K, Kingston HWF, Ghose A, et al. Cell-free hemoglobin mediated oxidative stress is associated with acute kidney injury and renal replacement therapy in severe falciparum malaria: an observational study. BMC Infect Dis. 2017;17(1):313. DOI:10.1186/s12879-017-2373-1
  • Rivera-Correa J, Conroy AL, Opoka RO, et al. Autoantibody levels are associated with acute kidney injury, anemia and post-discharge morbidity and mortality in Ugandan children with severe malaria. Sci Rep. 2019;9(1):14940. DOI:10.1038/s41598-019-51426-z
  • Silva LS, Peruchetti DB, Silva-Aguiar RP, et al. The angiotensin II/AT1 receptor pathway mediates malaria-induced acute kidney injury. PLoS One. 2018;13(9):e0203836. https://doi.org/10.1371/journal.pone.0203836
  • Bardají A, Martínez-Espinosa FE, Arévalo-Herrera M, et al. Burden and impact of Plasmodium vivax in pregnancy: a multi-centre prospective observational study. PLoS Negl Trop Dis. 2017;11(6):e0005606. DOI:10.1371/journal.pntd.0005606
  • Dombrowski JG, Barateiro A, Peixoto EPM, et al. Adverse pregnancy outcomes are associated with Plasmodium vivax malaria in a prospective cohort of women from the Brazilian Amazon. PLoS Negl Trop Dis. 2021;15(4):e0009390. DOI:10.1371/journal.pntd.0009390
  • McGready R, Lee SJ, Wiladphaingern J, et al. Adverse effects of falciparum and vivax malaria and the safety of antimalarial treatment in early pregnancy: a population-based study. Lancet Infect Dis. 2012;12(5):388–396. DOI:10.1016/S1473-3099(11)70339-5
  • Moore KA, Simpson JA, Scoullar MJL, et al. Quantification of the association between malaria in pregnancy and stillbirth: a systematic review and meta-analysis. Lancet Glob Health. 2017;5(11):e1101–1112. DOI:10.1016/S2214-109X(17)30340-6
  • Bulmer JN, Rasdheed FN, Francis N, et al. Placental malaria. I. Pathological classification. Histopathology. 1993;22(3):211–218. DOI:10.1111/j.1365-2559.1993.tb00110.x
  • Lufele E, Umbers A, Ordi J, et al. Risk factors and pregnancy outcomes associated with placental malaria in a prospective cohort of Papua New Guinean women. Malar J. 2017;16(1):427. DOI:10.1186/s12936-017-2077-4
  • Souza RM, Ataíde R, Dombrowski JG, et al. Placental Histopathological Changes Associated with Plasmodium vivax Infection during Pregnancy. PLoS Negl Trop Dis. 2013;7(2):e2071. DOI:10.1371/journal.pntd.0002071
  • Mayor A, Bardají A, Felger I, et al. Placental infection with Plasmodium vivax: a histopathological and molecular study. J Infect Dis. 2012;206(12):1904–1910. DOI:10.1093/infdis/jis614
  • Ismail MR, Ordi J, Menendez C, et al. Placental pathology in malaria: a histological, immunohistochemical, and quantitative study. Hum Pathol. 2000;31(1):85–93. DOI:10.1016/s0046-8177(00)80203-8
  • Crocker IP, Tanner OM, Myers JE, et al. Syncytiotrophoblast Degradation and the Pathophysiology of the Malaria-infected Placenta. Placenta. 2004;25:273–282. DOI:10.1016/j.placenta.2003.09.010
  • Moeller SL, Nyengaard JR, Larsen LG, et al. Malaria in Early Pregnancy and the Development of the Placental Vasculature. J Infect Dis. 2019;220(9):1425–1434. DOI:10.1093/infdis/jiy735
  • Chaikitgosiyakul S, Rijken MJ, Muehlenbachs A, et al. A morphometric and histological study of placental malaria shows significant changes to villous architecture in both Plasmodium falciparum and Plasmodium vivax infection. Malar J. 2014;13(1):4. DOI:10.1186/1475-2875-13-4
  • Rogerson SJ, Pollina E, Getachew A, et al. Placental monocyte infiltrates in response to Plasmodium falciparum malaria infection and their association with adverse pregnancy outcomes. Am J Trop Med Hyg. 2003;68(1):115–119. DOI:10.4269/ajtmh.2003.68.1.0680115
  • Batran SE, Salih MM, Elhassan EM, et al. CD20, CD3, placental malaria infections and low birth weight in an area of unstable malaria transmission in Central Sudan. Diagn Pathol. 2013;8(1):189. DOI:10.1186/1746-1596-8-189
  • Boström S, Schmiegelow C, Abu Abed U, et al. Neutrophil alterations in pregnancy-associated malaria and induction of neutrophil chemotaxis by Plasmodium falciparum. Parasite Immunol. 2017;39(6):e12433. DOI:10.1111/pim.12433
  • Umbers AJ, Boeuf P, Clapham C, et al. Placental malaria-associated inflammation disturbs the insulin-like growth factor axis of fetal growth regulation. J Infect Dis. 2011;203(4):561–569. DOI:10.1093/infdis/jiq080
  • Menendez C, Ordi J, Ismail MR, et al. The Impact of Placental Malaria on Gestational Age and Birth Weight. J Infect Dis. 2000;181(5):1740–1745. DOI:10.1086/315449
  • Ordi J, Ismail MR, Ventura PJ, et al. Massive Chronic Intervillositis of the Placenta Associated with Malaria Infection. Am J Surg Pathol. 1998;22(8):1006–1011. DOI:10.1097/00000478-199808000-00011
  • Conroy AL, Silver KL, Zhong K, et al. Complement Activation and the Resulting Placental Vascular Insufficiency Drives Fetal Growth Restriction Associated with Placental Malaria. Cell Host Microbe. 2013;13(2):215–226. DOI:10.1016/j.chom.2013.01.010
  • Elphinstone RE, Weckman AM, McDonald CR, et al. Early malaria infection, dysregulation of angiogenesis, metabolism and inflammation across pregnancy, and risk of preterm birth in Malawi: a cohort study. PLoS Med. 2019;16(10):e1002914. DOI:10.1371/journal.pmed.1002914
  • Kabyemela ER, Fried M, Kurtis JD, et al. Fetal responses during placental malaria modify the risk of low birth weight. Infect Immun. 2008;76(4):1527–1534. DOI:10.1128/IAI.00964-07
  • Dobaño C, Bardají A, Arévalo-Herrera M, et al. Cytokine signatures of Plasmodium vivax infection during pregnancy and delivery outcomes. PLoS Negl Trop Dis. 2020;14(5):e0008155. DOI:10.1371/journal.pntd.0008155
  • Singh KP, Shakeel S, Naskar N, et al. Role of IL-1β, IL-6 and TNF-α cytokines and TNF-α promoter variability in Plasmodium vivax infection during pregnancy in endemic population of Jharkhand, India. Mol Immunol. 2018;97:82–93. DOI:10.1016/j.molimm.2018.03.019
  • McDonald CR, Cahill LS, Gamble JL, et al. Malaria in pregnancy alters l-arginine bioavailability and placental vascular development. Sci Transl Med. 2018;10(431):eaan6007. DOI:10.1126/scitranslmed.aan6007
  • Mathenge PG, Low SK, Vuong NL, et al. Efficacy and resistance of different artemisinin-based combination therapies: a systematic review and network meta-analysis. Parasitol Int. 2020;74:101919. DOI:10.1016/j.parint.2019.04.016
  • WWARN K13 Genotype-Phenotype Study. Association of mutations in the Plasmodium falciparum Kelch13 gene (Pf3D7_1343700) with parasite clearance rates after artemisinin-based treatments—a WWARN individual patient data meta-analysis. BMC Med. 2019;17(1):1. doi: 10.1186/s12916-018-1207-3.
  • Hamilton WL, Amato R, van der Pluijm RW, et al. Evolution and expansion of multidrug-resistant malaria in southeast Asia: a genomic epidemiology study. Lancet Infect Dis. 2019;19(9):943–951. DOI:10.1016/S1473-3099(19)30392-5
  • RW VDP, Imwong M, Chau NH, et al. Determinants of dihydroartemisinin-piperaquine treatment failure in Plasmodium falciparum malaria in Cambodia, Thailand, and Vietnam: a prospective clinical, pharmacological, and genetic study. Lancet Infect Dis. 2019;19(9):952–961. DOI:10.1016/S1473-3099(19)30391-3
  • Na-Bangchang K, Muhamad P, Ruaengweerayut R, et al. Identification of resistance of Plasmodium falciparum to artesunate-mefloquine combination in an area along the Thai-Myanmar border: integration of clinico-parasitological response, systemic drug exposure, and in vitro parasite sensitivity. Malar J. 2013;12(1):263. DOI:10.1186/1475-2875-12-263
  • Leang R, Taylor WRJ, Bouth DM, et al. Evidence of Plasmodium falciparum Malaria Multidrug Resistance to Artemisinin and Piperaquine in Western Cambodia: dihydroartemisinin-Piperaquine Open-Label Multicenter Clinical Assessment. Antimicrob Agents Chemother. 2015;59(8):4719–4726. DOI:10.1128/AAC.00835-15
  • Smith SJ, Kamara ARY, Sahr F, et al. Efficacy of artemisinin-based combination therapies and prevalence of molecular markers associated with artemisinin, piperaquine and sulfadoxine-pyrimethamine resistance in Sierra Leone. Acta Trop. 2018;185:363–370. DOI:10.1016/j.actatropica.2018.06.016
  • Gansané A, Moriarty LF, Ménard D, et al. Anti-malarial efficacy and resistance monitoring of artemether-lumefantrine and dihydroartemisinin-piperaquine shows inadequate efficacy in children in Burkina Faso, 2017–2018. Malar J. 2021;20(1):48. DOI:10.1186/s12936-021-03585-6
  • Kakolwa MA, Mahende MK, Ishengoma DS, et al. Efficacy and safety of artemisinin-based combination therapy, and molecular markers for artemisinin and piperaquine resistance in Mainland Tanzania. Malar J. 2018;17(1):369. DOI:10.1186/s12936-018-2524-x
  • Mayengue PI, Niama RF, Kouhounina Batsimba D, et al. No polymorphisms in K13-propeller gene associated with artemisinin resistance in Plasmodium falciparum isolated from Brazzaville, Republic of Congo. BMC Infect Dis. 2018;18(1):538. DOI:10.1186/s12879-018-3453-6
  • Arya A, Kojom Foko LP, Chaudhry S, et al. Artemisinin-based combination therapy (ACT) and drug resistance molecular markers: a systematic review of clinical studies from two malaria endemic regions - India and sub-Saharan Africa. Int J Parasitol Drugs Drug Resist. 2021;15:43–56. DOI:10.1016/j.ijpddr.2020.11.006
  • Uwimana A, Legrand E, Stokes BH, et al. Emergence and clonal expansion of in vitro artemisinin-resistant Plasmodium falciparum kelch13 R561H mutant parasites in Rwanda. Nat Med. 2020;26(10):1602–1608. DOI:10.1038/s41591-020-1005-2
  • Ikeda M, Kaneko M, Tachibana S-I, et al. Artemisinin - Resistant Plasmodium falciparum with High Survival Rates, Uganda, 2014–2016. Emerg Infect Dis. 2018;24(4):718–726. DOI:10.3201/eid2404.170141
  • Scott N, Ataide R, Wilson DP, et al. Implications of population-level immunity for the emergence of artemisinin-resistant malaria: a mathematical model. Malar J. 2018;17(1):279. DOI:10.1186/s12936-018-2418-y
  • Arora T, Sharma S. Global scenario of counterfeit antimalarials: a potential threat. J Vector Borne Dis. 2019;56(4):288–294.
  • Hassett MR, Roepe PD. In vitro growth competition experiments that suggest consequences of the substandard artemisinin epidemic that may be accelerating drug resistance in P. falciparum malaria. PLoS One. 2021;16(3):e0248057.
  • Levin M, Cunnington AJ, Wilson C, et al. Effects of saline or albumin fluid bolus in resuscitation: evidence from re-analysis of the FEAST trial. Lancet Respir Med. 2019;7(7):581–593. DOI:10.1016/S2213-2600(19)30114-6
  • Meremikwu M, Marson AG. Routine anticonvulsants for treating cerebral malaria. Cochrane Database Syst Rev. 2002;2002:CD002152.
  • Birbeck GL, Herman ST, EV C, et al. A clinical trial of enteral Levetiracetam for acute seizures in pediatric cerebral malaria. BMC Pediatr. 2019;19(1):399. DOI:10.1186/s12887-019-1766-2
  • Mohanty S, Mishra SK, Patnaik R, et al. Brain swelling and mannitol therapy in adult cerebral malaria: a randomized trial. Clin Infect Dis. 2011;53(4):349–355. DOI:10.1093/cid/cir405
  • Commons RJ, Simpson JA, Thriemer K, et al. The effect of chloroquine dose and primaquine on Plasmodium vivax recurrence: a WorldWide Antimalarial Resistance Network systematic review and individual patient pooled meta-analysis. Lancet Infect Dis. 2018;18(9):1025–1034. DOI:10.1016/S1473-3099(18)30348-7
  • Stepniewska K, Humphreys GS, Gonçalves BP, et al. Efficacy of Single-Dose Primaquine with Artemisinin Combination Therapy on Plasmodium falciparum Gametocytes and Transmission: an Individual Patient Meta-Analysis. J Infect Dis. 2022;225(7):1215–1226. DOI:10.1093/infdis/jiaa498
  • Howes RE, Piel FB, Patil AP, et al. G6PD deficiency prevalence and estimates of affected populations in malaria endemic countries: a geostatistical model-based map. PLoS Med. 2012;9(11):e1001339. DOI:10.1371/journal.pmed.1001339
  • Lacerda MVG, Llanos-Cuentas A, Krudsood S, et al. Single-Dose Tafenoquine to Prevent Relapse of Plasmodium vivax Malaria. N Engl J Med. 2019;380(3):215–228. DOI:10.1056/NEJMoa1710775
  • Rueangweerayut R, Bancone G, Harrell EJ, et al. Hemolytic Potential of Tafenoquine in Female Volunteers Heterozygous for Glucose-6-Phosphate Dehydrogenase (G6PD) Deficiency (G6PD Mahidol Variant) versus G6PD-Normal Volunteers. Am J Trop Med Hyg. 2017;97(3):702–711. DOI:10.4269/ajtmh.16-0779
  • Chu CS, Bancone G, Nosten F, et al. Primaquine-induced haemolysis in females heterozygous for G6PD deficiency. Malar J. 2018;17(1):101. DOI:10.1186/s12936-018-2248-y
  • Recht J, Ashley EA, White NJ. Use of primaquine and glucose-6-phosphate dehydrogenase deficiency testing: divergent policies and practices in malaria endemic countries. PLoS Negl Trop Dis. 2018;12(4):e0006230.
  • Gaudinski MR, Berkowitz NM, Idris AH, et al. A Monoclonal Antibody for Malaria Prevention. N Engl J Med. 2021;385(9):803–814. DOI:10.1056/NEJMoa2034031
  • Bernabeu M, Gunnarsson C, Vishnyakova M, et al. Binding Heterogeneity of Plasmodium falciparum to Engineered 3D Brain Microvessels is Mediated by EPCR and ICAM-1. MBio. 2019;10(3): e00420-19. https://doi.org/10.1128/mBio.00420-19.
  • Sharma A, Jenkins B, Akue A, et al. Plasmodium falciparum in Aotus nancymaae: a New Model for Placental Malaria. J Infect Dis. 2022;226(3):jiac096. DOI:10.1093/infdis/jiac096
  • Schäfer C, Dambrauskas N, Reynolds LM, et al. Partial protection against P. vivax infection diminishes hypnozoite burden and blood-stage relapses. Cell Host Microbe. 2021;29(5):752–756.e4. DOI:10.1016/j.chom.2021.03.011
  • McCarthy JS, Griffin PM, Sekuloski S, et al. Experimentally induced blood-stage Plasmodium vivax infection in healthy volunteers. J Infect Dis. 2013;208(10):1688–1694. DOI:10.1093/infdis/jit394
  • Luiza-Batista C, Thiberge S, Serra-Hassoun M, et al. Humanized mice for investigating sustained Plasmodium vivax blood-stage infections and transmission. Nat Commun. 2022;13(1):4123. DOI:10.1038/s41467-022-31864-6
  • Tsuboi T, Del Portillo HA, Mueller I. Editorial on the special issue on Plasmodium vivax: current situation and challenges towards elimination. Parasitol Int. 2022;89:102594.
  • Lee W-C, Cheong FW, Amir A, et al. Plasmodium knowlesi: the game changer for malaria eradication. Malar J. 2022;21(1):140. DOI:10.1186/s12936-022-04131-8