Figures
Abstract
Malaria is a complex parasitic disease caused by species of Plasmodium parasites. Infection with the parasites can lead to a spectrum of symptoms and disease severity, influenced by various parasite, host, and environmental factors. There have been some successes in developing vaccines against the disease recently, but the vaccine efficacies require improvement. Some issues associated with the difficulties in developing a sterile vaccine include high antigenic diversity, switching expression of the immune targets, and inhibition of immune pathways. Current vaccine research focuses on identifying conserved and protective epitopes, developing multivalent vaccines (including the whole parasite), and using more powerful adjuvants. However, overcoming the systematic immune inhibition and immune cell dysfunction/exhaustion may be required before high titers of protective antibodies can be achieved. Increased expression of surface molecules such as CD86 and MHC II on antigen-presenting cells and blocking immune checkpoint pathways (interactions of PD-1 and PD-L1; CTLA-4 and CD80) using small molecules could be a promising approach for enhancing vaccine efficacy. This assay reviews the factors affecting the disease severity, the genetics of host–parasite interaction, immune evasion mechanisms, and approaches potentially to improve host immune response for vaccine development.
Citation: Su X-z, Xu F, Stadler RV, Teklemichael AA, Wu J (2025) Malaria: Factors affecting disease severity, immune evasion mechanisms, and reversal of immune inhibition to enhance vaccine efficacy. PLoS Pathog 21(1): e1012853. https://doi.org/10.1371/journal.ppat.1012853
Editor: Keke C. Fairfax, University of Utah, UNITED STATES OF AMERICA
Published: January 23, 2025
This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.
Funding: This work was supported by the Division of Intramural Research (to XzS, project # ZIA AI000892-23), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), USA. The funders had no role in the design, data collection, analysis, and decision to publish or prepare the manuscript.
Competing interests: The authors have declared that no competing interests exist.
1. Malaria and parasite life cycle
We typically describe malaria as a life-threatening disease caused by Plasmodium parasites through the bites of infected female Anopheles mosquitoes. However, malaria is not a single disease in many ways, and many factors can influence disease outcomes (Fig 1). First, humans can be infected by 6 Plasmodium species, including Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae, Plasmodium knowlesi, Plasmodium cynomolgi, and Plasmodium ovale, which has 2 subspecies, P. o. cutisi and P. o. wallikeri. Although infections with malaria parasites result in some common symptoms (fever and anemia), each parasite species or strain can cause specific disease phenotypes. Second, mix-infections of more than one malaria parasite species or strains and co-infections with other pathogens can alter disease progression and severity. Third, the host’s genetic background plays a significant role in the outcome of an infection. Fourth, the strain, timing, and number of prior exposures (immune status) may determine the level and direction of the host immune response. Fifth, host nutritional status and administration of anti-malarial drugs can also influence disease outcomes. Indeed, individuals infected with malaria parasites may be asymptomatic, can be mild with fever, or develop into a disease with severe anemia, respiratory distress, metabolic acidosis, cerebral malaria, and multi-organ failure leading to death. All malaria parasite species have similar life cycles and developmental stages, although the time to complete a life cycle is species specific (Fig 2).
Parasite factors include molecules mediating the invasion of RBCs and modulating host immune responses. Host genetic background can influence parasite invasion of RBCs and host immunity, too. Prior infections, nutritional status, and anti-malaria treatment also contribute to disease severity.
Sporozoites are injected into the human skin when an infected mosquito bites a human host. The parasites migrate to the liver, developing into schizonts containing thousands of merozoites. Merozoites are released into the blood and invade RBCs, within which they develop from rings to trophozoites to schizonts. Mature schizonts again release merozoites to infect more RBCs. Some of the merozoites differentiate into male and female gametocytes. When another mosquito takes blood, the male and female gametocytes fertilize to produce zygotes that differentiate into ookinetes and oocysts. Sporozoites within oocysts migrate to the salivary glands of the mosquito. When the mosquito bites another human host, the sporozoites will start a new cycle. iRBCs, parasite nucleic acids, and metabolites can be picked up by DCs and macrophages (Mac) to trigger immune responses through activation of Toll-like (TLR) and other receptors in immune organs such as the spleen or lymph nodes in early infection. Activation of DCs and other antigen-presenting cells will activate T and B cells, leading to antibody production later. Parasite materials such as hemozoin could also induce immune cell exhaustion and immune inhibition later in the infection. Other immune mechanisms are also activated, such as interferon signaling against liver stages and the Toll, Imd, JNK, and STAT pathways to kill mosquito stages. Some images of cells and receptors were adopted from BioRender.
2. Parasite genetic factors affecting disease outcomes
Both host and parasite populations are genetically complex, and the interaction of the complex genomes can influence disease outcomes. Dissecting the molecular mechanism of host–parasite interaction and a better understanding of the roles of host and parasite genetic factors during an infection will provide critical information for disease management and control.
Malaria parasite species and disease severity
Different parasite species inherently cause diseases with various symptoms or disease severity. For instance, most malaria-related deaths are caused by P. falciparum, while P. vivax infection is generally nonlethal, underscoring the crucial role of parasite genetics and biology in disease manifestation. P. vivax only invades reticulocytes and produces hypnozoites, which can cause relapses in the liver, whereas P. falciparum can infect all red blood cell (RBC) types and does not cause relapses. The selective invasion of reticulocytes by P. vivax could contribute to the relatively “mild” disease because the parasites will grow slowly in searching for a host cell suitable for invasion, which may give the host more time to produce antibodies to control the infection.
Genetic differences within a species
Infection with different P. falciparum strains can also contribute to the variation in disease manifestation. Infections of different P. falciparum strains or parasites carrying specific antigenic alleles were associated with parasite density and disease severity. For example, the merozoite surface protein 1 (MSP-1) allelic family MSP1-MAD20 was associated with high parasitemia [1]. Infections with parasites carrying MSP-1 RO33/K1 alleles were associated with fever, independent of age and parasite density [2].
Mixed species/strain infections
In malaria-endemic regions, patients have a high frequency of multi-strain infection (30% to 60% of the patients) [3]. Patients with mixed species infection had a higher proportion of severe anemia, pulmonary complications, and multiple organ failure than those with P. falciparum mono-infection [4]. The complexity of the FC27 genotypes was significantly higher in 405 children with severe malaria in Boulgou, Burkina Faso [5]. Therefore, the number of strains in a P. falciparum infection correlates with increased severity.
Co-infection with other pathogens
A malaria patient may be co-infected with other disease agents, including other parasite species, fungi, bacteria, and viruses. These pathogens can interact with each other and the host immune system to influence disease outcomes. Co-infection of mice with Trypanosoma brucei and Plasmodium berghei increased the number of both parasites, leading to more severe anemia, hypoglycemia, and lower survival [6]. Helminth infections have been reported to increase or decrease susceptibility against Plasmodium infections in animal models and human populations [7]. Patients with malaria appeared at greater risk of bacteremia and death, although the prevalence of co-infection was low [8].
Diverse disease pathologies and virulence from rodent parasite species and strains
Parasite genetics affecting malaria disease phenotypes have clearly been demonstrated using the rodent malaria parasite Plasmodium yoelii. Inbred mice can be infected with parasite strains having diverse or similar genetic backgrounds, and the disease symptoms or severity can be compared, providing direct evidence of the effect of parasite genetic variation on disease phenotypes. P. yoelii strains 17XNL, 17XL, and YM were derived from 17X during propagation in different laboratories. However, 17XL and YM are lethal, killing C57BL/6j mice in approximately 7 days after injections of 1 × 106 infected RBCs, whereas 17X and 17XNL are nonlethal and are cleared by the host day 20 postinfection (pi) [9]. The YM (17XL) parasites grow fast and likely kill their hosts through severe anemia. An amino acid (C713R) substitution in domain 6 of the P. yoelii erythrocyte-binding-like (PyEBL) altered parasite growth rate and disease phenotype [10]. Similarly, subspecies Plasmodium y. nigeriensis N67 and N67C are isogenic strains. N67C kills the host in 7 days due to a high level of inflammation, but mice infected with N67 can survive for 20 days [9]. Again, another amino acid substitution in the domain 6 (C741Y) of the PyEBL changed the parasite growth, host immune responses, and disease phenotypes [11]. A single amino acid change (S to F) in the Plasmodium berghei (Pb) NK65 ApiAP2 gene resulted in early IFN-γ responses and high levels of IgG2b and IgG2c antibodies [12].
Co-infections of mice with P. yoelii and Plasmodium vinckei or Plasmodium chabaudi increased virulence, leading to 100% mortality, compared to no mortality for single infections of P. yoelii CU strain or P. vinckei DS strain and 40% mortality for P. chabaudi AJ single infections [13]. Virulent P. chabaudi clones had a competitive advantage over nonvirulent clones in the acute phase of mixed infections [14]. Indeed, evolution modeling of rodent and human malaria infections showed strong positive correlations between asexual multiplication, transmission rate, infection length, morbidity, and mortality and predicted parasite populations evolving to new levels of virulence in response to vaccines and drugs [15].
3. Host factors affecting disease progression and severity
Many host factors can affect the efficiencies of parasite invasion of RBCs and host immunity. Understanding the effects of these factors on disease outcomes will help us design better laboratory experiments and clinical studies.
Host genetic variants affecting parasite invasion and growth
Host genetic variants can confer resistance to malaria infection. The Duffy Antigen Receptor for Chemokines (DARC) is expressed on the surface of RBCs, and P. vivax uses DARC to invade RBCs [16]. DARC-negative human erythrocytes are resistant to invasion by P. vivax, although complement receptor 1 (CR1) was recently shown to be another receptor for P. vivax invasion [17]. Similarly, P. falciparum erythrocyte binding antigen 175 (PfEBA-175) and P. falciparum MSP-1 can bind glycophorin A (GYPA) for invasion [18]. For additional host molecules that can impact parasite growth, invasion of RBCs, and host–parasite interaction, readers can consult the reviews published previously [19,20].
Host immune molecules affecting parasite growth and disease severity
The host’s genetic background can affect the level and direction of immune response to malaria parasite infections. For example, a Toll-like receptor 2 (TLR2) Δ22 polymorphism was associated with protection from cerebral malaria in a case-control study [21]. Proinflammatory cytokines such as IL-1, IL-6, IL-8, IL-12, IFN-γ, and TNF-α are critical for controlling Plasmodium infection, and the plasma IL-10:TNF-α ratio was associated with TNF promoter variants and could predict malarial complications [22]. Genetic variations in the IFN-γ gene were also associated with cerebral malaria (CM), suggesting that IFN-γ protects against CM through anti-parasite activity [23].
The timing of infection and cytokine dynamics
When parasites are injected, the host mounts an innate response with proinflammatory cytokine production and immune cell activation to control pathogen replication. As the infection progresses, the host responses turn into anti-inflammatory or anti-disease responses by producing IL-10 and induction of regulatory subsets of innate immune cells (immunoregulation) [24]. Levels of cytokines are dynamic, typically with an increase in early infection and decrease gradually as the infection progresses. For example, most cytokines and chemokines peaked days 4 to 7 postinjection of P. yoelii 17XNL infected red blood cells (iRBCs) and declined to baseline levels on day 10 postinjection [25], suggesting a state of immune suppression. In clinical studies, blood samples were generally collected from patients without knowing the time of infection or disease progression status. The readouts between individual clinical samples may not be comparable due to the timing of infection and the dynamic responses to malaria parasite infections and could present problems for comparative or association studies.
Prior exposure and persistent parasitemia on host immune capability
Previous exposure history can be a significant factor in determining disease outcomes and vaccination efficacy. Multiple episodes of infection or persistent blood-stage parasitemia will induce anti-disease immunity that can inhibit anti-infection immune response and negatively affect vaccine-induced protection [24,26]. Repeated Plasmodium infections will suppress protective inflammatory responses, which may also interfere with the ability to generate suitable protective immunity, including producing high titers of antibodies [27].
4. Mechanisms of immune inhibition and evasion
Malaria parasites developed various immune escape strategies to survive within their hosts [28,29]. One advantage of the blood stages of malaria parasites is that RBCs do not have a nucleus and lack the expression of immune genes. Thus, the parasites can avoid a direct attack from intracellular defense mechanisms such as autophagy or recognition of pathogen recognition receptors within the host cells. Here, we summarize the major evasion mechanisms employed by malaria parasites (Fig 3).
(A) Parasite lives within RBCs, avoiding various intracellular killing mechanisms, although parasite proteins expressed on the surface of RBCs can be recognized by the host immune system. (B) Parasite molecules exposed to host immunity are highly polymorphic, allowing survival under strain-specific immunity. (C) Expression of a different copy of a variant antigen gene (var) can also evade immunity against a previously expressed variant. (D) Binding of parasite proteins such as PfEMP1 and PfRIFIN to various host cells can induce immune inhibition and avoid clearance by the spleen. (E) Binding of parasite proteins to molecules in the host complement system blocks complement-mediated lysis and killing of the parasites. (F) Genome-wide transcriptional analyses show down-regulation of host genes in many immune pathways, including B and T cell activations. (G) DC dysfunction with reduced expression of many surface proteins required for T cell activation. (H) Malaria parasite infection also leads to T cell dysfunction and exhaustion with increased expression of T cell exhaustion and senescence markers. (I) Expansion of a group of B cells with reduced B cell receptor signaling was recognized in malaria. The status and functions of the atypical B cell required further investigation. (J) Dysfunction of macrophages and NK cells was also observed in malaria. Dysfunction of erythropoietic island macrophage can also cause anemia. (K) Antibodies from prior exposure or immunization may block the binding of new antibodies generated. (L) Many immune checkpoint pathways are activated in malaria, and blockade of the pathways can reverse immune inhibition, inhibit parasite growth, and enhance host survival in rodent malaria models. Note that many of these elements are overlapping and can be integrated into a broad systematic immune inhibition. Some images of cells and receptors were adopted from BioRender.
Antigen diversity
Malaria antigens such as MSP-1 and -2, apical membrane antigen 1 (AMA1), and circumsporozoite protein (CSP) are highly diverse in parasite populations [30]. The extensive diversity of malarial surface antigens is one of the mechanisms for immune evasion and the main difficulties in developing an effective vaccine (Fig 3). Immunity against the erythrocytic stages of the rodent malaria parasite P. c. chabaudi was shown to be strain specific, further supporting an immune escaping mechanism by changing the immune targets [31].
The variant var, rif, and stevor genes and antigenic variation
The protein encoded by the var, rif, and stevor genes, their binding receptors, and potential functions in parasite biology have been well summarized [32]. In addition to having many gene copies and switching gene expression leads to antigenic variation, the var genes encoded protein PfEMP-1 mediates cytoadherence of iRBCs to endothelium and formation of rosettes to avoid clearance by the spleen [32]. They can also suppress cytokine and chemokine production by inhibiting NF-κB signaling in monocytes and macrophages, reduce dendritic cell (DC) antigen-presenting ability by binding to CD36 and CD51 [33], and inhibit cytokine release by NK cells and γδ T cells [34]. Therefore, the var genes play diverse roles in immune evasion and suppression.
RIFIN-mediated inhibition
The P. falciparum genome also has approximately 150 copies of genes encoding RIFIN (repetitive interspersed families of proteins) expressed on the infected red cell surface [35]. RIFIN can suppress host immune cell activation through direct interaction with host receptors, including Type A erythrocyte antigen, sialic acid on glycophorin A, leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), leukocyte immunoglobulin-like receptor subfamily B 1 (LILRB1), and LILRB2 [36]. LAIR1, LILRB1, and LILRB2 are inhibitory immune receptors, and the binding of RIFIN to LILRB1 on immune cells can suppress IgM production by B cells and reduce cytotoxicity NK cells [36]. Interruption of the interaction between RIFIN and the inhibitory immune receptors may partially release immune suppression and improve host-protective immune responses.
STEVOR and antigenic variation
STEVORs (subtelomeric variant open reading frame, approximately 30 copies) are variant proteins that are expressed in Maurer’s clefts, the cytosol and membrane of schizont-infected RBCs, and at the apical end of merozoites [37,38]. The expression of different STEVORs on the surface of the iRBC plays a role in the antigenic diversity and agglutination of iRBCs [39]. Malaria parasites infecting rodents and nonhuman primates also have the Plasmodium interspersed repeat (pir) multigene gene family. The proteins encoded by these genes are expressed on or near the surface of iRBCs and are involved in antigenic variation and immune evasion [40].
Evasion of complement lysis
Human antibodies were found to activate complement against P. falciparum sporozoites and were associated with protection against malaria in children [41]. P. falciparum gamete surface protein PfGAP50 could bind to host complement regulator factor H (FH) and inactivate protein C3b to evade complement-mediated lysis within the mosquito midgut [42]. Similarly, P. falciparum merozoites recruited FH and C1 esterase inhibitor (C1-INH) to escape complement-mediated lysis [43,44]. More recently, P. falciparum gametes and sporozoites were shown to hijack plasmin to evade complement attack by degrading C3b [45].
Hemozoin-mediated immune inhibition
Hemozoin (HZ) is a key product produced by malaria parasites. HZ has been associated with severe malaria anemia (SMA), immunosuppression, and cytokine dysfunction [46]. Macrophages had impaired functions with a long-lasting oxidative burst after ingestion of P. falciparum-infected erythrocytes or isolated malarial HZ [47]. Differentiation and maturation to DC from HZ-fed monocytes were impaired with a blunted expression of MHC II and co-stimulatory molecules CD1a, CD40, CD54, CD80, and CD83 [48]. HZ-induced DC dysfunction compromised CD4+ Tfh function and reduced B cell responses via NLRP3 inflammasome activation [49]. In contrast, malaria HZ could act as a proinflammatory danger signal that activates the NALP3 inflammasome and releases IL-1β [50]. In another study, pure synthetic HZ stimulation of naive murine macrophages activated NF-κB and ERK signaling in naive murine macrophages independent of MyD88 and the NALP3 inflammasome pathway via release of uric acid in vivo [51]. Purified HZ was also found to up-regulate DC maturation with marked increases in cell-surface molecules and IL-12 production [52], which can be explored as an adjuvant to improve vaccine efficacy [53].
Systematic immune suppression
Malaria-induced immune suppression was reported in a human vaccination study decades ago [54]. In an analysis of gene expression of samples from experimental infection of malaria-naïve volunteers and Cameroonian adults infected with P. falciparum, 2 to 3 times more genes were repressed than induced compared to uninfected controls [55]. Additionally, up-regulated genes enriched in GO terms of the immune response, inflammatory response, antigen presentation, and other immune pathways were observed in the malaria-naïve volunteers but not in the Cameroonian adults. In a more recent study of controlled malaria infection of Europeans and Africans, preexposed African individuals showed an increase in regulatory T (Treg) cells, ILC2s, and cells expressing PD-1, whereas Europeans had increased classical monocytes and highly up-regulated interferons [56]. Asymptomatic malaria infections also showed immunosuppressive blood transcriptional signature with up-regulation of pathways involved in controlling T-cell function, including the gene encoding cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) [57]. Infections of different P. yoelii strains or subspecies down-regulated many genes in host immune response pathways such as antigen processing and presentation, DC and monocyte chemotaxis, positive regulation of αβT-cell activation, Th1 activation, DC maturation, and NFAT immune regulation [58].
Dysfunction and exhaustion of immune cells
The observations of systemic immune suppression indicate dysfunction of immune cells. Adherence of intact P. falciparum-infected erythrocytes to DCs derived from human peripheral blood cells can inhibit DC maturation and reduce their capacity to stimulate T cells [59]. Blood-stage parasites could inhibit mouse DC maturation and the capacity to initiate CD8+ T cell immune responses against the initial liver stage of P. yoelii parasites [60]. Repeated malaria parasite infection impaired DC function that could affect the generation of helper T cells and B cell responses in animal models and humans [61].
Malaria parasite infection of mice and humans also causes T-cell exhaustion characterized by poor effector functions and the loss of the cells by apoptosis [62]. Increased expression of T cell exhaustion and senescence markers (PD-1, CTLA-4, and CD57) on CD8+ and CD4+ T cells were observed in the symptomatic children compared to the asymptomatic and healthy controls [63]. Higher frequencies of PD-1-expressing CD4+ T cells were found in children in Mali after infection with P. falciparum, and blockade of PD-L1 and the inhibitory receptor LAG-3 restored CD4+ T cell function in mice infected with P. yoelii but not in those infected with P. chabaudi [64]. Similarly, PD-1 was expressed in human CD8+ T cells from individuals infected with malaria parasites [65] and mediated the loss of parasite-specific CD8+ T cells during the acute phase of P. chabaudi malaria in mice [66]. Therefore, a blockade of PD-1 will improve CD8 T cell function and protect against chronic malaria [62]. Atypical memory B cells (atMBCs) characterized by the expression of exhaustion markers with reduced B cell receptor signaling and effector function were also observed in malaria patients [67], but the roles of atMBCs in immune inhibition require additional investigation.
Plasmodium DNA, HZ, or extracellular vesicles can impair the function of monocytes and macrophages [68]. Malaria parasite infection induced macrophage dysfunction in P. yoelii 17XNL-infected C57BL/6j mice, leading to SMA [25]. Monocytes loaded with HZ could suppress erythropoiesis in the bone marrow through IFN-γ-mediated apoptosis of the erythroid progenitors and antagonized GATA1 transcriptional activity in P. cynomolgi-infected rhesus macaques [69]. P. falciparum infection polarized and accumulated M1 macrophages in the lungs of patients [70]. SMA patients had a greater proportion of monocytes loaded with HZ than controls, and children with severe malaria showed a lowered expression of TLR2 and TLR4, which correlates with monocyte inactivation [71].
RIFIN on the surface of human iRBCs could bind to the inhibitory receptors LILRB1 or LAIR1 on NK cells and reduce NK cell cytotoxicity [72]. Malaria parasite infection also induced neutrophils with reduced oxidative burst activity in Gambian children, which correlated significantly with markers of hemolysis and HO-1 induction [73].
Modulation of immune signaling pathways
CTLA-4 and PD-1 are negative checkpoint regulators of T cell immune function, and inhibition of these targets is being used as immunotherapies for various cancers. The activation of T cells requires the interaction of the T cell receptor (TCR) and CD28 with MHC II and B7 co-stimulatory molecules (CD80 and CD86) located on the antigen-presenting cells, respectively [74]. The interaction of CTLA-4 with the B7 molecules initiates an inhibitory signal, which can be blocked (checked) by CTLA-4 inhibitors. Similarly, the interaction of PD-1 and PD-L1 leads to the negative regulation of T cells, which can be blocked by inhibitors blocking the interaction [74]. CD80 and CD86 do not bind to T cell receptors with the same affinity or effect [75]. CD86 is important for initiating immune responses, whereas CD80 might dampen immune responses after binding to CTLA-4 or PD-L1 [76]. DCs expressing CD80, but not CD86, could significantly suppress lymphocyte proliferation and increase production of IL-10, TGF-β, and IDO [77].
The PD-1- and CTLA-4-mediated immune inhibition pathways also play an important role in the host response to malaria parasite infections [78]. Higher T cell activation and IFN-γ production levels were observed after antibody-mediated blockade of the CTLA-4 or PD-1 pathways during PbA-infection in the BALB/c mice. PD-1-deficient mice could rapidly and completely clear P. chabaudi infections by increasing parasite-specific CD8+ T cells [66]. Persistent P. falciparum exposure was associated with an increased frequency of CD4+ T cells expressing PD-1 alone and PD-1 in combination with lymphocyte-activation gene-3 (LAG-3) [65]. Similarly, increased CD4+ T cells expressing CTLA-4, OX40, GITR, TNFRII, and CD69 were found in patients acutely infected with P. vivax, P. falciparum, or both [79].
MARCH1 (or MARCHF1) is an E3 ubiquitin ligase and immune regulator that regulates protein levels of CD86, MHC II (HLA-DR alpha and beta), TFRC, FAS, and key molecules (MAVS and STING) in IFN-I pathways [80,81]. MARCH1 deficiency increased CD86+ DC populations and IFN-γ and IL-10 levels in C57BL/6j mice at day 4 after infection with P. yoelii YM, leading to improved host survival [81]. The functions of mDCs were compromised by P. falciparum exposure, with impaired HLA-DR and CD86 expression and effector T cell cytokine responses [82]. An early IFN-I response helps parasitemia control, but chronically elevated levels of IFN-I in later infection may impair adaptive immune response and increase disease severity [83]. CD83 could block IL-10-driven, MARCH1-dependent ubiquitination and degradation of MHC II and CD86 in DCs [84], which may be explored for immunotherapy. There are more immune regulatory molecules, such as SOCS1 and RTP4, that can affect malaria parasite infections [29,85,86]; due to space constraints, we cannot discuss them individually here.
Epitope masking
Antibody avidity and titer after vaccination were associated with increased levels of vaccine efficacy. However, induction and maintenance of high antibody titers above a protective threshold have proven challenging [87]. In one study, CSP-specific antibodies and B cells expanded in humans and Ig-knockin mice following the first and second immunization with PfSPZ, but not after the third dose, which might be mediated through epitope masking via the feedback of circulating anti-CSP antibodies [88]. Indeed, invasion-blocking antibodies were induced by a CSP N-terminal polypeptide of 99 amino acids only when it was presented in the absence of the rest of the protein [89]. Structural-based design of the mature and infection-relevant forms of antigens may improve vaccine efficacy [90].
5. Modulation of immune response to increase vaccine efficacy
To deal with antigen diversity and antigenic variation, much effort has been made to find conserved and protective epitopes for vaccine development. RTS,S/AS01 and R21 are the 2 vaccines approved by WHO. The vaccines are designed based on the “conserved” repeat region of the CSP protein to avoid epitope allelic variation and can provide partial protection [91]. Another approach is to use whole parasites, either sporozoites or asexual stages, as antigens, which has achieved promising results [92,93]. Whole parasite vaccines consist of multiple unknown antigen targets that may generate protective antibodies against more than 1 parasite strain infection. Various efforts have been employed to enhance molecular vaccine efficacy or increase antibody titer. The R21 vaccine used a single virus-like CSP-hepatitis B surface antigen (HBsAg) fusion protein with a much higher proportion of CSP than in the RTS,S, leading to a minimal antibody response to the HBsAg carrier and improved vaccine efficacy [94]. Engineered chimeric antigens of the AMA1 DII loop with RON2L enhanced antibodies targeting conserved epitopes on AMA1 and increased neutralization of nonvaccine type parasites [95]. However, these and other efforts to increase vaccination efficacy or antibody titers by refining epitopes and/or antigen presentation with known adjuvants may not be sufficient to overcome the immune inhibition induced by prior parasite exposure or the presence of parasites in the patient’s blood. Modulation of host immune responses may be required to overcome malaria-induced inhibition. Modulation of host metabolism or immune response to treat malaria has been reported previously. Administration of the glutamine analog 6-diazo-5-oxo-L-norleucine (DON), CD47, or interleukin-15 complex rescued mice from PbA ECM [96–98].
The expression of MHC II and the co-stimulatory molecule CD86 on DCs is required to activate T and B cells to produce antibodies. The key issue in increasing vaccine efficacy may be enhancing the expression of CD86/MHC II on antigen-presenting cells before vaccination, although whether enhancing CD86/MHC II expression can totally reverse malaria-induced immune inhibition remains to be tested. Postponing vaccination until a malaria infection has been cleared to “release” immune inhibition may be a good strategy for vaccinating populations in endemic areas where most individuals have been exposed to malaria multiple times [99]. Screening and testing small molecules (SMs) and peptides to block immune inhibition is becoming an emerging field in treating cancers and other diseases. SMs can also be explored to improve malaria vaccine efficacy by blocking various immune inhibition pathways and/or host–parasite molecular interactions (Fig 4). Vaccinating individuals with multiple prior exposures or active parasite infection could be like driving a car with the handbrake on.
Inhibition of interactions of PD-1/PD-L1 and CTLA-4/CD80 with SMs can block the immune checkpoint inhibition pathways to activate T cells. Additionally, inhibition of MARCH1 using SMs or specific peptides from CD83 may increase levels of CD86 and MHC II expression on DCs, promoting T cell activation. Combining monoclonal antibodies and SMs blocking PD-1/PD-L1 interaction or inhibiting MARCH1 may achieve better results than individually blocking PD-1/PD-L1 or inhibiting MARCH1. Enhancing T cell activities will promote antibody production by B cells, leading to better vaccination efficacy. Some images of cells and receptors were adopted from BioRender.
SM immune checkpoint modulators
Malaria is a curable disease, and artemisinin combination therapies are still effective [100]. Immunotherapies such as PD-1/PD-L1 blockade using monoclonal antibodies, developed for treating cancers, will not be practical for malaria treatment or activation of host immune response before vaccination, mainly due to high costs and potential immune-related adverse events (irAEs). SMs are being developed as checkpoint blockers with advantages over antibody treatment, including lower cost, easier dosing (oral), and better management of irEAs [101]. Antibodies typically have a relatively long half-life in vivo, and in the event of irAEs such as cytokine storm, corticosteroids are used to suppress inflammation, which may, in turn, put the patients at a higher risk of developing infections. SMs generally have a shorter half-life than antibodies and may be synthesized at lower costs. A novel series of [1,2,4]triazolo[4,3- a]pyridines were found to be potent inhibitors of the PD-1/PD-L1 interaction, particularly compound A22 having an IC50 of 92.3 nM in blocking PD-1/PD-L1 interaction [102]. Further structural refinement improved IC50 values to 3.0 to 25 nM [103]. Interestingly, screening random phage libraries for peptides bound to the recombinant human PD-1 identified potential PD-1 checkpoint inhibitors that could be used as adjuvants for vaccines against infectious diseases [104]. In a follow-up study, a 22-amino acid immunomodulatory peptide derived from a Bacillus bacterium (LD01) with similarity to a PD-1 peptide antagonist was a potent immunomodulator that could stimulate T cell responses [105]. When combined with an adenovirus-based or irradiated sporozoite-based malaria vaccine, LD01 significantly enhanced antigen-specific CD8+ T cell expansion. In a study using an AI (artificial intelligence) algorithm to screen a library of approximately 10 million compounds for SMs recognizing a putative binding pocket on CTLA-4, several lead SMs were found to bind to CTLA-4 and inhibit its interaction with CD80 [106]. Compounds bound to CTLA-4 at low micromolar concentrations could inhibit tumor development in hCTLA-4 mice at 5 to 25 mg/kg dosages.
Blockade of MARCH1 degradation of CD86 and MHC II
MARCH1 is a potent immune regulator that can be explored for immune therapies. MARCHI transduced DCs secreted high levels of IL-10 after LPS stimulation and stimulated T cells toward the Treg subset [107]. The TM domain of CD83 could enhance MHC II and CD86 expression by blocking MHC II association with MARCH1 [84]. CD83 may be a promising immune modulator with therapeutic potential, and soluble CD83 can be explored to treat various mouse models of autoimmune and inflammatory diseases [108]. Alternatively, SMs can be identified as inhibitors of MARCH1 activity, although no SM MARCH1 inhibitors have been reported.
Safety concerns on immune therapies
Immune checkpoint blockade therapies are often associated with a spectrum of side effects, with various onsets described for the different toxicities such as cytokine storm [109]. To reduce the chance of inducing irEAs, immune modulators and vaccination can be administered during the dry season when an individual has no active infection and strong inflammation. The SM dosage, half-life in vivo, and the timing of administration will have to be tested in clinical trials before field application.
6. Conclusions
Malaria is a complex disease with a spectrum of symptoms determined by various factors derived from host–parasite genetic interactions. Rodent and nonhuman primate malaria parasites are good models to dissect disease and immune mechanisms. Results from animal models can then be tested in human malaria patients. Clinical association studies are confounded by many unknown or uncontrollable conditions such as genetic variations in the host and parasite, mix-infection, unknown timing of infection, prior exposure, etc. Historically, most studies on host immunity focused on up-regulated genes in response to malaria parasite infections. It is time to pay more attention to the immune pathways inhibited in malaria. Measures to release immune inhibition will be critical to improve vaccine efficacy. Searching and testing SMs to block immune checkpoints (PD-1/PD-L1, for example), as is being done in anti-cancer therapies, may provide a fruitful approach to enhance vaccine efficacy. MARCH1 and CD83, in addition to PD-1 and CTLA-4, are promising immune regulators that can be explored to activate host immune responses, including activation of T-helper cells and production of IFN-I that appear to be associated with successful immunotherapy for cancers [110]. Finally, it will be critical and challenging to find SMs and dosages that can activate the host immune response to enhance vaccine efficacy without triggering a cytokine storm or irAEs.
Acknowledgments
We thank Yolanda L. Jones, NIH Library Editing Service, for manuscript editing assistance.
References
- 1. Sondo P, Derra K, Lefevre T, Diallo-Nakanabo S, Tarnagda Z, Zampa O, et al. Genetically diverse Plasmodium falciparum infections, within-host competition and symptomatic malaria in humans. Sci Rep. 2019;9(1):127. Epub 20190115. pmid:30644435; PubMed Central PMCID: PMC6333925.
- 2. Anong DN, Nkuo-Akenji T, Fru-Cho J, Amambua-Ngwa A, Titanji VP. Genetic diversity of Plasmodium falciparum in Bolifamba, on the slopes of Mount Cameroon: influence of MSP1 allelic variants on symptomatic malaria and anaemia. Ann Trop Med Parasitol. 2010;104(1):25–33. pmid:20149290.
- 3. Zhu SJ, Hendry JA, Almagro-Garcia J, Pearson RD, Amato R, Miles A, et al. The origins and relatedness structure of mixed infections vary with local prevalence of P. falciparum malaria. Elife. 2019;8. Epub 20190712. pmid:31298657; PubMed Central PMCID: PMC6684230.
- 4. Kotepui M, Kotepui KU, De Jesus Milanez G, Masangkay FR. Plasmodium spp. mixed infection leading to severe malaria: a systematic review and meta-analysis. Sci Rep. 2020;10(1):11068. Epub 20200706. pmid:32632180; PubMed Central PMCID: PMC7338391.
- 5. Soulama I, Sawadogo M, Nebie I, Diarra A, Tiono AB, Konate A, et al. [Genetic diversity of P. falciparum and pathogenesis of the severe malarial anaemia in children under 5 years old in the province of Boulgou, Burkina Faso]. Bull Soc Pathol Exot. 2006;99(3):166–70. pmid:16983818.
- 6. Ademola IO, Odeniran PO. Co-infection with Plasmodium berghei and Trypanosoma brucei increases severity of malaria and trypanosomiasis in mice. Acta Trop. 2016;159:29–35. Epub 20160325. pmid:27021269.
- 7. Hartgers FC, Yazdanbakhsh M. Co-infection of helminths and malaria: modulation of the immune responses to malaria. Parasite Immunol. 2006;28(10):497–506. pmid:16965285.
- 8. Wilairatana P, Mala W, Masangkay FR, Kotepui KU, Kotepui M. The Prevalence of Malaria and Bacteremia Co-Infections among Febrile Patients: A Systematic Review and Meta-Analysis. Trop Med Infect Dis. 2022;7(9). Epub 20220913. pmid:36136654; PubMed Central PMCID: PMC9503679.
- 9. Wu J, Oguz C, Teklemichael AA, Xu F, Stadler RV, Lucky AB, et al. Comparative genomics of Plasmodium yoelii nigeriensis N67 and N67C: genome-wide polymorphisms, differential gene expression, and drug resistance. BMC Genomics. 2024;25(1):1035. Epub 20241105. pmid:39497038; PubMed Central PMCID: PMC11536827.
- 10. Otsuki H, Kaneko O, Thongkukiatkul A, Tachibana M, Iriko H, Takeo S, et al. Single amino acid substitution in Plasmodium yoelii erythrocyte ligand determines its localization and controls parasite virulence. Proc Natl Acad Sci U S A. 2009;106(17):7167–72. Epub 20090403. pmid:19346470; PubMed Central PMCID: PMC2678433.
- 11. Peng YC, Qi Y, Zhang C, Yao X, Wu J, Pattaradilokrat S, et al. Plasmodium yoelii Erythrocyte-Binding-like Protein Modulates Host Cell Membrane Structure, Immunity, and Disease Severity. MBio. 2020;11(1). Epub 20200107. pmid:31911494; PubMed Central PMCID: PMC6946805.
- 12. Akkaya M, Bansal A, Sheehan PW, Pena M, Molina-Cruz A, Orchard LM, et al. A single-nucleotide polymorphism in a Plasmodium berghei ApiAP2 transcription factor alters the development of host immunity. Sci Adv. 2020;6(6):eaaw6957. Epub 20200205. pmid:32076635; PubMed Central PMCID: PMC7002124.
- 13. Tang J, Templeton TJ, Cao J, Culleton R. The Consequences of Mixed-Species Malaria Parasite Co-Infections in Mice and Mosquitoes for Disease Severity, Parasite Fitness, and Transmission Success. Front Immunol. 2019;10:3072. Epub 20200122. pmid:32038623; PubMed Central PMCID: PMC6987389.
- 14. Bell AS, de Roode JC, Sim D, Read AF. Within-host competition in genetically diverse malaria infections: parasite virulence and competitive success. Evolution. 2006;60(7):1358–71. pmid:16929653.
- 15. Mackinnon MJ, Read AF. Virulence in malaria: an evolutionary viewpoint. Philos Trans R Soc Lond B Biol Sci. 2004;359(1446):965–86. pmid:15306410; PubMed Central PMCID: PMC1693375.
- 16. Picon-Jaimes YA, Lozada-Martinez ID, Orozco-Chinome JE, Molina-Franky J, Acevedo-Lopez D, Acevedo-Lopez N, et al. Relationship between Duffy Genotype/Phenotype and Prevalence of Plasmodium vivax Infection: A Systematic Review. Trop Med Infect Dis. 2023;8(10). Epub 20230930. pmid:37888591; PubMed Central PMCID: PMC10610806.
- 17. Lee SK, Crosnier C, Valenzuela-Leon PC, Dizon BLP, Atkinson JP, Mu J, et al. Complement receptor 1 is the human erythrocyte receptor for Plasmodium vivax erythrocyte binding protein. Proc Natl Acad Sci U S A. 2024;121(5):e2316304121. Epub 20240123. pmid:38261617; PubMed Central PMCID: PMC10835065.
- 18. Baldwin MR, Li X, Hanada T, Liu SC, Chishti AH. Merozoite surface protein 1 recognition of host glycophorin A mediates malaria parasite invasion of red blood cells. Blood. 2015;125(17):2704–11. Epub 20150316. pmid:25778531; PubMed Central PMCID: PMC4408295.
- 19. Lelliott PM, McMorran BJ, Foote SJ, Burgio G. The influence of host genetics on erythrocytes and malaria infection: is there therapeutic potential? Malar J. 2015;14:289. Epub 20150729. pmid:26215182; PubMed Central PMCID: PMC4517643.
- 20. Kariuki SN, Williams TN. Human genetics and malaria resistance. Hum Genet. 2020;139(6–7):801–11. Epub 20200304. pmid:32130487; PubMed Central PMCID: PMC7271956.
- 21. Greene JA, Sam-Agudu N, John CC, Opoka RO, Zimmerman PA, Kazura JW. Toll-like receptor polymorphisms and cerebral malaria: TLR2 Delta22 polymorphism is associated with protection from cerebral malaria in a case control study. Malar J. 2012;11:47. Epub 20120215. pmid:22336003; PubMed Central PMCID: PMC3306729.
- 22. May J, Lell B, Luty AJ, Meyer CG, Kremsner PG. Plasma interleukin-10:Tumor necrosis factor (TNF)-alpha ratio is associated with TNF promoter variants and predicts malarial complications. J Infect Dis. 2000;182(5):1570–3. Epub 20001009. pmid:11023485.
- 23. Cabantous S, Poudiougou B, Traore A, Keita M, Cisse MB, Doumbo O, et al. Evidence that interferon-gamma plays a protective role during cerebral malaria. J Infect Dis. 2005;192(5):854–60. Epub 20050722. pmid:16088835.
- 24. Boyle MJ, Engwerda CR, Jagannathan P. The impact of Plasmodium-driven immunoregulatory networks on immunity to malaria. Nat Rev Immunol. 2024. Epub 20240611. pmid:38862638.
- 25. Tumas KC, Xu F, Wu J, Hernandez M, Pattaradilokrat S, Xia L, et al. Dysfunction of CD169(+) macrophages and blockage of erythrocyte maturation as a mechanism of anemia in Plasmodium yoelii infection. Proc Natl Acad Sci U S A. 2023;120(40):e2311557120. Epub 20230925. pmid:37748059; PubMed Central PMCID: PMC10556621.
- 26. Murphy SC, Deye GA, Sim BKL, Galbiati S, Kennedy JK, Cohen KW, 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. Epub 20210528. pmid:34048504; PubMed Central PMCID: PMC8191919.
- 27. Montes de Oca M, Good MF, McCarthy JS, Engwerda CR. The Impact of Established Immunoregulatory Networks on Vaccine Efficacy and the Development of Immunity to Malaria. J Immunol. 2016;197(12):4518–26. pmid:27913644.
- 28. Calle CL, Mordmuller B, Singh A. Immunosuppression in Malaria: Do Plasmodium falciparum Parasites Hijack the Host? Pathogens. 2021;10(10). Epub 20211003. pmid:34684226; PubMed Central PMCID: PMC8536967.
- 29. Cai C, Hu Z, Yu X. Accelerator or Brake: Immune Regulators in Malaria. Front Cell Infect Microbiol. 2020;10:610121. Epub 20201210. pmid:33363057; PubMed Central PMCID: PMC7758250.
- 30. Ferreira MU, da Silva Nunes M, Wunderlich G. Antigenic diversity and immune evasion by malaria parasites. Clin Diagn Lab Immunol. 2004;11(6):987–95. pmid:15539495; PubMed Central PMCID: PMC524792.
- 31. Culleton RL, Inoue M, Reece SE, Cheesman S, Carter R. Strain-specific immunity induced by immunization with pre-erythrocytic stages of Plasmodium chabaudi. Parasite Immunol. 2011;33(1):73–8. pmid:21189655; PubMed Central PMCID: PMC3937739.
- 32. Wahlgren M, Goel S, Akhouri RR. Variant surface antigens of Plasmodium falciparum and their roles in severe malaria. Nat Rev Microbiol. 2017;15(8):479–91. Epub 20170612. pmid:28603279.
- 33. Urban BC, Willcox N, Roberts DJ. A role for CD36 in the regulation of dendritic cell function. Proc Natl Acad Sci U S A. 2001;98(15):8750–5. Epub 20010710. pmid:11447263; PubMed Central PMCID: PMC37507.
- 34. D’Ombrain MC, Voss TS, Maier AG, Pearce JA, Hansen DS, Cowman AF, et al. Plasmodium falciparum erythrocyte membrane protein-1 specifically suppresses early production of host interferon-gamma. Cell Host Microbe. 2007;2(2):130–8. pmid:18005727.
- 35. Kyes SA, Rowe JA, Kriek N, Newbold CI. Rifins: a second family of clonally variant proteins expressed on the surface of red cells infected with Plasmodium falciparum. Proc Natl Acad Sci U S A. 1999;96(16):9333–8. pmid:10430943; PubMed Central PMCID: PMC17783.
- 36. Sakoguchi A, Arase H. Mechanisms for Host Immune Evasion Mediated by Plasmodium falciparum-Infected Erythrocyte Surface Antigens. Front Immunol. 2022;13:901864. Epub 20220615. pmid:35784341; PubMed Central PMCID: PMC9240312.
- 37. Przyborski JM, Miller SK, Pfahler JM, Henrich PP, Rohrbach P, Crabb BS, et al. Trafficking of STEVOR to the Maurer’s clefts in Plasmodium falciparum-infected erythrocytes. EMBO J. 2005;24(13):2306–17. Epub 20050616. pmid:15961998; PubMed Central PMCID: PMC1173160.
- 38. Blythe JE, Yam XY, Kuss C, Bozdech Z, Holder AA, Marsh K, et al. Plasmodium falciparum STEVOR proteins are highly expressed in patient isolates and located in the surface membranes of infected red blood cells and the apical tips of merozoites. Infect Immun. 2008;76(7):3329–36. Epub 20080512. pmid:18474651; PubMed Central PMCID: PMC2446718.
- 39. Niang M, Yan Yam X, Preiser PR. The Plasmodium falciparum STEVOR multigene family mediates antigenic variation of the infected erythrocyte. PLoS Pathog. 2009;5(2):e1000307. Epub 20090220. pmid:19229319; PubMed Central PMCID: PMC2637975.
- 40. Cunningham D, Lawton J, Jarra W, Preiser P, Langhorne J. The pir multigene family of Plasmodium: antigenic variation and beyond. Mol Biochem Parasitol. 2010;170(2):65–73. Epub 20100104. pmid:20045030.
- 41. Kurtovic L, Boyle MJ, Opi DH, Kennedy AT, Tham WH, Reiling L, et al. Complement in malaria immunity and vaccines. Immunol Rev. 2020;293(1):38–56. Epub 20190926. pmid:31556468; PubMed Central PMCID: PMC6972673.
- 42. Simon N, Lasonder E, Scheuermayer M, Kuehn A, Tews S, Fischer R, et al. Malaria parasites co-opt human factor H to prevent complement-mediated lysis in the mosquito midgut. Cell Host Microbe. 2013;13(1):29–41. Epub 20130116. pmid:23332154.
- 43. Kennedy AT, Wijeyewickrema LC, Huglo A, Lin C, Pike R, Cowman AF, et al. Recruitment of Human C1 Esterase Inhibitor Controls Complement Activation on Blood Stage Plasmodium falciparum Merozoites. J Immunol. 2017;198(12):4728–37. Epub 20170508. pmid:28484054.
- 44. Rosa TF, Flammersfeld A, Ngwa CJ, Kiesow M, Fischer R, Zipfel PF, et al. The Plasmodium falciparum blood stages acquire factor H family proteins to evade destruction by human complement. Cell Microbiol. 2016;18(4):573–90. Epub 20151112. pmid:26457721; PubMed Central PMCID: PMC5063132.
- 45. Ernest M, Rosa TFA, Pala ZR, Kudyba HM, Sweeney B, Reiss T, et al. Plasmodium falciparum Gametes and Sporozoites Hijack Plasmin and Factor H To Evade Host Complement Killing. Microbiol Spectr. 2023;11(3):e0449322. Epub 20230516. pmid:37191558; PubMed Central PMCID: PMC10269923.
- 46. Ihekwereme CP, Esimone CO, Nwanegbo EC. Hemozoin inhibition and control of clinical malaria. Adv Pharm Sci. 2014;2014:984150. Epub 20140209. pmid:24669217; PubMed Central PMCID: PMC3941158.
- 47. Schwarzer E, Turrini F, Ulliers D, Giribaldi G, Ginsburg H, Arese P. Impairment of macrophage functions after ingestion of Plasmodium falciparum-infected erythrocytes or isolated malarial pigment. J Exp Med. 1992;176(4):1033–41. pmid:1402649; PubMed Central PMCID: PMC2119406.
- 48. Skorokhod OA, Alessio M, Mordmuller B, Arese P, Schwarzer E. Hemozoin (malarial pigment) inhibits differentiation and maturation of human monocyte-derived dendritic cells: a peroxisome proliferator-activated receptor-gamma-mediated effect. J Immunol. 2004;173(6):4066–74. pmid:15356156.
- 49. Pack AD, Schwartzhoff PV, Zacharias ZR, Fernandez-Ruiz D, Heath WR, Gurung P, et al. Hemozoin-mediated inflammasome activation limits long-lived anti-malarial immunity. Cell Rep. 2021;36(8):109586. pmid:34433049; PubMed Central PMCID: PMC8432597.
- 50. Dostert C, Guarda G, Romero JF, Menu P, Gross O, Tardivel A, et al. Malarial hemozoin is a Nalp3 inflammasome activating danger signal. PLoS ONE. 2009;4(8):e6510. Epub 20090804. pmid:19652710; PubMed Central PMCID: PMC2714977.
- 51. Griffith JW, Sun T, McIntosh MT, Bucala R. Pure Hemozoin is inflammatory in vivo and activates the NALP3 inflammasome via release of uric acid. J Immunol. 2009;183(8):5208–20. Epub 20090925. pmid:19783673; PubMed Central PMCID: PMC3612522.
- 52. Coban C, Ishii KJ, Sullivan DJ, Kumar N. Purified malaria pigment (hemozoin) enhances dendritic cell maturation and modulates the isotype of antibodies induced by a DNA vaccine. Infect Immun. 2002;70(7):3939–43. pmid:12065539; PubMed Central PMCID: PMC128057.
- 53. Lee MS, Igari Y, Tsukui T, Ishii KJ, Coban C. Current status of synthetic hemozoin adjuvant: A preliminary safety evaluation. Vaccine. 2016;34(18):2055–61. Epub 20160311. pmid:26976665.
- 54. Williamson WA, Greenwood BM. Impairment of the immune response to vaccination after acute malaria. Lancet. 1978;1(8078):1328–9. pmid:78096
- 55. Ockenhouse CF, Hu WC, Kester KE, Cummings JF, Stewart A, Heppner DG, et al. Common and divergent immune response signaling pathways discovered in peripheral blood mononuclear cell gene expression patterns in presymptomatic and clinically apparent malaria. Infect Immun. 2006;74(10):5561–73. pmid:16988231; PubMed Central PMCID: PMC1594921.
- 56. de Jong SE, Olin A, Pulendran B. The Impact of the Microbiome on Immunity to Vaccination in Humans. Cell Host Microbe. 2020;28(2):169–79. pmid:32791110; PubMed Central PMCID: PMC7422826.
- 57. Studniberg SI, Ioannidis LJ, Utami RAS, Trianty L, Liao Y, Abeysekera W, et al. Molecular profiling reveals features of clinical immunity and immunosuppression in asymptomatic P. falciparum malaria. Mol Syst Biol. 2022;18(4):e10824. pmid:35475529; PubMed Central PMCID: PMC9045086.
- 58. Xia L, Wu J, Pattaradilokrat S, Tumas K, He X, Peng YC, et al. Detection of host pathways universally inhibited after Plasmodium yoelii infection for immune intervention. Sci Rep. 2018;8(1):15280. Epub 20181016. pmid:30327482; PubMed Central PMCID: PMC6191451.
- 59. Urban BC, Ferguson DJ, Pain A, Willcox N, Plebanski M, Austyn JM, et al. Plasmodium falciparum-infected erythrocytes modulate the maturation of dendritic cells. Nature. 1999;400(6739):73–7. pmid:10403251.
- 60. Ocana-Morgner C, Mota MM, Rodriguez A. Malaria blood stage suppression of liver stage immunity by dendritic cells. J Exp Med. 2003;197(2):143–51. pmid:12538654; PubMed Central PMCID: PMC2193811.
- 61. Osii RS, Otto TD, Garside P, Ndungu FM, Brewer JM. The Impact of Malaria Parasites on Dendritic Cell-T Cell Interaction. Front Immunol. 2020;11:1597. Epub 20200724. pmid:32793231; PubMed Central PMCID: PMC7393936.
- 62. Wykes MN, Horne-Debets JM, Leow CY, Karunarathne DS. Malaria drives T cells to exhaustion. Front Microbiol. 2014;5:249. Epub 20140527. pmid:24904561; PubMed Central PMCID: PMC4034037.
- 63. Frimpong A, Kusi KA, Adu-Gyasi D, Amponsah J, Ofori MF, Ndifon W. Phenotypic Evidence of T Cell Exhaustion and Senescence During Symptomatic Plasmodium falciparum Malaria. Front Immunol. 2019;10:1345. Epub 20190618. pmid:31316497; PubMed Central PMCID: PMC6611412.
- 64. Butler NS, Moebius J, Pewe LL, Traore B, Doumbo OK, Tygrett LT, et al. Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection. Nat Immunol. 2011;13(2):188–95. Epub 20111211. pmid:22157630; PubMed Central PMCID: PMC3262959.
- 65. Illingworth J, Butler NS, Roetynck S, Mwacharo J, Pierce SK, Bejon P, et al. Chronic exposure to Plasmodium falciparum is associated with phenotypic evidence of B and T cell exhaustion. J Immunol. 2013;190(3):1038–47. Epub 20121221. pmid:23264654; PubMed Central PMCID: PMC3549224.
- 66. Horne-Debets JM, Faleiro R, Karunarathne DS, Liu XQ, Lineburg KE, Poh CM, et al. PD-1 dependent exhaustion of CD8+ T cells drives chronic malaria. Cell Rep. 2013;5(5):1204–13. Epub 20131205. pmid:24316071.
- 67. Portugal S, Tipton CM, Sohn H, Kone Y, Wang J, Li S, et al. Malaria-associated atypical memory B cells exhibit markedly reduced B cell receptor signaling and effector function. Elife. 2015;4. Epub 20150508. pmid:25955968; PubMed Central PMCID: PMC4444601.
- 68. Ortega-Pajares A, Rogerson SJ. The Rough Guide to Monocytes in Malaria Infection. Front Immunol. 2018;9:2888. Epub 20181207. pmid:30581439; PubMed Central PMCID: PMC6292935.
- 69. Tang Y, Joyner CJ, Cabrera-Mora M, Saney CL, Lapp SA, Nural MV, et al. Integrative analysis associates monocytes with insufficient erythropoiesis during acute Plasmodium cynomolgi malaria in rhesus macaques. Malar J. 2017;16(1):384. Epub 20170922. pmid:28938907; PubMed Central PMCID: PMC5610412.
- 70. Klinkhamhom A, Glaharn S, Srisook C, Ampawong S, Krudsood S, Ward SA, et al. M1 macrophage features in severe Plasmodium falciparum malaria patients with pulmonary oedema. Malar J. 2020;19(1):182. Epub 20200515. pmid:32414377; PubMed Central PMCID: PMC7226720.
- 71. Mandala WL, Msefula CL, Gondwe EN, Drayson MT, Molyneux ME, MacLennan CA. Monocyte activation and cytokine production in Malawian children presenting with P. falciparum malaria. Parasite Immunol. 2016;38(5):317–25. pmid:27027867; PubMed Central PMCID: PMC4850749.
- 72. Saito F, Hirayasu K, Satoh T, Wang CW, Lusingu J, Arimori T, et al. Immune evasion of Plasmodium falciparum by RIFIN via inhibitory receptors. Nature. 2017;552(7683):101–5. Epub 20171129. pmid:29186116; PubMed Central PMCID: PMC5748893.
- 73. Cunnington AJ, Njie M, Correa S, Takem EN, Riley EM, Walther M. Prolonged neutrophil dysfunction after Plasmodium falciparum malaria is related to hemolysis and heme oxygenase-1 induction. J Immunol. 2012;189(11):5336–46. Epub 20121024. pmid:23100518; PubMed Central PMCID: PMC3504608.
- 74. Chae YK, Arya A, Iams W, Cruz MR, Chandra S, Choi J, et al. Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC). J Immunother Cancer. 2018;6(1):39. Epub 20180516. pmid:29769148; PubMed Central PMCID: PMC5956851.
- 75. Xiang J, Gu X, Qian S, Chen Z. Graded function of CD80 and CD86 in initiation of T-cell immune response and cardiac allograft survival. Transpl Int. 2008;21(2):163–8. Epub 20071029. pmid:17971032.
- 76. Zheng Y, Manzotti CN, Liu M, Burke F, Mead KI, Sansom DM. CD86 and CD80 differentially modulate the suppressive function of human regulatory T cells. J Immunol. 2004;172(5):2778–84. pmid:14978077.
- 77. Ke N, Su A, Huang W, Szatmary P, Zhang Z. Regulating the expression of CD80/CD86 on dendritic cells to induce immune tolerance after xeno-islet transplantation. Immunobiology. 2016;221(7):803–12. Epub 20160203. pmid:26879762.
- 78. Faleiro R, Karunarathne DS, Horne-Debets JM, Wykes M. The Contribution of Co-signaling Pathways to Anti-malarial T Cell Immunity. Front Immunol. 2018;9:2926. Epub 20181214. pmid:30631323; PubMed Central PMCID: PMC6315188.
- 79. Goncalves-Lopes RM, Lima NF, Carvalho KI, Scopel KK, Kallas EG, Ferreira MU. Surface expression of inhibitory (CTLA-4) and stimulatory (OX40) receptors by CD4(+) regulatory T cell subsets circulating in human malaria. Microbes Infect. 2016;18(10):639–48. Epub 20160616. pmid:27320393; PubMed Central PMCID: PMC5087799.
- 80. Ishido S, Matsuki Y, Goto E, Kajikawa M, Ohmura-Hoshino M. MARCH-I: a new regulator of dendritic cell function. Mol Cells. 2010;29(3):229–32. Epub 20100304. pmid:20213309.
- 81. Wu J, Xia L, Yao X, Yu X, Tumas KC, Sun W, et al. The E3 ubiquitin ligase MARCH1 regulates antimalaria immunity through interferon signaling and T cell activation. Proc Natl Acad Sci U S A. 2020;117(28):16567–78. Epub 20200630. pmid:32606244; PubMed Central PMCID: PMC7368286.
- 82. Loughland JR, Minigo G, Burel J, Tipping PE, Piera KA, Amante FH, et al. Profoundly Reduced CD1c+ Myeloid Dendritic Cell HLA-DR and CD86 Expression and Increased Tumor Necrosis Factor Production in Experimental Human Blood-Stage Malaria Infection. Infect Immun. 2016;84(5):1403–12. Epub 20160422. pmid:26902728; PubMed Central PMCID: PMC4862702.
- 83. He X, Xia L, Tumas KC, Wu J, Su XZ. Type I Interferons and Malaria: A Double-Edge Sword Against a Complex Parasitic Disease. Front Cell Infect Microbiol. 2020;10:594621. Epub 20201202. pmid:33344264; PubMed Central PMCID: PMC7738626.
- 84. Tze LE, Horikawa K, Domaschenz H, Howard DR, Roots CM, Rigby RJ, et al. CD83 increases MHC II and CD86 on dendritic cells by opposing IL-10-driven MARCH1-mediated ubiquitination and degradation. J Exp Med. 2011;208(1):149–65. Epub 20110110. pmid:21220452; PubMed Central PMCID: PMC3023131.
- 85. Zander RA, Obeng-Adjei N, Guthmiller JJ, Kulu DI, Li J, Ongoiba A, et al. PD-1 Co-inhibitory and OX40 Co-stimulatory Crosstalk Regulates Helper T Cell Differentiation and Anti-Plasmodium Humoral Immunity. Cell Host Microbe. 2015;17(5):628–41. Epub 20150416. pmid:25891357; PubMed Central PMCID: PMC4433434.
- 86. Wu J, Cai B, Sun W, Huang R, Liu X, Lin M, et al. Genome-wide Analysis of Host-Plasmodium yoelii Interactions Reveals Regulators of the Type I Interferon Response. Cell Rep. 2015;12(4):661–72. Epub 20150716. pmid:26190101; PubMed Central PMCID: PMC4520759.
- 87. White MT, Bejon P, Olotu A, Griffin JT, Bojang K, Lusingu J, et al. A combined analysis of immunogenicity, antibody kinetics and vaccine efficacy from phase 2 trials of the RTS,S malaria vaccine. BMC Med. 2014;12:117. Epub 20140710. pmid:25012228; PubMed Central PMCID: PMC4227280.
- 88. McNamara HA, Idris AH, Sutton HJ, Vistein R, Flynn BJ, Cai Y, et al. Antibody Feedback Limits the Expansion of B Cell Responses to Malaria Vaccination but Drives Diversification of the Humoral Response. Cell Host Microbe. 2020;28(4):572–85 e7. Epub 20200721. pmid:32697938.
- 89. Rathore D, Nagarkatti R, Jani D, Chattopadhyay R, de la Vega P, Kumar S, et al. An immunologically cryptic epitope of Plasmodium falciparum circumsporozoite protein facilitates liver cell recognition and induces protective antibodies that block liver cell invasion. J Biol Chem. 2005;280(21):20524–9. Epub 20050321. pmid:15781464.
- 90. Patel PN, Tolia N. Structural vaccinology of malaria transmission-blocking vaccines. Expert Rev Vaccines. 2021;20(2):199–214. Epub 20210119. pmid:33430656; PubMed Central PMCID: PMC11077433.
- 91. Duffy PE, Gorres JP, Healy SA, Fried M. Malaria vaccines: a new era of prevention and control. Nat Rev Microbiol. 2024. Epub 20240718. pmid:39025972.
- 92. Sirima SB, Ouedraogo A, Tiono AB, Kabore JM, Bougouma EC, Ouattara MS, et al. A randomized controlled trial showing safety and efficacy of a whole sporozoite vaccine against endemic malaria. Sci Transl Med. 2022;14(674):eabj3776. Epub 20221207. pmid:36475905; PubMed Central PMCID: PMC10041996.
- 93. Cai J, Chen S, Zhu F, Lu X, Liu T, Xu W. Whole-Killed Blood-Stage Vaccine: Is It Worthwhile to Further Develop It to Control Malaria? Front Microbiol. 2021;12:670775. Epub 20210430. pmid:33995336; PubMed Central PMCID: PMC8119638.
- 94. Collins KA, Snaith R, Cottingham MG, Gilbert SC, Hill AVS. Enhancing protective immunity to malaria with a highly immunogenic virus-like particle vaccine. Sci Rep. 2017;7:46621. Epub 20170419. pmid:28422178; PubMed Central PMCID: PMC5395940.
- 95. Yanik S, Venkatesh V, Parker ML, Ramaswamy R, Diouf A, Sarkar D, et al. Structure guided mimicry of an essential P. falciparum receptor-ligand complex enhances cross neutralizing antibodies. Nat Commun. 2023;14(1):5879. Epub 20230921. pmid:37735574; PubMed Central PMCID: PMC10514071.
- 96. Gordon EB, Hart GT, Tran TM, Waisberg M, Akkaya M, Kim AS, et al. Targeting glutamine metabolism rescues mice from late-stage cerebral malaria. Proc Natl Acad Sci U S A. 2015;112(42):13075–80. Epub 20151005. pmid:26438846; PubMed Central PMCID: PMC4620853.
- 97. Bravo M, Dileepan T, Dolan M, Hildebrand J, Wolford J, Hanson ID, et al. IL-15 Complex-Induced IL-10 Enhances Plasmodium-specific CD4+ T Follicular Helper Differentiation and Antibody Production. J Immunol. 2024;212(6):992–1001. pmid:38305633; PubMed Central PMCID: PMC10932862.
- 98. Torrez Dulgeroff LB, Oakley MS, Tal MC, Yiu YY, He JQ, Shoham M, et al. CD47 blockade reduces the pathologic features of experimental cerebral malaria and promotes survival of hosts with Plasmodium infection. Proc Natl Acad Sci U S A. 2021;118(11). pmid:33836556; PubMed Central PMCID: PMC7980459.
- 99. Cunnington AJ, Riley EM. Suppression of vaccine responses by malaria: insignificant or overlooked? Expert Rev Vaccines. 2010;9(4):409–29. pmid:20370551.
- 100. Wellems TE, Sa JM, Su XZ, Connelly SV, Ellis AC. ‘Artemisinin Resistance’: Something New or Old? Something of a Misnomer? Trends Parasitol. 2020;36(9):735–44. Epub 20200622. pmid:32586776.
- 101. Sasikumar PG, Ramachandra M. Small Molecule Agents Targeting PD-1 Checkpoint Pathway for Cancer Immunotherapy: Mechanisms of Action and Other Considerations for Their Advanced Development. Front Immunol. 2022;13:752065. Epub 20220502. pmid:35585982; PubMed Central PMCID: PMC9108255.
- 102. Qin M, Cao Q, Zheng S, Tian Y, Zhang H, Xie J, et al. Discovery of [1,2,4]Triazolo[4,3- a]pyridines as Potent Inhibitors Targeting the Programmed Cell Death-1/Programmed Cell Death-Ligand 1 Interaction. J Med Chem. 2019;62(9):4703–15. Epub 20190419. pmid:30964291
- 103. Basu S, Yang J, Xu B, Magiera-Mularz K, Skalniak L, Musielak B, et al. Design, Synthesis, Evaluation, and Structural Studies of C(2)-Symmetric Small Molecule Inhibitors of Programmed Cell Death-1/Programmed Death-Ligand 1 Protein-Protein Interaction. J Med Chem. 2019;62(15):7250–63. Epub 20190725. pmid:31298541.
- 104. Kotraiah V, Phares TW, Browne CD, Pannucci J, Mansour M, Noe AR, et al. Novel Peptide-Based PD1 Immunomodulators Demonstrate Efficacy in Infectious Disease Vaccines and Therapeutics. Front Immunol. 2020;11:264. Epub 20200306. pmid:32210956; PubMed Central PMCID: PMC7068811.
- 105. Phares TW, Kotraiah V, Karunarathne DS, Huang J, Browne CD, Buontempo P, et al. A Peptide-Based PD1 Antagonist Enhances T-Cell Priming and Efficacy of a Prophylactic Malaria Vaccine and Promotes Survival in a Lethal Malaria Model. Front Immunol. 2020;11:1377. Epub 20200709. pmid:32733457; PubMed Central PMCID: PMC7363839.
- 106. Sobhani N, Tardiel-Cyril DR, Chai D, Generali D, Li JR, Vazquez-Perez J, et al. Artificial intelligence-powered discovery of small molecules inhibiting CTLA-4 in cancer. BJC Rep. 2024;2. Epub 20240123. pmid:38312352; PubMed Central PMCID: PMC10838660.
- 107. Valizadeh M, Raoofian R, Homayoonfar A, Hajati E, Pourfathollah AA. MARCH-I: A negative regulator of dendritic cell maturation. Exp Cell Res. 2024;436(1):113946. Epub 20240207. pmid:38331309.
- 108. Li Z, Ju X, Silveira PA, Abadir E, Hsu WH, Hart DNJ, et al. CD83: Activation Marker for Antigen Presenting Cells and Its Therapeutic Potential. Front Immunol. 2019;10:1312. Epub 20190607. pmid:31231400; PubMed Central PMCID: PMC6568190.
- 109. Schneider BJ, Naidoo J, Santomasso BD, Lacchetti C, Adkins S, Anadkat M, et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J Clin Oncol. 2021;39(36):4073–126. Epub 20211101. pmid:34724392.
- 110. Mathew D, Marmarelis ME, Foley C, Bauml JM, Ye D, Ghinnagow R, et al. Combined JAK inhibition and PD-1 immunotherapy for non-small cell lung cancer patients. Science. 2024;384(6702):eadf1329. Epub 20240621. pmid:38900877.