Skip to main content

Malaria therapeutics: are we close enough?

Abstract

Malaria is a vector-borne parasitic disease caused by the apicomplexan protozoan parasite Plasmodium. Malaria is a significant health problem and the leading cause of socioeconomic losses in developing countries. WHO approved several antimalarials in the last 2 decades, but the growing resistance against the available drugs has worsened the scenario. Drug resistance and diversity among Plasmodium strains hinder the path of eradicating malaria leading to the use of new technologies and strategies to develop effective vaccines and drugs. A timely and accurate diagnosis is crucial for any disease, including malaria. The available diagnostic methods for malaria include microscopy, RDT, PCR, and non-invasive diagnosis. Recently, there have been several developments in detecting malaria, with improvements leading to achieving an accurate, quick, cost-effective, and non-invasive diagnostic tool for malaria. Several vaccine candidates with new methods and antigens are under investigation and moving forward to be considered for clinical trials. This article concisely reviews basic malaria biology, the parasite's life cycle, approved drugs, vaccine candidates, and available diagnostic approaches. It emphasizes new avenues of therapeutics for malaria.

Graphical Abstract

Background

Malaria is one of the most severe and life-threatening diseases. It is a mosquito-transmitted infectious disease and a major global health issue in tropical and subtropical regions. The mortality rate of malaria is very high compared to other protozoan diseases. As per the World Health Organization (WHO) 2020 reports, 241 million malaria cases were recorded worldwide; 627,000 people died from malaria. WHO announced the Global technical strategy (GTS) 2016–2030 to eradicate malaria by reducing malaria case incidence and mortality rates by at least 90% [1]. Malaria elimination depends on (i) preventive measures (including vaccination) and vector control, (ii) a sensitive diagnostic technique, and (iii) proper treatment of malaria infection on time [2]. Malaria is categorized as (i) asymptomatic malaria (caused by most of the Plasmodium species; infected individuals exhibit no symptoms or clinical signs), (ii) uncomplicated malaria (caused by human infecting Plasmodium species; symptoms include fever, moderate to severe body shaking, chills, sweating, anemia, vomiting, and nausea), and (iii) severe malaria (mainly caused by Plasmodium falciparum; symptoms include severe anemia, multiple organ failure, coma in case of cerebral malaria, pulmonary complications, acute kidney-associated injury, blood coagulation problems, metabolic acidosis, high temperature of 39 to 41 ºC, polyuria, and myalgia). Malaria is often fatal when not diagnosed and treated in a timely manner [3].

Five different species of Plasmodium (P. falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesi) cause malaria infection in humans. The parasite is infective and motile to the vertebrate host, and the malaria parasite life cycle involves two hosts (digenetic): human (intermediate host) and female Anopheles mosquito (definitive host) [3]. When an Anopheles mosquito bites a healthy human being, it injects sporozoites (infective stage for humans) while sucking the blood meal. These sporozoites are transferred to the hepatic cells (in the liver) through the blood circulatory system. These sporozoites mature into merozoites (exo-erythrocytic cycle) inside the hepatic cells. They are released into the blood vessel and invade the erythrocytes in which they grow and re-invade the fresh red blood cells (RBCs) for the completion of the erythrocytic cycle (asexual stage) (Fig. 1) [3,4,5].

Fig. 1
figure 1

Life cycle of malaria parasite

In the erythrocytic cycle, merozoites convert into ring-stage trophozoites, further developing into mature trophozoites and then into schizonts, and this development cycle takes around 48 h for P. falciparum. Later, schizonts rupture and release 8 to 36 merozoites to invade the new RBCs and continue the cycle where some of the merozoites undergo sexual development and mature into the male and female gametocytes (infective stage for mosquito); this process is called gametocytogenesis [6]. These gametocytes are taken by mosquitos while sucking the blood meal. Inside the mosquito gut, the microgamete (male gamete) fuses with the macrogamete (female gamete) and produces a zygote (gametogenesis) [6]. The zygote matures into ookinetes, which takes approximately 24 h. The ookinetes then develop into oocysts; this maturation occurs between the epithelium and basal lamina of the mosquito gut. The oocysts grow and rupture, releasing sporozoites (formed by asexual replication). The sporozoites are released and migrate through the hemocoel; they invade and are stored in the salivary gland of mosquitoes (Fig. 1).

Malaria is one of the major global health problems and annually causes significant mortality, morbidity, and socioeconomic burden. In the last 20 years, the world has achieved enormous progress in eliminating malaria. Timely diagnosis and effective treatment can prevent human health from diseases, including malaria [7, 8]. Several methods, such as rapid diagnostic test (RDT), microscopy-based analysis, and thick and thin layer blood smear analysis, are available to diagnose malaria. RDT is an accurate, quick, and WHO-recommended approach for identifying persons with symptomatic malaria and high parasite counts, and it is also used outside health institutions (remote areas) where a sophisticated lab is not established for malaria testing [9]. The RDTs are very affordable and straightforward to use. There is no requirement for a trained person. The available malaria diagnostic methods are explained in this review article.

A therapeutic approach can be preventive or curing. A vaccine is the most trusted preventative measure to control many infectious diseases. A vaccine is vital in eliminating any disease because it provides sterile lifelong immunity [8]. WHO recently approved RTS,S vaccine to cure malaria. The RTS,S vaccine targets the P. falciparum circumsporozoite surface protein (PfCSP) and shows < 37% protection against the P. falciparum parasite in the third phase of clinical trials [6, 10]. Several malaria vaccines targeting various stages of the parasite’s life cycle have been tried, but vaccine development against the eukaryotic malarial parasite is still very challenging. This review also discusses the potential malarial vaccines [11]. In addition, the availability of effective drugs for any disease may lower the overall mortality rate. Commonly used antimalarials are chloroquine, amodiaquine, quinine, mefloquine, halofantrine, lumefantrine, primaquine, and atovaquone. These drugs provide first-line protection to malaria patients. Moreover, WHO recommended artemisinin-based combination therapies (ACTs) as the first- and second-line treatment for the uncomplicated P. falciparum and chloroquine-resistant P. vivax malaria [12, 13]. In the ACT approach, artemisinin is combined with its derivatives, such as artesunate, dihydro-artemisinin, and artemether, and given to malaria patients for their betterment. Unfortunately, the malaria parasites are becoming resistant to the existing anti-malarial drugs, so there is an urgent need to develop or identify new potential drug/s against the malaria parasite [9, 13, 14]. This review article touches upon all the aspects of malaria therapeutics, explaining available therapeutics (vaccine/s and drug/s), diagnostic methods, and possible advancement in the respective fields.

Antimalarial drugs and their targets

Malaria is a severe global health problem that can lead to death if not treated in a timely manner. A drug is a chemical substance (natural or synthetic) used to diagnose, prevent, treat, and cure a disease [13]. Several antimalarial drugs are used worldwide to treat and prevent malaria infection [15]. Antimalarials are divided into several classes based on quinoline (4 and 8 aminoquinolines), cinchona alkaloids, diaminopyrimidine, sulfonamides, tetracyclines, naphthoquinones, and sesquiterpenes [16, 17]. Antimalarials are used as prophylaxis for malaria, and most kill parasites in the infected RBCs. Unfortunately, mosquitoes are becoming resistant to most of the approved antimalarials. Many have severe side effects such as blurred vision, stomach upset, nausea, vomiting, insomnia, headache, loss of appetite, hair loss, and mood swings [18]. One of the focuses of this review paper is to list all the anti-malarials and their drug targets (Table 1 and Fig. 2).

Table 1 List of antimalarial drugs along with their drug targets and site of action
Fig. 2
figure 2

List of approved antimalarials

Primarily, drugs interact with biological macromolecules (protein/DNA/RNA) in the human body to alter the physiological function and produce the desired effect. Drugs perform specific acts by interfering with macromolecules and altering cellular or biochemical processes, often called ‘drug targets.’ The structure and biochemistry of the Plasmodium parasite were studied very well, leading to understanding and identifying potential drug targets to overcome malaria. Malarial drug targets are classified based on their location in the parasite (e.g. food vacuole, apicoplast, cytosol, membrane, and mitochondria) and their mechanism (e.g. heme polymerization, pyrimidines de novo synthesis, DNA/protein synthesis, TCA cycle, ETC pathway, membrane transport, and hemoglobin hydrolysis). The essentiality of any protein in parasite development makes them potential drug targets [40, 41]. Table 2 and Fig. 3 collect all the reported drug targets for malaria disease.

Table 2 List of potential antimalarial targets
Fig. 3
figure 3

Antimalarials targeting the parasite’s life cycle

Malaria vaccine

The vaccine as a preventive measure or to generate herd immunity in the community is essential in eradicating any disease. Some WHO-approved or under clinical trial vaccines for protozoan diseases are Leish-F1 and ChAd63-KH vaccine (human leishmaniasis), Leishmune and CaniLeish (canine leishmaniasis), s48 vaccine (toxoplasmosis in sheep), Mosquirix or RTS,S (malaria) [106, 107]. RTS,S is the only malaria vaccine approved by WHO for its pilot implementation in the malaria-endemic regions. Despite several efforts, RTS,S could generate only 50% protection and a subsequent decline in efficacy after a four-dose regimen. Therefore, there is an urgent need to develop an improved, efficient, and potential vaccine against malaria. The increase in cases or deaths and drug-resistant strains is dismaying and demands a safe and immunogenic vaccine against malaria.

It has been more than 5 decades since Nussenzweig et al. used irradiated Plasmodium berghei sporozoite to immunize mice in their study and found that mice were protected when challenged with infectious sporozoite [108, 109]. However, mice were infected when challenged with blood-stage parasites, meaning only stage-specific protection was generated. An important lesson concluded from this study was to explore the parasite’s proteome, select essential protein/s from different stages of the Plasmodium’s life cycle, and develop stage-specific vaccine candidates to eradicate malaria worldwide. Stage-specific malaria vaccine could be produced by targeting the life-cycle stages such as (i) pre-erythrocyte stage, (ii) erythrocyte or blood stage, and (iii) sexual or transmission-blocking vaccine (TBV) (Fig. 4) [110]. Keeping this in mind, several proteins or biomolecules present on the surface of sporozoite, [circumsporozoite protein (CSP), cell-traversal protein for ookinetes and sporozoite (CELTOS)], blood-stage merozoite [merozoite surface protein (MSP), apical membrane antigen 1 (AMA1)], gametocyte (Pfs230), and ookinete (Pfs25 or Pvs25) have been characterized [111]. For instance, the identification of the CSP and its characterization has created a path ahead for malaria vaccine development [112]. CSP found abundantly on the sporozoite surface is the most potent candidate due to its high immunogenicity. It has a central repeat sequence [Asn-Ala-Asn-Pro (NANP) in P. falciparum], N-terminal region I, and a C-terminal region II which show similarity to thrombospondin and other adhesive proteins (Fig. 5) [113]. However, there is a distinct difference (sequence heterogenicity) between the repeat region of P. vivax VK210 (Type 1) and VK247 (Type 2) strains [114]. A chimeric construct targeting conserved C-terminal and repeat regions from these diverge strains would be suitable against pre-erythrocyte stage infection and prevent the hypnozoite dormant stage of P. vivax [115]. A junction between N-terminal and central repeats was recognized by antibodies specific to NANP repeats. The dual-capacity antibody can bind to the junction region and the NANP region and is characterized as the most potent among other antibodies against PfCSP [116]. Several vaccines have been designed and developed taking CSP as an antigen and tested in mice models [117]. A trend of subunit vaccines has been set employing NANP repeats and the C terminal region of CSP antigen for a malaria vaccine [118]. Further in this review, we will discuss the emerging candidates for the malaria vaccine (Table 3 and Fig. 4).

Fig. 4
figure 4

An overview of malaria vaccines and their mechanism. a Target stage (sporozoite, merozoites or gametocytes and ookinete). b Vaccine type (whole parasite vaccines, subunit vaccines, recombinant DNA/RNA vaccines) and target antigens. c Immune cell and response (cell-mediated or humoral antibody response). d Mode of action (killing of infected cell or cell invasion inhibition) of vaccine. (e) Vaccine name/s. DNA, deoxyribonucleic acid; RNA, ribonucleic acid

Fig. 5
figure 5

Schematic representation of (a) CSP region I and region II at N-terminal and C-terminal, respectively, and a central repeat region consists of NANP amino acid repeats. The junction region joins central repeat region to N-terminal and a signal sequence and GPI anchor sequence at N-terminal and C-terminal end, respectively. Central repeats (NANP) and junction region induce antibody generation while CD4 and CD8 T-cell epitopes at C-terminal trigger cell-mediated immune response. b RTS,S vaccine consists of B-cell epitopes from central repeat region at N-terminal and T-cell epitopes from region II of CS protein fused with hepatitis surface antigen (HBsAg) at C-terminal along with three copies of HBsAg (not in fusion with CS protein). c Yeast cell producing VLP (RTS,S and R21 vaccines) expressing CSP antigen on surface fused with hepatitis B surface antigen (HBsAg). GPI, glycosylphosphatidylinositol; CD, clusters of differentiation; CSP, circumsporozoite protein; VLP, virus-like particle

Table 3 List of malarial vaccines (approved or under clinical trial)

Whole parasite vaccine

After Nussenwieig’s research, several studies were performed by taking the whole parasite to develop a malaria vaccine using malaria-infected mosquitoes, irradiated by gamma radiation and allowed to feed on non-human primates and human subjects [108]. The idea was to mimic the natural infection. Complete protection was observed for weeks and months when challenged with infectious sporozoite or controlled human malaria infection (CHMI). Sanaria P. falciparum sporozoite (PfSPZ), a fast-track designated vaccine, has shown promising efficacy during its first and second clinical phase trial and is likely to move into the phase IIb/III trial. This non-replicating whole parasite was purified from irradiated mosquito's salivary glands and injected intravenously [119]. High-level protection was obtained against malaria up to 14 months after final immunization at a 9.0 × 105 sporozoites dose when administered intravenously [120]. PfSPZ or whole parasite vaccine efficacy depends upon the number of sporozoites and dose regimen and the route of vaccine administration. Another method employed to use the live parasite is to inject PfSPZ along with anti-malarial drugs, e.g. chloroquine or pyrimethamine (termed as chemoprophylaxis vaccine PfSPZ-CVac), which kills the parasite and checks the intra-erythrocyte development cycle [121]. However, it does not affect the parasite’s liver stage development and continues to produce sporozoite, which triggers the immune response without causing the disease.

Since the PfSPZ-CVac has live sporozoite to induce an immune response, an increased fold of protection can be generated through this method compared with radiation attenuated sporozoite [122]. In parallel, PfSPZ-GA1, a genetically attenuated parasite (GAP), is a candidate vaccine (phase I/IIa) in which attenuation was obtained through genetic modification in the parasite genome [123]. GAP showcases all the sporozoite surface protein but fails to produce merozoites and prevent progression to the blood stage. In genetically attenuated vaccines, the modifications are being done by deletion of essential genes(s) (single, double, or triple gene knockout), resulting in loss of function or gain of function, or by overexpression of immunogenic protein or toxins (referred to as a suicidal parasite) [123,124,125]. Several researchers are trying to produce chimeric parasites that express the surface immunogen but cannot perform invasion activity. For example, the P. falciparum parasite expressing P. vivax CSP is a partial functional replacement of CSP and could be an excellent choice for whole parasite vaccine candidate as this chimeric parasite fails to generate infection [126]. The whole parasite or attenuated vaccine is always questioned for safety concerns due to incomplete attenuation and chances of revival of the parasite. Another major problem associated with the whole-parasite vaccine is the production of attenuated sporozoites (irradiated or genetically modified) and their storage and transportation, especially to the remote area of Saharan countries.

Subunit vaccine

Subunit vaccine is the safest method of immunization with the most negligible probability of toxicity and reactogenicity. Also, the scale-up process of recombinant antigens is much easier than large-scale production and maintenance of attenuated parasites, making subunit vaccines a better choice for community immunization. Here are some examples of subunit vaccines for malaria, which are broadly categorized based on their target at various developmental stages of the parasite.

Pre-erythrocyte stage vaccine

RTS,S (Mosquirix), the first WHO-approved malaria vaccine, has set a milestone in WHO’s malaria eradication roadmap effort [127]. Based on virus-like particle (VLP) technology, RTS,S is a subunit vaccine having CSP antigen (NANP repeats and C-term region for P. falciparum vaccine) fused with hepatitis B surface antigen (HBsAg) on VLPs (Fig. 5) [128,129,130]. Four doses of RTS,S elicited short-lived protection that varied with different age groups and parasite strains [110]. R21/Matrix M, a subunit vaccine, has shown an increased efficacy compared with RTS,S in its second phase of clinical trials, employing the VLP technique and CSP antigen similar to RTS,S [131]. Unlike RTS,S, R21 does not contain hepatitis B surface antigen (HBsAg) in a separate form. HBsAg is expressed as a fusion protein with CSP antigen. In response to this fusion protein present on VLPs, most antibodies will be produced against the CSP and not against the HBsAg. This makes R21 (Rv21, P. vivax vaccine candidate) a promising vaccine candidate for malaria in the coming year [131]. However, high efficacy vaccine against malaria remains a challenge.

Various adjuvants have been tested with the CSP-based malaria vaccine for their efficacy, including AS02A, an oil-in-water-based adjuvant, and a polymeric glyco adjuvant p (Man-TLR7) conjugate with CSP, adenovirus 35/26 expressing CSP. The approved version of RTS,S vaccine for malaria, employed adjuvant system 01 (AS01) consisting of MPL A, a TLR4 agonist that induces biased Th1 response, and QS21, a highly purified saponin that does not work through only one such PRR or signaling cascade; instead, they enhanced antigen uptake and induced a strong Th1 and Th2 response [117, 132,133,134,135]. R21 formulated with different adjuvant Matrix M, a saponin-based adjuvant, has achieved 77% efficacy in clinical trials.

Erythrocyte or blood stage vaccine

MSPs present on the surface of erythrocytes infecting merozoites are the most studied blood-stage antigen(s) for drug and vaccine development among all Plasmodium species. This includes MSP1, MSP2, MSP3, MSP4, MSP8, and MSP10, characterized in parallel for vaccine development [136, 137]. MSP1, the prime target of antibody response of naturally acquired immunity during the parasite's blood stage, is often considered for the blood-stage vaccine. MSP1 is a high-molecular-weight protein (185 kDa) with multiple proteolytic cleavage sites. A C-terminal fragment of 42 kDa is found to be immunogenic and cleaves again into 33 kDa and 19 kDa fragments during invasion [138]. This 42 kDa (19 kDa fragment) fragment of MSP1 alone or in combination with other merozoite antigens (AMA1) was tested in clinical trials [136, 139]. Likewise, PvMSP1 (42 kDa) fused with PvMSP8 has been recently tested as a vaccine candidate against P. vivax infection [138]. Controlled blood-stage human malaria infection using inoculum of parasitized blood is a new approach to developing P. vivax vaccine. Currently, vaccine candidates targeting blood-stage which are under trials include ChAd63-MVA RH5 and MSP3-CRM-Vac4All.

Other vaccine candidates based on merozoite antigens for inhibition of erythrocyte invasion or clinical symptom progression are AMA1, RH5, SERA5, and PvDBP (P. vivax duffy binding protein), which measurably fail to generate a protective immune response [140]. Reticulocyte binding protein homolog 5 (PfRH5) can induce antibody response, which can inhibit parasite growth more efficiently than antibody response by PfMSP1 and PfAMA1, suggesting the critical role of RH5 in parasite growth and survival [11]. An effective delivery system and TLR-based adjuvants are required to enhance the immunogenicity of these polymorphic antigens. P27A is one such vaccine candidate that showed good immunogenicity in its first clinical trial and needed to be improved by considering an immunogenic delivery system [141].

Transmission blocking vaccine (TBV)

Candidates for TBV or sexual stage vaccine are Pfs25M-EPA/AS01 (Phase I) and Pfs230D1M-EPA/AS01 (Phase II). The parasite's blood stage is linked to causing symptomatic/clinical malaria and disease transmission through the transfer of gametocytes from an infected human to vector Anopheles. In parallel with pre-erythrocyte and erythrocyte stage-based vaccines, sexual-stage or gametocyte surface antigen-targeting vaccines are used to treat malaria [142]. Targeting the sexual stage or gametocyte antigen prevents ookinete maturation and sporozoite development, release, and transmission. Still, it does not stop malaria symptoms in infected individuals. The antigens that are being targeted are ookinete surface protein Pfs25, a male gametocyte protein P48/45 (P. vivax Pvs48/45 and Pvs47), Pfs47, and gametocyte antigen Pf230 [143,144,145,146]. Cell-transversal protein for ookinete and sporozoite (CELTOS), an anticipated vaccine candidate required for ookinete transversal and sporozoite infection, has shown increased immunogenicity when adjuvanted with CpG or poly IC or both [147]; 25 kDa ookinete surface protein (Pfs25) fused with a complement inhibitor C4b-binding protein IMX313 encoded by ChAd63 and modified vaccinia virus Ankara (MVA) viral vector is a recombinant DNA or vectored vaccine candidate for transmission blocking [143]. Recombinant IMX313 protein acts as a carrier by creating heptamer with antigen and generating a solid antibody response [143]. Antibodies that respond to these antigens have been tested for blocking activity through a standard membrane-feeding assay (SMFA). A certain antibody titer level is required to secure the parasite's sexual stage progression in mosquitos. Pfs47 sexual stage antigen display on Acinetobacter Phage AP205 VLP elicited a robust transmission reducing activity (TRA) by antibody at a 5 µg/ml concentration purified from immunized mice [145]. In addition, considering mosquito midgut protein anopheline alanyl aminopeptidase N (AnAPN1) critical for traversal of parasite ookinete in mosquito midgut can be a proven potent strategy for transmission-blocking activity. A second-generation AnAPN1 vaccine construct containing crucial peptide epitopes adjuvanted with glucopyranosyl lipid adjuvant and saponin QS21 in liposomal formulation elicited antibody production [148].

Recombinant DNA or viral vectored vaccine

DNA or vectored vaccine, ChAd63 MVA ME-TRAP (phase II), is a current vaccine technology to present intracellular antigens and induces a strong CD8 + mediated immune response with pro-inflammatory cytokine production required against malaria infection. Chimpanzee Adenovirus 63 (ChAd63) and MVA, a non-replicating viral vector encoding different malaria proteins, includes 25 kDa ookinete protein Pfs25, RH5, PvDBP, CSP, and multiepitope chain of TRAP (ME-TRAP) [143, 149,150,151]. Self-amplifying RNA vaccine is a novel vaccine technology that introduces an mRNA construct encoding an antigen Plasmodium macrophage migration inhibitory factor, a Plasmodium protein that can quench the host pro-inflammatory cytokines, and a replication machine for self-amplification [152]. This novel self-amplifying RNA vaccine will minimize the number of doses and reduce the declination of antibody titer over a while. Table 3 summarizes the malarial vaccines, approved or under clinical trial or investigation, along with their vaccine type, target antigen, and mode of action.

Vaccine delivery, antigen uptake, and accurate antigen presentation are crucial for a vaccine's efficiency. A self-assembling protein nanoparticle is a current strategy to present Th or Tc cell epitopes of CSP or other blood and sexual stage proteins [153, 154]. Synthetic or inorganic nanoparticles can be proved to be a safe and novel approach to delivering or presenting antigens without any negative impact in the murine model [155, 156]. A carrier protein is sometimes required to particulate the antigen in nano size and simultaneously works as an adjuvant. Exo-protein A from Pseudomonas aeruginosa and IMX313, a homolog of human complement four binding protein (C4bp), were tested for their reactogenicity and immunogenicity with different vaccine candidates. At the same time, Advax (delta inulin polysaccharide), a co-adjuvant with poly (I:C), increases the half-life of the antigen, resulting in persisting immune response [136, 143, 157].

Diagnosis

According to National Institutes of Health, diagnosis is a complex process to identify a disease, illness, or injury by examining the signs and symptoms and comparing them to an existing set of categories that define a particular condition, as the medical profession agrees. Diagnosis is the distinguishing of a diseased condition from health, and it leads to the appropriate treatment and prognosis [158]. Diagnosis occurs at three levels: first, where a class of disease is determined (such as a cardiac disorder); second, the subject to be diagnosed is particularized (such as a 45-year-old male); third, a specific reasoned categorization is made (such as coronary artery disease) [158].

The detection of malaria is essential at the initial stage. Otherwise, the disease might develop severe complications, especially P. falciparum infections, which may be fatal [159]. The review paper explains the available diagnostic methods, such as RDT, polymerase chain reaction (PCR), and microscopy (Fig. 6), to detect malaria parasite infection and their recent advancements.

Fig. 6
figure 6

Diagnosis methods for malaria detection using (a) PCR. (b) Use of microscopy in malaria diagnosis: A blood sample is taken by pricking the finger, and two types of smears can be prepared: thick (for the presence of Plasmodium) and thin (for identification of species of Plasmodium). The figure shows how the trophozoite stage is visualized in thick smear and thin smear. c Flow cytometry in malaria diagnosis: Fluorochrome staining and analysis by flow cytometry. d Rapid diagnostic test (RDT) for malaria: Cassette and interpretation of the assay results as positive, negative, or invalid. (e) Diagnosis by RDTs using samples other than blood such as saliva and urine. PCR, polymerase chain reaction

Diagnostic methods currently in use

Clinical diagnosis

This type of malaria diagnosis is based on the symptoms displayed by the infected individual. It is used for diagnosis in case of the unavailability of laboratory facilities or self-diagnosis [160, 161]. Like malaria, many diseases cause symptoms such as fever, headache, fatigue, and anemia at later stages. In ancient times, people found it clinically challenging to distinguish malaria from other fevers. Significantly, the two conditions (malaria and typhoid) were most likely to be confused [162]. Self-diagnosis and self-treatment are prone to errors [163]. Therefore, clinically suspected individuals should constantly be tested using the diagnostic tools available at the hospitals.

Microscopy

A famous German chemist and bacteriologist, Gustav Giemsa, introduced a mixture of methylene blue and eosin in 1904, after which Giemsa staining, followed by imaging, was used for malaria diagnosis [164]. Although advanced diagnostic methods and automatic devices are being developed, the microscopic examination of blood films remains the gold standard method.

Eosin and methylene blue are the two main components that make up the Giemsa staining solution from which eosin stains the parasite’s nucleus red. In contrast, methylene blue causes the cytoplasm to appear blue colored [165]. A thick blood film stained using Giemsa stain is usually used to determine the presence of parasites, whereas a Giemsa-stained thin blood film helps identify the species under light microscopy. Approximately 50 times more blood is examined in a thick blood film than in a thin blood film [165]. However, while staining, 60–80% of parasites may be lost in the case of Giemsa-stained thick blood film [166]. The inexpensive Giemsa microscopy differentiates the Plasmodium species and quantifies the parasites.

Since mature erythrocytes do not contain DNA or RNA, whereas the parasites do, fluorescent dyes such as acridine orange are used to detect the Plasmodium parasites. For this, the patient’s blood sample is incubated with acridine orange, staining the DNA and RNA of different developmental stages of Plasmodium [167, 168]. The fluorescent parasites are then observed using a conventional fluorescence microscope or a fluorescence microscope based on LED [167, 168]. A fluorescent microscope with an interference filter was developed to image the thin blood films stained with acridine orange [169]. Although this method is feasible, trained personnel must correctly label the patient’s blood sample and have the expertise to read the slides.

Rapid diagnostic test

RDT is a device that detects the malaria parasite and proves to be an important alternative in cases where there is a lack of a microscope or time to scan the blood films [170]. It is a simple and fast diagnostic tool that detects the parasite in a small amount of blood sample (5–15 µl) by immunochromatographic assay, which involves monoclonal antibodies against the parasite’s antigen [171]. Currently, the available RDTs detect HRP2 (histidine-rich protein), pLDH (parasite lactate dehydrogenase), and aldolase [170]. They are more than 95% sensitive in P. falciparum infections, but this sensitivity level has not yet been achieved for non-P. falciparum infections [171]. The RDTs are available in different formats, such as dipsticks, cards, and cassettes [172].

Dipsticks have been used worldwide to detect malaria antigens in the blood. These rapid immuno-chromatographic tests (based on detecting the circulating antigens) are specific to the parasite with the help of specific antibodies attached to a membrane [173]. Dipstick tests were initially used to detect P. falciparum infections only as they target HRP2 expressed by P. falciparum during the trophozoite stage [174, 175]. Examples include Parasight-F, ICT Malaria P.f., and PATH Falciparum Malaria IC Strip [176]. However, newer tests, such as the OptiMAL assay and the ICT Malaria P.f/P.v assay, can detect P. falciparum and P. vivax infections. Both tests can also differentiate between the two species. The OptiMAL assay is based on pLDH detection, and these two parasitic species show antigenic differences in their pLDH isoenzymes [177]. The ICT Malaria P.f/P.v assay targets HRP2 of P. falciparum and P. vivax [178]. The dipsticks are simple to use and handle. They can be used for malaria self-testing, so travelers and tourists visiting malaria-endemic regions are advised to carry them [173]. In febrile cases, they can use these test kits when they cannot reach adequate professional help in time [179].

Cards and cassettes are safer to use than dipsticks as they can prevent blood contamination, but the disadvantage lies in their cost, which is 40% higher than for dipsticks. Another issue is that they are more time-consuming than dipsticks. Most available cards and cassettes target two antigens: HRP II/pLDH, HRP II/pan pLDH, or HRP II/pan aldolase [172].

Polymerase chain reaction

PCR-based tests have improved the limit for detection of malaria infection with < 0.02 parasites/µl [180, 181]. Although PCR detects cases with low parasitemia, it has been observed that it may miss some cases even with high parasitemia [182]. The PCR uses thermostable DNA polymerases of bacterial origin and amplifies even tiny fragments of DNA by using different temperatures at different stages of the cycle. Several PCR approaches are used to detect Plasmodium infection.

Nested PCR involves two consecutive rounds of amplification with two sets of primers. The first amplification product is used as the template for the second round, in which species-specific primers are used [183, 184]. For the first reaction, the primers rPLU1 and rPLU5 are used for the amplification of the genomic DNA of Plasmodium; for detecting P. falciparum, the products of the first reaction are then amplified using rFAL1 and rFAL2, rVIV1 and rVIV2 for P. vivax, rOVA1 and rOVA2 for P. ovale, and rMAL1 and rMAL2 for P. malariae [185]. Furthermore, the PCR products are separated by running agarose gel electrophoresis and stained with ethidium bromide, followed by visualization under UV light to check which lanes contain products positive for malaria [186].

Real-time PCR or qPCR is used for real-time observation of the replication and amplification process [184]. Fluorescent labels such as SYBR green, sequence-specific oligonucleotide probes, and photo-induced electron transfer fluorogenic primers are used to monitor the amplicon formation [187,188,189]. This is based on the principle that their fluorescence intensity is closely related to the number of amplification products [184].

Direct PCR assays are also available for Plasmodium detection. In most forms of PCR, DNA extraction from blood samples is a crucial step, but direct PCR bypasses DNA extraction [190]. Therefore, the time, cost, and labor required to get DNA is reduced, but this procedure might miss the asymptomatic infections because of relatively low parasitemia [184]. Phusion blood direct PCR kit (Thermo Scientific, Waltham, MA) has been used to perform direct PCR using dried blood spots as the sample [191].

Reverse-transcriptase PCR allows the targeting of expressed RNA sequence instead of the gene, which enables the determination of Plasmodium in its specific stages [184]. A real-time reverse-transcriptase PCR was developed to detect Plasmodium by amplifying the RNA and DNA of 18S rRNA genes [192]. It can detect infections with parasitemia as low as 0.002 parasite/μl [184].

Flow cytometry

During the life cycle of Plasmodium, the parasites invade the RBCs and further grow and multiply in these cells. Therefore, these stages cause clinical symptoms and are the targets for various drugs [193]. Also, the detection of the presence of Plasmodium in the blood is used to diagnose malaria for which flow cytometry has proved useful [193]. The analysis of the development of the blood stages by flow cytometry is reproducible and rapid [193, 194]. Flow cytometry is performed using fluorescent dyes specific to nucleic acids since RBCs do not contain DNA. Any DNA-specific fluorescence detected in the RBC population results from the fluorescent dyes bound to Plasmodium DNA [193]. Therefore, infected cells can be differentiated from non-infected cells, and this method can even be used to determine the parasite’s developmental stage. As the plasmodia multiply in the RBCs, the stained parasites' fluorescence intensity increases during their development [193]. It is a sophisticated approach to diagnosing malaria, but the equipment is expensive and requires trained personnel for operation and maintenance, which would affect the accuracy severely [170].

Automated blood cell analyzers

These can detect parasites at low parasitemia levels, such as 5–20 parasites/µl of blood, but light microscopy, if performed by an experienced pathologist, might detect even lower parasitemia levels [195]. Therefore, the automated blood analyzer is not appropriate as a screening test. Still, it plays a role in detecting additional cases, such as those with no clinical suspicion that lead to a specific request for a malaria test [196]. For determining the species of Plasmodium and parasitemia, microscopy is required as the automated blood cell analyzer, Abbott Cell-Dyn 3500, only lets one know about the presence of abnormal monocyte and neutrophil cell populations [197]. The instrument’s sensitivity is based on pigmentation. Therefore, early infections might not get detected because of the low abundance of malaria pigment in the initial stage [198]. New models with higher sensitivity for malaria detection have been developed [199].

Serological detection

The antibodies to Plasmodium may persist for months once they appear after the erythrocytes are invaded by the parasite [196]. This can be used to diagnose the presence of Plasmodium in the serum of the patients. The immunofluorescence antibody test has been used to detect Plasmodium-specific antibodies in serum samples [200]. The serum sample is applied to a slide on which Plasmodium antigen was prepared and stored at –30 ºC, followed by a quantitative result using fluorescence microscopy to determine the amount of IgG and IgM [200]. Enzyme-linked immunosorbent assay can also be used for antibody detection [201]. Although these two techniques are simple, they require more time and trained personnel [170].

Non-invasive diagnosis

Samples other than blood, such as body fluids (urine and saliva) or fecal matter or hair, represent an alternative as they are obtained without invasion, thus avoiding the pain associated with invasive procedures and the consequences of the social and cultural beliefs related to blood sampling, leading to an increase in the participation in mass screening programs [202, 203]. Techniques like PCR, immunoassay, microfluidics, and immunochromatography are used for Plasmodium detection in such samples [170].

Recent developments in malaria diagnosis

Malaria detection is a crucial step for proper and timely treatment. Moreover, the two most commonly used malaria diagnostic approaches, microscopy and RDT, can diagnose symptomatic infections but are not sensitive enough to detect low-parasitemia asymptomatic infections. This means that even if asymptomatic people reach the health centers for diagnostic examination, they will probably remain undiagnosed. The PCR can detect disease with lower parasitemia, but those with very low density cannot be detected (Fig. 7) [204]. Several recently developed and emerging techniques seem promising. However, they still have a few limitations regarding detecting certain parasite stages, and discrimination between different stages is also a concern [205]. For example, the detection of dormant stages or hypnozoites of P. vivax and P. ovale cannot be done with the currently available tools [205]. Among the recently developed techniques for malaria diagnosis, one is loop-mediated isothermal amplification, a molecular approach based on the amplification of nucleic acid during which specific genes are converted to loops so that continuous amplification can occur (Fig. 8) [205, 206]. Gazelle is a new device for malaria diagnosis based on detecting malaria pigment, hemozoin. It quickly detects hemozoin particles in the blood sample and is cost-effective (Fig. 8) [207]. Since hemozoin contains iron components, the magnetic field aligns the particles. An internal light source shines a light on the sample, and the measurement is made of the amount of transmitted light in both the presence and absence of a magnetic field [207]. In its absence, the hemozoin particles become randomly oriented because of Brownian motion, while in a magnetic field, the particles become aligned and block light transmission [207]. Since all five species produce hemozoin, this device allows the detection of malaria caused by all five species. A non-invasive, rapid technique based on near infrared spectroscopy has been developed recently, which can diagnose malaria due to P. falciparum and P. vivax through the skin of malaria patients. It uses only a hand-held spectrometer, thus making it a reagent-free technique; this miniature spectrometer is used to shine near-infrared light on the ear, arm, or finger of the individual, and spectra are generated, which are then used for making predictions using machine learning algorithms. The study in Brazil showed 92% accuracy for the arm and 93% predictive accuracy for differentiating between P. falciparum and P. vivax. The bands observed in the spectra enable the identification of positive and negative malaria cases and are mainly due to hemozoin [208].

Fig. 7
figure 7

The range of malaria infection and a comparison of the sensitivity of three diagnostic methods: microscopy, RDT, and PCR. Microscopy and RDTs are sensitive enough to detect symptomatic infections but not those with low parasite density. PCR is more sensitive than the other two techniques (RDT and microscopy) but is unable to detect infections with very low parasitemia. PCR, polymerase chain reaction; RDT, rapid diagnostic test

Fig. 8
figure 8

Future tools for malaria diagnosis. a Gazelle device and its mechanism based on magneto-optical detection. b TMek: The iRBCs and hemozoin crystals get captured on the cylindrical Ni concentrators of microchip and the healthy RBCs are sedimented. (c) LAMP for malaria diagnosis: Procedure for DNA extraction and LAMP assay. TMek, Tid Mekii; iRBC, infected red blood cell; LAMP, loop-mediated isothermal amplification; DNA, deoxyribonucleic acid

Anticipated future developments

Many efforts are being made worldwide to understand the parasite Plasmodium and the Anopheles mosquito to control malaria successfully. The massive number of malaria cases and deaths distress the African region, tropical countries, and many others. Over the last century, efforts have been made to eradicate malaria globally. However, there are several roadblocks to eliminating malaria, including our understanding of the biology of the malaria parasites, the complex life cycle, and the parasite’s immune evasion. Simultaneously, the parasite has evolved drug resistance against most of the available antimalarial drugs, including WHO-recommended ACT. There is an urgent need to find novel antimalarials with the help of the latest strategy such as computer-aided drug design, fragment-based drug design, high-throughput screening, and drug repurposing approach. Apart from drug development, it has increased the need for an effective vaccine to eliminate malaria. No malaria vaccine can generate 100% sterile protection to achieve WHO's 90% malaria eradication goal by 2030. However, with low efficacy and subsequent antibody decline with time, the world has its first malaria vaccine, RTS,S (Mosquirix).

Following RTS,S, its second-generation vaccine R21 with improved efficacy is also moving forward quickly. The major obstacle in developing immunogenic malaria vaccines is the parasite's multi-stage life cycle, antigenic variation and polymorphisms, limited choice of immunogen, and vaccine candidate evaluation. Efforts can be made to remove these obstacles, and further steps should be taken to improve the efficacy of the existing RTS,S vaccine. This can be done by introducing better adjuvants and new technologies to present more and more antigens to the host immune system. Qubevirus durum bacteriophage and SpyTag/SpyCatcher system are a few to multimerize the antigen on the surface of a VLP [209, 210]. Bacterial vector vaccines expressing pathogen proteins and antigen-presenting cells are new tools under investigation that can be considered for malaria. However, it is too early to know the efficiency of these vaccine technologies [211]. Multiple anticipated vaccine candidates still need to be considered for improvement and newer technologies.

Apart from the treatment, a timely and accurate diagnosis of malaria is an important event that leads to saving humans from malaria disease. Malaria diagnosis is one of the effective strategies for disease management since it is curable if diagnosed promptly. Although many developments and newer techniques are continuously emerging, microscopy remains the gold standard. Several methods are better than microscopy in terms of accuracy or sensitivity. The methods discussed above have certain advantages and disadvantages, and it cannot be determined which is the best or most appropriate among them. It is unknown whether there is an ideal diagnostic method for malaria that is simple, accurate, quick, affordable, easy to handle, and painless. So, the search continues, and we have high hopes for new ideas and techniques. Currently, microscopy and RDTs are the most commonly used methods to diagnose malaria. Still, they will not be sufficient as the world moves towards malaria elimination, and newer techniques are required, enabling mass screening for asymptomatic infections, the surveillance of continued transmission, and the management of symptomatic infections [204, 212]. One such diagnostic technique that has been developed is known as TMek, a lab-on-a-chip diagnostic method that directly quantifies the level of parasitemia (Fig. 8) [213]. It exploits the magnetic properties of hemozoin (malaria pigment) nanocrystals and provides the quantification value in 10 min [213]. Also, the laser-based non-invasive method of malaria detection is an excellent step in the pain-free detection of malaria. It may be developed into a handy instrument to screen large populations without a sophisticated setup, similar to the infrared thermometer.

Conclusion

Malaria is an infectious disease affecting people globally. Without prompt diagnosis and treatment, the condition can worsen, which is why proper diagnosis and treatment are essential. Overall, the progress in eliminating malaria globally has been satisfactory, and the number of malaria cases has declined [214]. Also, the available therapeutics in the form of drugs and vaccine have played a significant role in restraining malaria aftermaths. The presence of diagnostic approaches for malaria detection has also made it possible to reach the last corner of society. But still, diagnosis of asymptomatic malaria and removal of false-positive results even after a recovery has remained the bottleneck for accurate diagnosis for containing the spread of malaria. Moreover, the lack of a highly efficient vaccine has also hampered malaria infection preventive measures, especially in African regions. In addition, the growing concern about parasite resistance to the available medicines has worsened the scenario. Therefore, it is difficult to assess how close we are to the 2021 theme of malaria day, i.e. 'Zeroing in on malaria elimination.' Hence, we must align with the 2022 theme of malaria day by focusing on new and innovative approaches to reduce malaria's burden and save lives.

Availability of data and materials

Not applicable.

Abbreviations

ACT:

Artemisinin combination therapy

AMA:

Apical membrane antigen

CELTOS:

Cell traversal protein for ookinete and sporozoites

CHMI:

Controlled human malaria infection

CSP:

Circumsporozoite protein

GAP:

Genetically attenuated parasite

GTS:

Global technical strategy

HRP:

Histidine-rich protein

LAMP:

Loop-mediated isothermal amplification

MSP:

Merozoite surface protein

PCR:

Polymerase chain reaction

Pf:

Plasmodium falciparum

PfSPZ:

Plasmodium falciparum sporozoite

PfSPZ-CVac:

Plasmodium falciparum sporozoite chemoprophylaxis vaccine

Pv:

Plasmodium vivax

PvDBP:

Plasmodium vivax duffy binding protein

RBC:

Red blood cells

RDTs:

Rapid diagnostic tests

RH5:

Reticulocyte binding protein homolog 5

SMFA:

Standard membrane feeding assay

TBV:

Transmission blocking vaccine

TLR:

Toll-like receptor

VLP:

Virus-like particle

WHO:

World Health Organization

References

  1. Roche B, Broutin H, Simard F. Optimizing public health strategies in low-income countries: the challenges to apply the scientific knowledge for disease control and for which diseases. Oxford: Oxford University Press; 2018.

    Google Scholar 

  2. Animut A, Lindtjørn B. Use of epidemiological and entomological tools in the control and elimination of malaria in Ethiopia. Malar J. 2018;17:1–8.

    Article  Google Scholar 

  3. Makanjuola RO, Taylor-Robinson AW. Improving accuracy of malaria diagnosis in underserved rural and remote endemic areas of Sub-Saharan Africa: a call to develop multiplexing rapid diagnostic tests. Scientifica. 2020;2020:3901409.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Soni R, Sharma D, Bhatt TK. Plasmodium falciparum secretome in erythrocyte and beyond. Front Microbiol. 2016;7:194.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Borgheti-Cardoso LN, Anselmo MS, Lantero E, Lancelot A, Serrano JL, Hernández-Ainsa S, et al. Promising nanomaterials in the fight against malaria. J Mater Chem B. 2020;8:9428–48.

    Article  Google Scholar 

  6. Keleta Y, Ramelow J, Cui L, Li J. Molecular interactions between parasite and mosquito during midgut invasion as targets to block malaria transmission. NPJ Vaccines. 2021;6:140.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Kalantari P. The emerging role of pattern recognition receptors in the pathogenesis of malaria. Vaccines. 2018;6:13.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Mwakingwe-Omari A, Healy SA, Lane J, Cook DM, Kalhori S, Wyatt C, et al. Two chemoattenuated PfSPZ malaria vaccines induce sterile hepatic immunity. Nature. 2021;595:289–94.

    Article  CAS  PubMed  Google Scholar 

  9. Alonso PL, Tanner M. Public health challenges and prospects for malaria control and elimination. Nat Med. 2013;19:150–5.

    Article  CAS  PubMed  Google Scholar 

  10. Camponovo F, Ockenhouse CF, Lee C, Penny MA. Mass campaigns combining antimalarial drugs and anti-infective vaccines as seasonal interventions for malaria control, elimination and prevention of resurgence: a modelling study. BMC Infect Dis. 2019;19:1–15.

    Article  Google Scholar 

  11. Draper SJ, Sack BK, King CR, Nielsen CM, Rayner JC, Higgins MK, et al. Malaria vaccines: recent advances and new horizons. Cell Host Microbe. 2018;24:43–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Abay SM. Blocking malaria transmission to Anopheles mosquitoes using artemisinin derivatives and primaquine: a systematic review and meta-analysis. Parasit Vectors. 2013;6:278.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Talapko, Škrlec, Alebić, Jukić, Včev. Malaria: the past and the present. Microorganisms. 2019;7:179.

  14. Rahman K, Khan SU, Fahad S, Chang MX, Abbas A, Khan WU, et al. Nano-biotechnology: a new approach to treat and prevent malaria. Int J Nanomedicine. 2019;14:1401–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Alam A, Goyal M, Iqbal MS, Pal C, Dey S, Bindu S, et al. Novel antimalarial drug targets: hope for new antimalarial drugs. Expert Rev Clin Pharmacol. 2009;2:469–89.

    Article  CAS  PubMed  Google Scholar 

  16. Yadav N, Sharma C, Awasthi SK. Diversification in the synthesis of antimalarial trioxane and tetraoxane analogs. RSC Adv. 2014;4:5469–98.

    Article  CAS  Google Scholar 

  17. Chakraborti S, Chhibber-Goel J, Sharma A. Drug targeting of aminoacyl-tRNA synthetases in Anopheles species and Aedes aegypti that cause malaria and dengue. Parasit Vectors. 2021;14:1–11.

    Article  Google Scholar 

  18. Tse EG, Korsik M, Todd MH. The past, present and future of anti-malarial medicines. Malar J. 2019;18:1–21.

    Article  Google Scholar 

  19. White NJ. The treatment of malaria. N Engl J Med. 1996;335:800–6.

    Article  CAS  PubMed  Google Scholar 

  20. Abamecha A, Yilma D, Adissu W, Yewhalaw D, Abdissa A. Efficacy and safety of artemether–lumefantrine for treatment of uncomplicated Plasmodium falciparum malaria in Ethiopia: a systematic review and meta-analysis. Malar J. 2021;20:213.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Eckstein-Ludwig U, Webb RJ, van Goethem IDA, East JM, Lee AG, Kimura M, et al. Artemisinins target the SERCA of Plasmodium falciparum. Nature. 2003;424:957–61.

    Article  CAS  PubMed  Google Scholar 

  22. Nixon GL, Moss DM, Shone AE, Lalloo DG, Fisher N, O’Neill PM, et al. Antimalarial pharmacology and therapeutics of atovaquone. J Antimicrob Chemother. 2013;68:977–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Coban C. The host targeting effect of chloroquine in malaria. Curr Opin Immunol. 2020;66:98–107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Obonyo CO, Juma EA. Clindamycin plus quinine for treating uncomplicated falciparum malaria: a systematic review and meta-analysis. Malar J. 2012;11:1–11.

    Article  Google Scholar 

  25. Okada M, Guo P, Nalder S, Sigala PA. Doxycycline has distinct apicoplast-specific mechanisms of antimalarial activity. Life. 2020;9:e60246.

    CAS  Google Scholar 

  26. Nothdurft HD, Clemens R, Bock HL, Löscher T. Halofantrine: a new substance for treatment of multidrug-resistant malaria. J Clin Investig. 1993;71:69–73.

    Article  CAS  Google Scholar 

  27. Al-Bari MdAA. Chloroquine analogues in drug discovery: new directions of uses, mechanisms of actions and toxic manifestations from malaria to multifarious diseases. J Antimicrob Chemother. 2015;70:1608–21.

    Article  PubMed  Google Scholar 

  28. Bassat Q. The use of artemether-lumefantrine for the treatment of uncomplicated Plasmodium vivax malaria. PLOS Negl Trop Dis. 2011;5:e1325.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wong W, Bai X-C, Sleebs BE, Triglia T, Brown A, Thompson JK, et al. Mefloquine targets the Plasmodium falciparum 80S ribosome to inhibit protein synthesis. Nat Microbiol. 2017;2:1–9.

    Article  Google Scholar 

  30. Meissner PE, Mandi G, Coulibaly B, Witte S, Tapsoba T, Mansmann U, et al. Methylene blue for malaria in Africa: results from a dose-finding study in combination with chloroquine. Malar J. 2006;5:1–5.

    Article  Google Scholar 

  31. Davis TME, Hung T-Y, Sim I-K, Karunajeewa HA, Ilett KF. Piperaquine A Resurgent Antimalarial Drug. Drugs. 2005;65:75–87.

    Article  CAS  PubMed  Google Scholar 

  32. Baird JK, Hoffman SL. Primaquine therapy for malaria. Clin Infect Dis. 2004;39:1336–45.

    Article  CAS  PubMed  Google Scholar 

  33. Srivastava IK, Vaidya AB. A mechanism for the synergistic antimalarial action of atovaquone and proguanil. Antimicrob Agents Chemother. 1999;43:1334–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Peters PJ, Thigpen MC, Parise ME, Newman RD. Safety and toxicity of sulfadoxine/pyrimethamine. Drug Saf. 2007;30:481–501.

    Article  CAS  PubMed  Google Scholar 

  35. Bailly C. Pyronaridine: an update of its pharmacological activities and mechanisms of action. Biopolymers. 2021;112:e23398.

    Article  CAS  PubMed  Google Scholar 

  36. van Dyke K, Lantz C, Szustkiewicz C. Quinacrine: mechanisms of antimalarial action. Science. 1970;169:492–3.

    Article  PubMed  Google Scholar 

  37. Sanchez CP, Stein WD, Lanzer M. Dissecting the components of quinine accumulation in Plasmodium falciparum. Mol Microbiol. 2008;67:1081–93.

    Article  CAS  PubMed  Google Scholar 

  38. Junghanss T, Lanzer M. Antiprotozoal Drugs Encyclopedia of Molecular Pharmacology. Berlin: Springer; 2008.

    Google Scholar 

  39. Ebstie YA, Abay SM, Tadesse WT, A. Ejigu D. Tafenoquine and its potential in the treatment and relapse prevention of Plasmodium vivax malaria: the evidence to date. Drug Des Devel Ther. 2016;10:2387–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Erhirhie EO. Antimalarial therapies and infertility: a comprehensive review. Toxicol Int. 2016;23:107–11.

    Article  Google Scholar 

  41. Bhatt TK, Kapil C, Khan S, Jairajpuri MA, Sharma V, Santoni D, et al. A genomic glimpse of aminoacyl-tRNA synthetases in malaria parasite Plasmodium falciparum. BMC Genom. 2009;10:1–4.

    Article  Google Scholar 

  42. Summers RL, Pasaje CFA, Pisco JP, Striepen J, Luth MR, Kumpornsin K, et al. Chemogenomics identifies acetyl-coenzyme A synthetase as a target for malaria treatment and prevention. Cell Chem Biol. 2022;29:191–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Forte B, Ottilie S, Plater A, Campo B, Dechering KJ, Gamo FJ, et al. Prioritization of molecular targets for antimalarial drug discovery. ACS Infect Dis. 2021;7:2764–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Manickam Y, Chaturvedi R, Babbar P, Malhotra N, Jain V, Sharma A. Drug targeting of one or more aminoacyl-tRNA synthetase in the malaria parasite Plasmodium falciparum. Drug Discov Today. 2018;23:1233–40.

    Article  CAS  PubMed  Google Scholar 

  45. Wrenger C, Müller IB, Schifferdecker AJ, Jain R, Jordanova R, Groves MR. Specific inhibition of the aspartate aminotransferase of Plasmodium falciparum. J Mol Biol. 2011;405:956–71.

    Article  CAS  PubMed  Google Scholar 

  46. Wang C, Krüger A, Du X, Wrenger C, Groves MR. Novel highlight in malarial drug discovery: aspartate transcarbamoylase. Front Cell Infect Microbiol. 2022;12:232.

    CAS  Google Scholar 

  47. Lunev S, Batista FA, Bosch SS, Wrenger C, Groves MR. Identification and validation of novel drug targets for the treatment of Plasmodium falciparum malaria: new insights. Current Topics Malaria. 2016. https://0-doi-org.brum.beds.ac.uk/10.5772/65659.

    Article  Google Scholar 

  48. Bansal A, Singh S, More KR, Hans D, Nangalia K, Yogavel M, et al. Characterization of Plasmodium falciparum calcium-dependent protein kinase 1 (PfCDPK1) and its role in microneme secretion during erythrocyte invasion. J Biol Chem. 2013;288:1590–602.

    Article  CAS  PubMed  Google Scholar 

  49. Belen Cassera M, Zhang Y, Hazleton ZK, Schramm LV. Purine and pyrimidine pathways as targets in Plasmodium falciparum. Curr Top Med Chem. 2011;11:2103–15.

    Article  Google Scholar 

  50. Krungkrai SR, Krungkrai J. Malaria parasite carbonic anhydrase: inhibition of aromatic/heterocyclic sulfonamides and its therapeutic potential. Asian Pac j trop med. 2011;1:233–42.

    Article  CAS  Google Scholar 

  51. Graciotti M, Alam M, Solyakov L, Schmid R, Burley G, Bottrill AR, et al. Malaria protein kinase CK2 (PfCK2) shows novel mechanisms of regulation. PLoS ONE. 2014;9:e85391.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Rotella D, Siekierka J, Bhanot P. Plasmodium falciparum cGMP-dependent protein kinase – A novel chemotherapeutic target. Front Microbiol. 2021;11:610408.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Choubey V, Maity P, Guha M, Kumar S, Srivastava K, Puri SK, et al. Inhibition of Plasmodium falciparum choline kinase by hexadecyltrimethylammonium bromide: a possible antimalarial mechanism. Antimicrob Agents Chemother. 2007;51:696–706.

    Article  CAS  PubMed  Google Scholar 

  54. Guca E, Nagy GN, Hajdú F, Marton L, Izrael R, Hoh F, et al. Structural determinants of the catalytic mechanism of Plasmodium CCT, a key enzyme of malaria lipid biosynthesis. Sci Rep. 2018;8:11215.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Rosenthal P, Sijwali P, Singh A, Shenai B. Cysteine proteases of malaria parasites: targets for chemotherapy. Curr Pharm Des. 2002;8:1659–72.

    Article  CAS  PubMed  Google Scholar 

  56. Nixon GL, Pidathala C, Shone AE, Antoine T, Fisher N, O’Neill PM, et al. Targeting the mitochondrial electron transport chain of Plasmodium falciparum: new strategies towards the development of improved antimalarials for the elimination era. Future Med Chem. 2013;5:1573–91.

    Article  CAS  PubMed  Google Scholar 

  57. Mishra R, Mishra B, Moorthy NH. Dihydrofolate reductase enzyme: a potent target for antimalarial research. Asian J Cell Biol. 2005;1:48–58.

    Article  Google Scholar 

  58. Pornthanakasem W, Riangrungroj P, Chitnumsub P, Ittarat W, Kongkasuriyachai D, Uthaipibull C, et al. Role of Plasmodium vivax dihydropteroate synthase polymorphisms in sulfa drug resistance. Antimicrob Agents Chemother. 2016;60:4453–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tanaka TQ, Deu E, Molina-Cruz A, Ashburne MJ, Ali O, Suri A, et al. Plasmodium dipeptidyl aminopeptidases as malaria transmission-blocking drug targets. Antimicrob Agents Chemother. 2013;57:4645–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Koumpoura CL, Robert A, Athanassopoulos CM, Baltas M. Antimalarial inhibitors targeting epigenetics or mitochondria in Plasmodium falciparum: recent survey upon synthesis and biological evaluation of potential drugs against malaria. Molecules. 2021;26:5711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Belete TM. Recent progress in the development of new antimalarial drugs with novel targets. Drug Des Devel Ther. 2020;14:3875–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Marco M, Miguel CJ. Falcipain inhibition as a promising antimalarial target. Curr Top Med Chem. 2012;12:408–44.

    Article  CAS  PubMed  Google Scholar 

  63. Sharma K. A Review on Plasmodium falciparum-protein farnesyltransferase inhibitors as antimalarial drug targets. Curr Drug Targets. 2017;18:1676–86.

    Article  CAS  PubMed  Google Scholar 

  64. Lim S, Prieto JH. Glutathione reductase of Plasmodium falciparum as an antimalarial drug target of methylene blue. Biophys J. 2015;108:55a–6a.

    Article  Google Scholar 

  65. Colón-Lorenzo EE, Colón-López DD, Vega-Rodríguez J, Dupin A, Fidock DA, Baerga-Ortiz A, et al. Structure-based screening of Plasmodium berghei glutathione S-transferase identifies CB-27 as a novel antiplasmodial compound. Front Pharmacol. 2020;11:246.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Bruno S, Pinto A, Paredi G, Tamborini L, de Micheli C, la Pietra V, et al. Discovery of covalent inhibitors of glyceraldehyde-3-phosphate dehydrogenase, a target for the treatment of malaria. J Med Chem. 2014;57:7465–71.

    Article  CAS  PubMed  Google Scholar 

  67. Alam A, Neyaz MK, Ikramul Hasan S. Exploiting unique structural and functional properties of malarial glycolytic enzymes for antimalarial drug development. Malar Res Treat. 2014;2014:451065.

    PubMed  PubMed Central  Google Scholar 

  68. Kumar R, Musiyenko A, Barik S. The heat shock protein 90 of Plasmodium falciparum and antimalarial activity of its inhibitor, geldanamycin. Malar J. 2003;2:1–11.

    Article  Google Scholar 

  69. Fong KY, Wright DW. Hemozoin and antimalarial drug discovery. Future Med Chem. 2013;5:1437–50.

    Article  CAS  PubMed  Google Scholar 

  70. Joët T, Eckstein-Ludwig U, Morin C, Krishna S. Validation of the hexose transporter of Plasmodium falciparum as a novel drug target. Proc Natl Acad Sci. 2003;100:7476–9.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Patel V, Mazitschek R, Coleman B, Nguyen C, Urgaonkar S, Cortese J, et al. Identification and characterization of small molecule inhibitors of a class I histone deacetylase from Plasmodium falciparum. J Med Chem. 2009;52:2185–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Miao J, Wang C, Lucky AB, Liang X, Min H, Adapa SR, et al. A unique GCN5 histone acetyltransferase complex controls erythrocyte invasion and virulence in the malaria parasite Plasmodium falciparum. PLoS Pathog. 2021;17:e1009351.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Keough DT, Hocková D, Krečmerová M, Česnek M, Holý A, Naesens L, et al. Plasmodium vivax hypoxanthine-guanine phosphoribosyltransferase: a target for anti-malarial chemotherapy. Mol Biochem Parasitol. 2010;173:165–9.

    Article  CAS  PubMed  Google Scholar 

  74. Penna-Coutinho J, Cortopassi WA, Oliveira AA, França TCC, Krettli AU. Antimalarial activity of potential inhibitors of Plasmodium falciparum lactate dehydrogenase enzyme selected by docking studies. PLoS ONE. 2011;6:e21237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Reyes Romero A, Lunev S, Popowicz GM, Calderone V, Gentili M, Sattler M, et al. A fragment-based approach identifies an allosteric pocket that impacts malate dehydrogenase activity. Commun Biol. 2021;4:949.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Hartuti ED, Inaoka DK, Komatsuya K, Miyazaki Y, Miller RJ, Xinying W, et al. Biochemical studies of membrane bound Plasmodium falciparum mitochondrial L-malate:quinone oxidoreductase, a potential drug target. Biochim Biophys Acta Bioenerg. 2018;1859:191–200.

    Article  CAS  PubMed  Google Scholar 

  77. Chen X, Chong CR, Shi L, Yoshimoto T, Sullivan DJ, Liu JO. Inhibitors of Plasmodium falciparum methionine aminopeptidase 1b possess antimalarial activity. Proc Natl Acad Sci. 2006;103:14548–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Dorin-Semblat D, Quashie N, Halbert J, Sicard A, Doerig C, Peat E, et al. Functional characterization of both MAP kinases of the human malaria parasite Plasmodium falciparum by reverse genetics. Mol Microbiol. 2007;65:1170–80.

    Article  CAS  PubMed  Google Scholar 

  79. Schlott AC, Holder AA, Tate EW. N -myristoylation as a drug target in malaria: exploring the role of N -myristoyltransferase substrates in the inhibitor mode of action. ACS Infect Dis. 2018;4:449–57.

    Article  CAS  PubMed  Google Scholar 

  80. Ke H, Ganesan SM, Dass S, Morrisey JM, Pou S, Nilsen A, et al. Mitochondrial type II NADH dehydrogenase of Plasmodium falciparum (PfNDH2) is dispensable in the asexual blood stages. PLoS ONE. 2019;14:e0214023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Istvan ES, Das S, Bhatnagar S, Beck JR, Owen E, Llinas M, et al. Plasmodium Niemann-Pick type C1-related protein is a druggable target required for parasite membrane homeostasis. eLife. 2019;8:e40529.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Henry MX. A double line of defense: heat shock proteins and polyamines act as contributing factors to drug resistance of some Plasmodium parasites. In: Tyagi RK, editor. Plasmodium species and drug resistance. London: IntechOpen; 2021.

    Google Scholar 

  83. Kumar S, Krishnamoorthy K, Mudeppa DG, Rathod PK. Structure of Plasmodium falciparum orotate phosphoribosyltransferase with autologous inhibitory protein–protein interactions. Acta Crystallogr F Struct Biol Commun. 2015;71:600–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Krungkrai SR, DelFraino BJ, Smiley JA, Prapunwattana P, Mitamura T, Horii T, et al. A novel enzyme complex of orotate phosphoribosyltransferase and orotidine 5‘-monophosphate decarboxylase in human malaria parasite Plasmodium falciparum: physical association, kinetics, and inhibition characterization. Biochemistry. 2005;44:1643–52.

    Article  CAS  PubMed  Google Scholar 

  85. Koyama FC, Ribeiro RY, Garcia JL, Azevedo MF, Chakrabarti D, Garcia CRS. Ubiquitin proteasome system and the atypical kinase PfPK7 are involved in melatonin signaling in Plasmodium falciparum. J Pineal Res. 2012;53:147–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Pett HE, Jansen PA, Hermkens PH, Botman PN, Beuckens-Schortinghuis CA, Blaauw RH, et al. Novel pantothenate derivatives for anti-malarial chemotherapy. Malar J. 2015;14:1–8.

    Article  CAS  Google Scholar 

  87. Flueck C, Drought LG, Jones A, Patel A, Perrin AJ, Walker EM, et al. Phosphodiesterase beta is the master regulator of cAMP signaling during malaria parasite invasion. PLoS Biol. 2019;17:3000154.

    Article  Google Scholar 

  88. Arendse LB, Wyllie S, Chibale K, Gilbert IH. Plasmodium kinases as potential drug targets for malaria: challenges and opportunities. ACS Infect Dis. 2021;7:518–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Tawk L, Chicanne G, Dubremetz J-F, Richard V, Payrastre B, Vial HJ, et al. Phosphatidylinositol 3-phosphate, an essential lipid in Plasmodium, localizes to the food vacuole membrane and the apicoplast. Eukaryot Cell. 2010;9:1519–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Raman J, Ashok CS, Subbayya IN, Anand RP, Senthamizh T, Selvi S, et al. Plasmodium falciparum hypoxanthine guanine phosphoribosyltransferase. FEBS J. 2005;272:1900–11.

    Article  CAS  PubMed  Google Scholar 

  91. Liu P. Plasmepsin: function, characterization and targeted antimalarial drug development. Nat Remed Fight Against Parasites. 2017. https://0-doi-org.brum.beds.ac.uk/10.5772/66716.

    Article  Google Scholar 

  92. Egwu CO, Augereau J-M, Reybier K, Benoit-Vical F. Reactive oxygen species as the brainbox in malaria treatment. Antioxidants. 2021;10:1872.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Ghosh S, Kennedy K, Sanders P, Matthews K, Ralph SA, Counihan NA, et al. The Plasmodium rhoptry associated protein complex is important for parasitophorous vacuole membrane structure and intraerythrocytic parasite growth. Cell Microbiol. 2017;19:e12733.

    Article  Google Scholar 

  94. Nakanishi M. S-Adenosyl-L-homocysteine hydrolase as an attractive target for antimicrobial drugs. ChemInform. 2007;127:977–82.

    CAS  Google Scholar 

  95. McCoubrie JE, Miller SK, Sargeant T, Good RT, Hodder AN, Speed TP, et al. Evidence for a common role for the serine-type Plasmodium falciparum serine repeat antigen proteases: implications for vaccine and drug design. Infect Immun. 2007;75:5565–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Holland Z, Prudent R, Reiser J-B, Cochet C, Doerig C. functional analysis of protein kinase ck2 of the human malaria parasite Plasmodium falciparum. Eukaryot Cell. 2009;8:388–97.

    Article  CAS  PubMed  Google Scholar 

  97. Tuteja R, Pradhan A, Sharma S. Plasmodium falciparum signal peptidase is regulated by phosphorylation and required for intra-erythrocytic growth. Mol Biochem Parasitol. 2008;157:137–47.

    Article  CAS  PubMed  Google Scholar 

  98. Tarr SJ, Withers-Martinez C, Flynn HR, Snijders AP, Masino L, Koussis K, et al. A malaria parasite subtilisin propeptide-like protein is a potent inhibitor of the egress protease SUB1. Biochem J. 2020;477:525–40.

    Article  CAS  PubMed  Google Scholar 

  99. Tanaka TQ, Hirai M, Watanabe Y, Kita K. Toward understanding the role of mitochondrial complex II in the intraerythrocytic stages of Plasmodium falciparum: gene targeting of the Fp subunit. Parasitol Int. 2012;61:726–8.

    Article  CAS  PubMed  Google Scholar 

  100. Sumam de Oliveira D, Kronenberger T, Palmisano G, Wrenger C, de Souza EE. Targeting SUMOylation in Plasmodium as a potential target for malaria therapy. Front Cell Infect Microbiol. 2021;11:685866.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Lisk G, Pain M, Gluzman IY, Kambhampati S, Furuya T, Su X, et al. Changes in the plasmodial surface anion channel reduce leupeptin uptake and can confer drug resistance in Plasmodium falciparum -infected erythrocytes. Antimicrob Agents Chemother. 2008;52:2346–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Kanzok SM, Schirmer RH, Türbachova I, Iozef R, Becker K. The thioredoxin system of the malaria parasite Plasmodium falciparum. J Biol Chem. 2000;275:40180–6.

    Article  CAS  PubMed  Google Scholar 

  103. Yuthavong Y, Yuvaniyama J, Chitnumsub P, Vanichtanankul J, Chusacultanachai S, Tarnchompoo B, et al. Malarial (Plasmodium falciparum) dihydrofolate reductase-thymidylate synthase: structural basis for antifolate resistance and development of effective inhibitors. Parasitology. 2005;130:249–59.

    Article  CAS  PubMed  Google Scholar 

  104. Mudeppa DG, Kumar S, Kokkonda S, White J, Rathod PK. Topoisomerase II from human malaria parasites. J Biol Chem. 2015;290:20313–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Marchesini N, Vieira M, Luo S, Moreno SNJ, Docampo R. A malaria parasite-encoded vacuolar H+-ATPase is targeted to the host erythrocyte. J Biol Chem. 2005;280:36841–7.

    Article  CAS  PubMed  Google Scholar 

  106. Moafi M, Rezvan H, Sherkat R, Taleban R. Leishmania vaccines entered in clinical trials: a review of literature. Int J Prev Med. 2019;10:95.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Innes EA, Hamilton C, Garcia JL, Chryssafidis A, Smith D. A one health approach to vaccines against Toxoplasma gondii. Food Waterborne Parasitol. 2019;15:e00053.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Nussenzweig RS, Vanderberg J, Most H, Orton C. Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature. 1967;216:160–2.

    Article  CAS  PubMed  Google Scholar 

  109. Hollingdale MR, Sedegah M. Development of whole sporozoite malaria vaccines. Expert Rev Vaccines. 2017;16:45–54.

    Article  CAS  PubMed  Google Scholar 

  110. Arora N, Anbalagan LC, Pannu AK. Towards eradication of malaria: is the WHO’s RTS, S/AS01 vaccination effective enough? Risk Manag Healthc Policy. 2021;14:1033–9.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Nureye D, Assefa S. Old and recent advances in life cycle, pathogenesis, diagnosis, prevention, and treatment of malaria including perspectives in Ethiopia. Sci World J. 2020;1295381:1–17.

    Article  Google Scholar 

  112. Hoffman SL, Goh LML, Luke TC, Schneider I, Le TP, Doolan DL, et al. Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis. 2002;185:1155–64.

    Article  PubMed  Google Scholar 

  113. Oyen D, Torres JL, Wille-Reece U, Ockenhouse CF, Emerling D, Glanville J, et al. Structural basis for antibody recognition of the NANP repeats in Plasmodium falciparum circumsporozoite protein. Proc Natl Acad Sci. 2017;114:E10438-10445.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kucharska I, Hossain L, Ivanochko D, Yang Q, Rubinstein JL, Pomès R, et al. Structural basis of Plasmodium vivax inhibition by antibodies binding to the circumsporozoite protein repeats. eLife. 2022;11:e72908.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Bennett JW, Yadava A, Tosh D, Sattabongkot J, Komisar J, Ware LA, et al. Phase 1/2a trial of Plasmodium vivax malaria vaccine candidate VMP001/AS01B in malaria-naive adults: safety, immunogenicity, and efficacy. PLOS Negl Trop Dis. 2016;10:e0004423.

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Almeida MEM, Vasconcelos MGS, Tarragô AM, Mariúba LAM. Circumsporozoite surface protein-based malaria vaccines: a review. Rev Inst Med Trop Sao Paulo. 2021;63:e11.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Francica JR, Shi W, Chuang G-Y, Chen SJ, Pereira LDS, Farney SK, et al. Design of alphavirus virus-like particles presenting circumsporozoite junctional epitopes that elicit protection against malaria. Vaccines. 2021;9:272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  122. Jongo SA, Urbano V, Church LWP, Olotu A, Manock SR, Schindler T, et al. Immunogenicity and protective efficacy of radiation-attenuated and chemo-attenuated PfSPZ vaccines in equatoguinean adults. Am J Trop Med Hyg. 2021;104:283–93.

    Article  CAS  PubMed  Google Scholar 

  123. Roestenberg M, Walk J, van der Boor SC, Langenberg MCC, Hoogerwerf M-A, Janse JJ, et al. A double-blind, placebo-controlled phase 1/2a trial of the genetically attenuated malaria vaccine PfSPZ-GA1. Sci Transl Med. 2020;12:5629.

    Article  Google Scholar 

  124. Kublin JG, Mikolajczak SA, Sack BK, Fishbaugher ME, Seilie A, Shelton L, et al. Complete attenuation of genetically engineered Plasmodium falciparum sporozoites in human subjects. Sci Transl Med. 2017;9:9099.

    Article  Google Scholar 

  125. Singer M, Frischknecht F. Time for genome editing: next-generation attenuated malaria parasites. Trends Parasitol. 2017;33:202–13.

    Article  CAS  PubMed  Google Scholar 

  126. Marin-Mogollon C, van Pul FJA, Miyazaki S, Imai T, Ramesar J, Salman AM, et al. Chimeric Plasmodium falciparum parasites expressing Plasmodium vivax circumsporozoite protein fail to produce salivary gland sporozoites. Malar J. 2018;17:1–16.

    Article  Google Scholar 

  127. World malaria report 2021. WHO. 2022.

  128. Hume HKC, Vidigal J, Carrondo MJT, Middelberg APJ, Roldão A, Lua LHL. Synthetic biology for bioengineering virus-like particle vaccines. Biotechnol Bioeng. 2019;116:919–35.

    Article  Google Scholar 

  129. Nooraei S, Bahrulolum H, Hoseini ZS, Katalani C, Hajizade A, Easton AJ, et al. Virus-like particles: preparation, immunogenicity and their roles as nanovaccines and drug nanocarriers. J Nanobiotechnology. 2021;19:1–27.

    Article  Google Scholar 

  130. Laurens MB. RTS, S/AS01 vaccine (MosquirixTM): an overview. Hum Vaccines Immunother. 2020;16:480–9.

    Article  CAS  Google Scholar 

  131. Datoo MS, Natama MH, Somé A, Traoré O, Rouamba T, Bellamy D, 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;397:1809–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Wilson DS, Hirosue S, Raczy MM, Bonilla-Ramirez L, Jeanbart L, Wang R, et al. Antigens reversibly conjugated to a polymeric glyco-adjuvant induce protective humoral and cellular immunity. Nat Mater. 2019;18:175–85.

    Article  CAS  PubMed  Google Scholar 

  133. Alving CR, Peachman KK, Matyas GR, Rao M, Beck Z. Army Liposome Formulation (ALF) family of vaccine adjuvants. Expert Rev Vaccines. 2020;19:279–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Coler RN, Bertholet S, Moutaftsi M, Guderian JA, Windish HP, Baldwin SL, et al. Development and characterization of synthetic glucopyranosyl lipid adjuvant system as a vaccine adjuvant. PLoS ONE. 2011;6:e16333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  136. Mehrizi AA, Verjani SB, Zakeri S. Advax, as a co-adjuvant, in combination with Poly(I:C) elicits enhanced Th1 immune responses and parasite growth-inhibitory antibodies against Plasmodium falciparum merozoite surface protein-1 (PfMSP-142) in BALB/c mice. Iran J Immunol. 2021;18:279–91.

    Google Scholar 

  137. Perraut R, Varela M-L, Joos C, Diouf B, Sokhna C, Mbengue B, et al. Association of antibodies to Plasmodium falciparum merozoite surface protein-4 with protection against clinical malaria. Vaccine. 2017;35:6720–6.

    Article  CAS  PubMed  Google Scholar 

  138. Shen F, Ong JJY, Sun Y, Lei Y, Chu R, Kassegne K, et al. A chimeric Plasmodium vivax merozoite surface protein antibody recognizes and blocks erythrocytic P. cynomolgi berok merozoites in vitro. Infect Immun. 2021;89:00645–20.

    Article  Google Scholar 

  139. Elias SC, Choudhary P, Cassan SC, Biswas S, Collins KA, Halstead FD, et al. Analysis of human B cell responses following ChAd63-MVA MSP1 and AMA1 immunization and controlled malaria infection. Immunology. 2014;141:628–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Mahmoudi S, Keshavarz H. Malaria vaccine development: the need for novel approaches: a review article. Iran J Parasitol. 2018;13:1–10.

    PubMed  PubMed Central  Google Scholar 

  141. Karch CP, Doll TAPF, Paulillo SM, Nebie I, Lanar DE, Corradin G, et al. The use of a P falciparum specific coiled-coil domain to construct a self-assembling protein nanoparticle vaccine to prevent malaria. J Nanobiotechnology. 2017;15:1–10.

    Article  Google Scholar 

  142. Brod F, Miura K, Taylor I, Li Y, Marini A, Salman AM, et al. Combination of RTS, S and Pfs25-IMX313 induces a functional antibody response against malaria infection and transmission in mice. Front immunol. 2018;9:2780.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. De Graaf H, Payne RO, Taylor I, Miura K, Long CA, Elias SC, et al. Safety and immunogenicity of ChAd63/MVA Pfs25-IMX313 in a phase I first-in-human trial. Front immunol. 2021;12:694759.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Lee S-M, Hickey JM, Miura K, Joshi SB, Volkin DB, King CR, et al. A C-terminal Pfs48/45 malaria transmission-blocking vaccine candidate produced in the baculovirus expression system. Sci Rep. 2020;10:1–14.

    Google Scholar 

  145. Yenkoidiok-Douti L, Williams AE, Canepa GE, Molina-Cruz A, Barillas-Mury C. Engineering a virus-like particle as an antigenic platform for a Pfs47-targeted malaria transmission-blocking vaccine. Sci Rep. 2019;9:16833.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Huang W-C, Deng B, Mabrouk MT, Seffouh A, Ortega J, Long C, et al. Particle-based, Pfs230 and Pfs25 immunization is effective, but not improved by duplexing at fixed total antigen dose. Malar J. 2020;19:1–12.

    Article  Google Scholar 

  147. Pirahmadi S, Zakeri S, Mehrizi AA, Djadid ND, Raz A-A, Sani JJ, et al. Cell-traversal protein for ookinetes and sporozoites (CelTOS) formulated with potent TLR adjuvants induces high-affinity antibodies that inhibit Plasmodium falciparum infection in Anopheles stephensi. Malar J. 2019;18:1–16.

    Article  Google Scholar 

  148. Bender NG, Khare P, Martinez J, Tweedell RE, Nyasembe VO, López-Gutiérrez B, et al. Immunofocusing humoral immunity potentiates the functional efficacy of the AnAPN1 malaria transmission-blocking vaccine antigen. NPJ Vaccines. 2021;6:49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Payne RO, Silk SE, Elias SC, Miura K, Diouf A, Galaway F, et al. Human vaccination against RH5 induces neutralizing antimalarial antibodies that inhibit RH5 invasion complex interactions. JCI Insight. 2017;2:e96381.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Molina-Franky J, Cuy-Chaparro L, Camargo A, Reyes C, Gómez M, Salamanca DR, et al. Plasmodium falciparum pre-erythrocytic stage vaccine development. Malar J. 2020;19:1–18.

    Article  Google Scholar 

  151. Payne RO, Silk SE, Elias SC, Milne KH, Rawlinson TA, Llewellyn D, et al. Human vaccination against Plasmodium vivax Duffy-binding protein induces strain-transcending antibodies. JCI Insight. 2017;2:e93683.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Laurens MB. Novel malaria vaccines. Hum Vaccines Immunother. 2021;17:4549–52.

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  154. Schneider CG, Taylor JA, Sibilo MQ, Miura K, Mallory KL, Mann C, et al. Orientation of antigen display on self-assembling protein nanoparticles influences immunogenicity. Vaccines. 2021;9:103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Wilson KL, Pouniotis D, Hanley J, Xiang SD, Ma C, Coppel RL, et al. A synthetic nanoparticle based vaccine approach targeting MSP4/5 is immunogenic and induces moderate protection against murine blood-stage malaria. Front Immunol. 2019;10:331.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Powles L, Wilson KL, Xiang SD, Coppel RL, Ma C, Selomulya C, et al. Pullulan-coated iron oxide nanoparticles for blood-stage malaria vaccine delivery. Vaccines. 2020;8:651.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Burkhardt M, Reiter K, Nguyen V, Suzuki M, Herrera R, Duffy PE, et al. Assessment of the impact of manufacturing changes on the physicochemical properties of the recombinant vaccine carrier ExoProtein A. Vaccine. 2019;37:5762–9.

    Article  CAS  PubMed  Google Scholar 

  158. Pearce JMS. Disease, diagnosis or syndrome? Pract Neurol. 2011;11:91–7.

    Article  CAS  PubMed  Google Scholar 

  159. Most H. Falciparum malaria. JAMA. 1944;124:71–6.

    Article  Google Scholar 

  160. Bisoffi Z, Gobbi F, Buonfrate D, den Ende J, van. Diagnosis of malaria infection with or without disease. JAMA. 2012;4:e2012036.

    Google Scholar 

  161. Orish VN, Ansong JY, Onyeabor OS, Sanyaolu AO, Oyibo WA, Iriemenam NC. Overdiagnosis and overtreatment of malaria in children in a secondary healthcare centre in Sekondi-Takoradi. Ghana Trop Doct. 2016;46:191–8.

    Article  PubMed  Google Scholar 

  162. Cunha CB, Cunha BA. Brief history of the clinical diagnosis of malaria: from Hippocrates to Osler. J Vector Borne Dis. 2008;45:194–9.

    PubMed  Google Scholar 

  163. Oladosu OO, Oyibo WA. Overdiagnosis and overtreatment of malaria in children that presented with fever in Lagos. Nigeria ISRN Infect Dis. 2013;914675:1–6.

    Google Scholar 

  164. Fleischer B. Editorial: 100 years ago: Giemsa’s solution for staining of plasmodia. Trop Med Int Health. 2004;9:755–6.

    Article  PubMed  Google Scholar 

  165. Organization WH. Giemsa staining of malaria blood films malaria microscopy standard operating procedure-MM-SOP-07A 1. Purpose And Scope. 2016;7:1–6.

    Google Scholar 

  166. Dowling MA, Shute GT. A comparative study of thick and thin blood films in the diagnosis of scanty malaria parasitaemia. Bull World Health Organ. 1966;34:249–67.

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Lenz D, Kremsner PG, Lell B, Biallas B, Boettcher M, Mordmüller B, et al. Assessment of LED fluorescence microscopy for the diagnosis of Plasmodium falciparum infections in Gabon. Malar J. 2011;10:1–6.

    Article  Google Scholar 

  168. Kimura M, Teramoto I, Chan CW, Idris ZM, Kongere J, Kagaya W, et al. Improvement of malaria diagnostic system based on acridine orange staining. Malar J. 2018;17:1–6.

    Article  Google Scholar 

  169. Kawamoto F. Rapid diagnosis of malaria by fluorescence microscopy with light microscope and interference filter. The Lancet. 1991;337:200–2.

    Article  CAS  Google Scholar 

  170. Gitta B, Kilian N. Diagnosis of malaria parasites Plasmodium spp. in endemic areas: current strategies for an ancient disease. BioEssays. 2020;42:1900138.

    Article  Google Scholar 

  171. Wongsrichanalai C, Barcus MJ, Muth S, Sutamihardja A, Wernsdorfer WH. A review of malaria diagnostic tools: microscopy and rapid diagnostic test (RDT). Am J Trop Med. 2007;77:119–27.

    Article  Google Scholar 

  172. Obeagu EI, Chijioke UO, Ekelozie IS. Malaria rapid diagnostic test (RDTs). Ann Clin Lab Res. 2018;2018:275.

    Google Scholar 

  173. Jelinek T. Malaria self-testing by travellers: opportunities and limitations. Travel Med Infect Dis. 2004;2:143–8.

    Article  PubMed  Google Scholar 

  174. Mills CD, Burgess DC, Taylor HJ, Kain KC. Evaluation of a rapid and inexpensive dipstick assay for the diagnosis of Plasmodium falciparum malaria. Bull World Health Organ. 1999;77:553–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Thompson MJ. Rapid “dipstick” assays for the detection of malaria. Am Fam Physician. 2000;61:1640–3.

    CAS  PubMed  Google Scholar 

  176. Pieroni P, Mills CD, Ohrt C, Harrington MA, Kain KC. Comparison of the ParaSight™-F test and the ICT Malaria Pf™ test with the polymerase chain reaction for the diagnosis of Plasmodium falciparum malaria in travellers. Trans R Soc Trop Med Hyg. 1998;92:166–9.

    Article  CAS  PubMed  Google Scholar 

  177. Palmer CJ, Lindo JF, Klaskala WI, Quesada JA, Kaminsky R, Baum MK, et al. Evaluation of the OptiMAL test for rapid diagnosis of Plasmodium vivax and Plasmodium falciparum malaria. J Clin Microbiol. 1998;36:203–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Tjitra E, Suprianto S, Dyer M, Currie BJ, Anstey NM. Field evaluation of the ICT malaria Pf/Pv immunochromatographic test for detection of Plasmodium falciparum and Plasmodium vivax in patients with a presumptive clinical diagnosis of malaria in eastern Indonesia. J Clin Microbiol. 1999;37:2412–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Moody AH, Chiodini PL. Non-microscopic method for malaria diagnosis using OptiMAL IT, a second-generation dipstick for malaria pLDH antigen detection. Br J Biomed Sci. 2002;59:228–31.

    Article  CAS  PubMed  Google Scholar 

  180. Imwong M, Hanchana S, Malleret B, Rénia L, Day NPJ, Dondorp A, et al. High-throughput ultrasensitive molecular techniques for quantifying low-density malaria parasitemias. J Clin Microbiol. 2014;52:3303–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Cheng Z, Wang D, Tian X, Sun Y, Sun X, Xiao N, et al. Capture and ligation probe-PCR (CLIP-PCR) for molecular screening, with application to active malaria surveillance for elimination. Clin Chem. 2015;61:821–8.

    Article  CAS  PubMed  Google Scholar 

  182. Kain KC, Brown AE, Mirabelli L, Webster HK. Detection of Plasmodium vivax by polymerase chain reaction in a field study. J Infect Dis. 1993;168:1323–6.

    Article  CAS  PubMed  Google Scholar 

  183. Snounou G, Viriyakosol S, Zhu XP, Jarra W, Pinheiro L, do Rosario VE, et al. High sensitivity of detection of human malaria parasites by the use of nested polymerase chain reaction. Mol Biochem Parasitol. 1993;61:315–20.

    Article  CAS  PubMed  Google Scholar 

  184. Zheng Z, Cheng Z. Advances in molecular diagnosis of malaria. Adv Clin Chem. 2017;80:155–92.

    Article  CAS  PubMed  Google Scholar 

  185. Thongdee P, Chaijaroenkul W, Kuesap J, Na-Bangchang K. Nested-PCR and a new ELISA-based novalisa test kit for malaria diagnosis in an endemic area of Thailand. Korean J Parasitol. 2014;52:377–81.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Okyere B, Owusu-Ofori A, Ansong D, Buxton R, Benson S, Osei-Akoto A, et al. Point prevalence of asymptomatic Plasmodium infection and the comparison of microscopy, rapid diagnostic test and nested PCR for the diagnosis of asymptomatic malaria among children under 5 years in Ghana. PLoS ONE. 2020;15:0232874.

    Article  Google Scholar 

  187. Polanco JC, Rodrı́guez JA, Corredor V, Patarroyo MA. Plasmodium vivax: parasitemia determination by real-time quantitative PCR in Aotus monkeys. Exp Parasitol. 2002;100:131–4.

    Article  CAS  PubMed  Google Scholar 

  188. Lucchi NW, Narayanan J, Karell MA, Xayavong M, Kariuki S, DaSilva AJ, et al. Molecular diagnosis of malaria by photo-induced electron transfer fluorogenic primers: PET-PCR. PLoS ONE. 2013;8:56677.

    Article  Google Scholar 

  189. Perandin F, Manca N, Calderaro A, Piccolo G, Galati L, Ricci L, et al. Development of a real-time PCR assay for detection of Plasmodium falciparum, Plasmodium vivax, and Plasmodium ovale for routine clinical diagnosis. J Clin Microbiol. 2004;42:1214–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Mens PF, de Bes HM, Sondo P, Laochan N, Keereecharoen L, van Amerongen A, et al. Direct blood PCR in combination with nucleic acid lateral flow immunoassay for detection of Plasmodium species in settings where malaria is endemic. J Clin Microbiol. 2012;50:3520–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Echeverry DF, Deason NA, Davidson J, Makuru V, Xiao H, Niedbalski J, et al. Human malaria diagnosis using a single-step direct-PCR based on the Plasmodium cytochrome oxidase III gene. Malar J. 2016;15:1–12.

    Article  Google Scholar 

  192. Kamau E, Tolbert LS, Kortepeter L, Pratt M, Nyakoe N, Muringo L, et al. Development of a highly sensitive genus-specific quantitative reverse transcriptase real-time PCR assay for detection and quantitation of Plasmodium by amplifying RNA and DNA of the 18S rRNA genes. J Clin Microbiol. 2011;49:2946–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Janse CJ, van Vianen PH. Flow cytometry in malaria detection. Methods Cell Biol. 1994;42:295–318.

    Article  PubMed  Google Scholar 

  194. Janse CJ, van Vianen PH, Tanke HJ, Mons B, Ponnudurai T, Overdulve JP. Plasmodium species: flow cytometry and microfluorometry assessments of DNA content and synthesis. Exp Parasitol. 1987;64:88–94.

    Article  CAS  PubMed  Google Scholar 

  195. Jones KN, Mascia B, Waggoner-Fountain L, Pearson RD. Photo quiz. Diagnosis by automated blood analyzer. Clin Infect Dis. 2001;33:1944–95.

    Article  Google Scholar 

  196. Hänscheid T. Diagnosis of malaria: a review of alternatives to conventional microscopy. Clin Lab Haematol. 1999;21:235–45.

    Article  PubMed  Google Scholar 

  197. Mendelow B, Lyons C, Nhlangothi P, Tana M, Munster M, Wypkema E, et al. Automated malaria detection by depolarization of laser light. Br J Haematol. 1999;104:499–503.

    Article  CAS  PubMed  Google Scholar 

  198. Makler MT, Piper RC, Milhous WK. Lactate dehydrogenase and the diagnosis of malaria. Parasitol Today. 1998;14:376–7.

    Article  CAS  PubMed  Google Scholar 

  199. Kim YRAN, Yee M, Chupp K, et al. Simultaneous differentiation and quantitation of erythroblasts and white blood cells on a high throughput clinical haematology analyser. Clin Lab Haematol. 1998;20:21–9.

    Article  CAS  PubMed  Google Scholar 

  200. Tangpukdee N, Duangdee C, Wilairatana P, Krudsood S. Malaria diagnosis: a brief review. Korean J Parasitol. 2009;47:93–102.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Doderer C, Heschung A, Guntz P, Cazenave J-P, Hansmann Y, Senegas A, et al. A new ELISA kit which uses a combination of Plasmodium falciparum extract and recombinant Plasmodium vivax antigens as an alternative to IFAT for detection of malaria antibodies. Malar J. 2007;6:1–8.

    Article  Google Scholar 

  202. Sin MLY, Mach KE, Wong PK, Liao JC. Advances and challenges in biosensor-based diagnosis of infectious diseases. Expert Rev Mol Diagn. 2014;14:225–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Alnasser Y, Ferradas C, Clark T, Calderon M, Gurbillon A, Gamboa D, et al. Colorimetric detection of Plasmodium vivax in urine using MSP10 oligonucleotides and gold nanoparticles. Plos Negl Trop Dis. 2016;10:0005029.

    Google Scholar 

  204. Kolluri N, Klapperich CM, Cabodi M. Towards lab-on-a-chip diagnostics for malaria elimination. Lab Chip. 2018;18:75–94.

    Article  CAS  Google Scholar 

  205. Rubio M, Bassat Q, Estivill X, Mayor A. Tying malaria and microRNAs: from the biology to future diagnostic perspectives. Malar J. 2016;15:1–14.

    Article  Google Scholar 

  206. Selvarajah D, Naing C, Htet NH, Mak JW. Loop-mediated isothermal amplification (LAMP) test for diagnosis of uncomplicated malaria in endemic areas: a meta-analysis of diagnostic test accuracy. Malar J. 2020;19:1–10.

    Article  Google Scholar 

  207. Kumar R, Verma AK, Shrivas S, Thota P, Singh MP, Rajasubramaniam S, et al. First successful field evaluation of new, one-minute haemozoin-based malaria diagnostic device. EClinicalMedicine. 2020;22:100347.

    Article  PubMed  PubMed Central  Google Scholar 

  208. Garcia G, Lord A, Chaves L, Lima-Junior J, Sikulu-Lord M. First report of rapid, non-invasive, and reagent-free detection of malaria through the skin of patients with a beam of infrared light. Res Sq. 2022;1:1–19.

    Google Scholar 

  209. Tan TK, Rijal P, Rahikainen R, Keeble AH, Schimanski L, Hussain S, et al. A COVID-19 vaccine candidate using SpyCatcher multimerization of the SARS-CoV-2 spike protein receptor-binding domain induces potent neutralising antibody responses. Nat Commun. 2021;12:542.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Warner NL, Frietze KM. Development of bacteriophage virus-like particle vaccines displaying conserved epitopes of dengue virus non-structural protein 1. Vaccines. 2021;9:726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Pollard AJ, Bijker EM. A guide to vaccinology: from basic principles to new developments. Nat Rev Immunol. 2021;21:83–100.

    Article  CAS  PubMed  Google Scholar 

  212. Bell D, Fleurent AE, Hegg MC, Boomgard JD, McConnico CC. Development of new malaria diagnostics: matching performance and need. Malar J. 2016;15:1–12.

    Article  Google Scholar 

  213. Giacometti M, Milesi F, Coppadoro PL, Rizzo A, Fagiani F, Rinaldi C, et al. A lab-on-chip tool for rapid, quantitative, and stage-selective diagnosis of malaria. Adv Sci. 2021;8:2004101.

    Article  CAS  Google Scholar 

  214. Hemingway J, Shretta R, Wells TNC, Bell D, Djimdé AA, Achee N, et al. Tools and strategies for malaria control and elimination: what do we need to achieve a grand convergence in malaria? PLoS Biol. 2016;14:e1002380.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank the Department of Biotechnology, GOI, Department of Science and Technology, GOI, University Grants Commission, GOI, Indian Council of Medical Research, GOI, and Central University of Rajasthan for providing funds and infrastructure facilities. The authors would like to thank the Deanship of Scientific Research at Shaqra University for supporting this work.

Funding

Himani Tripathi thanks DST, Government of India, for providing INSPIRE fellowship (IF190844), and Preshita Bhalerao thanks UGC, Government of India, for providing NFSC [F. 82-44/2020 (SA-III), UGC-Ref no.: 201610039436] fellowship to pursue their doctoral degree. Hemant Arya thanks the Indian Council of Medical Research, Government of India, for the Research Associate Fellowship (ISRM/11(35)/2019). Sujeet Singh and Tarun Kumar Bhatt thank DBT, Government of India, for project funding (BT/PR24504/NER/95/746/2017). Bader S. Alotaibi, Summya Rashid and Mohammad R. Hasan appreciate the funding from Deanship of Scientific Research at Shaqra University for supporting this work.

Author information

Authors and Affiliations

Authors

Contributions

HT, PB, SS, HA, BSA, SR, and MRH wrote the review paper. HA and TKB did the final editing. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Hemant Arya, Mohammad Raghibul Hasan or Tarun Kumar Bhatt.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors have no conflicts of interest to declare.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tripathi, H., Bhalerao, P., Singh, S. et al. Malaria therapeutics: are we close enough?. Parasites Vectors 16, 130 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s13071-023-05755-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13071-023-05755-8

Keywords