You are on page 1of 12

REVIEW ESSAY

Prospects & Overviews www.bioessays-journal.com

Diagnosis of Malaria Parasites Plasmodium spp. in Endemic


Areas: Current Strategies for an Ancient Disease
Brian Gitta* and Nicole Kilian*

and P. falciparum (malaria tropica), with


Fast and effective detection of the causative agent of malaria in humans, the latter two species being responsible for
protozoan Plasmodium parasites, is of crucial importance for increasing the majority of clinical cases and deaths
the effectiveness of treatment and to control a devastating disease that affects worldwide.[1,4–6]
millions of people living in endemic areas. The microscopic examination
of Giemsa-stained blood films still remains the gold-standard in Plasmodium
1.2. Asexual Replication and Host
detection today. However, there is a high demand for alternative diagnostic Erythrocyte Reorganization Can Cause
methods that are simple, fast, highly sensitive, ideally do not rely on blood- Severe Complications
drawing and can potentially be conducted by the patients themselves. Here,
the history of Plasmodium detection is discussed, and advantages and disad- The life cycle of Plasmodium parasites
(Figure 2) involves asexual as well as sex-
vantages of diagnostic methods that are currently being applied are assessed.
ual replication that is linked to an obligate
host change from a human intermediate
host to a female mosquito of the genus Anopheles as the final
1. Introduction host.[7,8] During this complex life cycle, Plasmodium parasites
must infect and inhabit multiple cell types to ensure develop-
1.1. Malaria mental stage progression and progeny production.[7,8] The ery-
throcytic schizogony, which occurs within the erythrocytes of
Malaria (mala aria: “bad air”; a portmanteau word from the 18th the human host, involves a sophisticated reorganization of the
century), ague, or swamp fever (frz. paludisme) is a mosquito- terminally differentiated and metabolically reduced host cell in
borne parasitosis that is endemic in 87 countries and causes ap- the process of securing nutrient supply from hemoglobin di-
proximately 219 million clinical cases and 435 000 deaths per year gestion as well as blood serum uptake, removal of toxic waste,
(Figure 1).[1] Morbidity and mortality especially affect children protection against the host immune response, and life cycle
and pregnant women living in the World Health Organization progression.[1,7,9–13]
(WHO) African Region.[1,2] During schizogony, P. falciparum establishes the Maurer’s
The causative agents of malaria are protozoan parasites of the clefts, a secretory organelle that resides outside the parasite’s
genus Plasmodium.[1] This genus includes more than 200 differ- cellular boundaries within the erythrocyte cytoplasm, to facili-
ent species that parasitize various hosts such as reptiles, birds, tate host–parasite interaction and sequestration to avoid splenic
amphibians, and mammals (53 species total, of which 30 species clearance of the infected erythrocyte.[14] Sequestration is either
parasitize primates).[3] To date, six Plasmodium species have been achieved via cytoadherence of the infected erythrocyte to various
identified as human pathogenic, and they cause different types receptors presented on the surface of vascular endothelial cells
of malaria: P. malariae (malaria quartana), P. ovale (with two in the postcapillary venules of different organs or via rosetting
subspecies P. o. curtisi and P. o. wallikeri causing malaria ter- of infected erythrocytes with uninfected erythrocytes.[9,15,16] The
tiana), P. knowlesi (zoonotic malaria), P. vivax (malaria tertiana), cytoadherence of P. falciparum-parasitized erythrocytes is estab-
lished by members of the diverse var multigene family encoding
several different versions of P. falciparum erythrocyte membrane
B. Gitta protein 1 (PfEMP1), which are exclusively expressed in P. falci-
Matibabu
120 Semawata Rd, Ntinda, Kampala 00256, Uganda parum and presented on knob-like protrusions on the erythrocyte
E-mail: gitta@matibabu.io plasma membrane.[17,18]
Dr. N. Kilian The causative agent of zoonotic malaria in Southeast Asia, P.
Centre for Infectious Diseases knowlesi, also generates a secretory organelle in the erythrocyte
Parasitology Heidelberg University Hospital cytoplasm called Sinton Mulligan’s clefts that appears to play a
Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
crucial role in receiving and harboring proteins produced by the
E-mail: nicole.kilian@med.uni-heidelberg.de
erythrocyte-residing parasite.[19,20] It has been further reported
© 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc. This in an in vitro study by Fatih et al. that P. knowlesi might be
is an open access article under the terms of the Creative Commons able to establish a cytoadhesive phenotype that would allow at-
Attribution License, which permits use, distribution and reproduction in tachment of the host erythrocytes to the inducible human en-
any medium, provided the original work is properly cited. dothelial receptors intercellular adhesion molecule-1 (ICAM-1)
DOI: 10.1002/bies.201900138 and vascular cellular adhesion molecule (VCAM).[21] However,

BioEssays 2020, 42, 1900138 1900138 (1 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

Figure 1. Countries with indigenous cases in 2000 and their status by 2017. Countries with zero indigenous cases over at least the past 3 con-
secutive years are considered to be malaria free. All countries in the WHO European Region reported zero indigenous cases in 2016 and again in
2017. In 2017, both China and El Salvador reported zero indigenous cases. Source: WHO database.[1] Image published with permission from the
WHO.

a more recent study did not observe P. falciparum-like endothe- 1.3. The History of the Malaria Parasite and Vector Discovery
lial cytoadherence and subsequent sequestration of P. knowlesi- Sets the Stage for Today’s Gold Standard of Malaria Diagnosis
infected erythrocytes, but suggested that intravascular hemolysis
might instead be an important factor in the development of se- Malaria is a disease that might already have been documented by
vere symptoms.[22] the cultures of ancient societies, such as those of ancient China,
During an infection with P. ovale and P. vivax, the erythro- Egypt, India, Mesopotamia, and Greece (Figure 3).[34,35] The early
cyte cytoplasm does not contain membrane structures. Instead, Greeks, including Homer, Plato, Empedocles, and Hippocrates,
the erythrocyte plasma membrane consists of parasite antigen- were well aware of the characteristic symptoms of malaria such
containing caveolae called the Schüffner’s dots.[23,24] To date not as distinct fevers, splenomegaly, and generally poor health.[34,36]
much is known about the establishment and function of the In the 17th century, these symptoms were already treated with
Schüffner dots, but some evidence currently supports the no- “Jesuits’ powder,” a component of the cinchona tree bark, which
tion that P. vivax-infected erythrocytes are also able to display is now known as quinine, and is still in use as an antimalar-
cytoadherence.[18,25] Both P. ovale- and P. vivax-infected erythro- ial drug today.[37] However, another century would pass until the
cytes are able to form rosettes, but rosette formation in vivax and first histopathological discovery of the parasites themselves was
ovale malaria is, in contrast to P. falciparum, not associated with made by military physician Charles Louis Alphonse Laveran in
severe malaria.[26–28] An infection with P. malariae is not associ- 1880 (Figure 3).[38] By using direct microscopy, Alphonse Laveran
ated with severe malaria either, although the parasite is still able was able to identify granules of black pigment in the erythro-
to establish a successful erythrocyte infection and reorganization cytes of a feverish soldier in a military hospital in Constantine
generating the Ziemann stipplings (intraerythrocytic dots similar (Algeria).[38,39] Laveran further suspected that mosquitoes were
to Schüffner dots) in the process.[29] transmitting the disease, but it was Sir Ronald Ross who veri-
Parasite replication, host erythrocyte reorganization, host– fied this hypothesis and solved the life cycle mystery by examin-
parasite interaction, cytoadherence, and rosetting can cause var- ing birds that were infected with P. relictum almost two decades
ious severe symptoms such as severe anemia, jaundice, shock, later.[36,40] In 1898, Italian malariologists, among them Camillo
multiple organ dysfunction syndrome (MODS), acute pulmonary Golgi, identified Anopheles mosquitoes as transmitters of human
edema, acute kidney injury, acidosis and hypoglycemia, cerebral malaria.[40] Camillo Golgi was further able to connect the dis-
malaria, and coma, which can ultimately lead to the death of the tinct fever curves with the rupture of blood schizonts and mero-
patient.[30–33] zoite release.[40,41] It took another 50 years until Henry Shortt and

BioEssays 2020, 42, 1900138 1900138 (2 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

Figure 2. Life cycle of human-pathogenic Plasmodium parasites. Schizogony and sporogony of human-pathogenic Plasmodium parasites occurring
between the intermediate host (Homo sapiens) and the final host (Anopheles), respectively. An infected female Anopheles mosquito inoculates Plasmodium
sporozoites into the skin of the human host during the blood meal. The sporozoites glide away from the inoculation site to reach a blood vessel that
quickly carries them to the liver sinusoids. The sporozoites leave the bloodstream, infect the hepatocytes, and develop into liver schizonts. The liver
schizonts produce and release thousands of haploid merozoites into the bloodstream after rupture (liver schizogony). P. vivax and P. ovale can further
form a dormant developmental stage (hypnozoites) that is able to persist in the liver, and can cause a malaria relapse even years after the original
Plasmodium infection. The freshly released liver merozoites are able to infect the erythrocytes of the human host. Within the erythrocytes, the parasite
matures from the juvenile ring developmental stage to the mature schizont developmental stage, which consists of various merozoites (erythrocytic
schizogony). These merozoites are released into the bloodstream after schizont rupture and invade new erythrocytes. Eventually, some parasites begin
their differentiation into sexual developmental stages (gametocytes: female macrogametocytes and male microgametocytes). The sexual developmental
stages are then ingested by the next female Anopheles mosquito during the blood meal. Within the midgut of the mosquito, macrogamet and microgamet
are mating and form a zygote. This zygote develops into an ookinete that penetrates the midgut wall, where it further develops into an oocyst. The
oocyst then produces sporozoites, which are released after oocyst rupture and invade the salivary glands of the mosquito. The sporozoites that have
been produced during this sporogony can then again be inoculated into the human intermediate host during the next blood meal via the secretion of
the mosquito’s anticoagulant saliva.

BioEssays 2020, 42, 1900138 1900138 (3 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

Figure 3. Milestones of Plasmodium parasite discovery and malaria diagnosis. Cultures of ancient societies might already have observed and described
malaria symptoms. The actual discovery of Plasmodium came centuries later with the use of direct microscopy. Giemsa-stained blood film microscopy is
still the gold standard for malaria diagnosis today. Development of various PCR approaches, several different RDTs, and countless other methods have
provided more diagnostic tools with varying sensitivity, and practical advantages as well as disadvantages.

Cyril Garnham discovered that a pre-erythrocytic liver schizogony always be tested at a hospital by applying the diagnostic tools dis-
has to occur before the schizogony in the erythrocytes can take cussed in the following paragraphs.
place.[36] In 1982, Wojciech Krotoski observed dormant hypno-
zoite stages that were being produced in the liver of the human
host by P. vivax (as well as P. ovale) and caused relapses of malaria
tertiana.[42,43] 2.2. Microscopy of Thick and Thin Blood Films

The microscopic examination of Giemsa-stained blood films re-


mains the gold standard method to detect Plasmodium parasites
2. Diagnosis of Plasmodium Parasites Today: Old in the erythrocytes of patients, although devices that automati-
but Gold versus New and Fast cally analyze the blood of the patients are currently being devel-
The detection of Plasmodium parasites replicating in the ery- oped and tested.[49] Automatic slide reading approaches or vision-
throcytes of the human host is challenging because the parasite based devices have been developed and tested as well, but the
density of a patient can range from below 1 parasite/µL to tens of manual microscopic examination conducted by trained person-
thousands of parasites per microliter.[44] Parasite densities are a nel can still be seen as the preferred approach when it comes to
result of pronounced age patterns and reflect lifetime exposure as thin or thick blood film analysis.[50]
well as naturally acquired immunity at a population level.[44] Any Regardless, proper diagnosis of malaria and the examination
given diagnostic tool must be able to provide a fast and accurate of Plasmodium parasites are still challenging almost over 140
result to allow an appropriate treatment of the patient.[44,45] years after the discovery of the parasites by Alphonse Laveran.
In 1880, Laveran was relying on direct microscopy while he ex-
amined the unstained samples of his patients until he discovered
parasite-derived changes in the infected erythrocytes.[35,38] The ex-
2.1. Clinical Diagnosis amination of the blood samples was simplified after the develop-
ment of various methylene blue-based stains by Paul Ehrlich.[51]
Clinical diagnosis is solely based on the symptoms that a pa- With these newly developed stains, it was discovered that human
tient displays. It is used as a diagnostic method when labora- malaria is not caused by one Plasmodium species, which was orig-
tory facilities are not available or when the patient is performing inally hypothesized by Alphonse Laveran, but by various Plasmod-
self-diagnosis at home.[46,47] Several tropical diseases cause symp- ium species.
toms that are similar to malaria.[48] Self-diagnosis as well as self- In 1891, Dimitry Romanowsky further extended and modi-
treatment are error-prone procedures that can lead to overdiag- fied the original staining protocol.[52] By adding eosin to methy-
nosis and overtreatment.[48] Clinical suspicion of malaria should lene blue, he was able to differentially stain the nucleus and

BioEssays 2020, 42, 1900138 1900138 (4 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

the cytoplasm of the Plasmodium parasites replicating within the silent reservoirs by maintaining low-level residual malaria trans-
erythrocytes.[52] However, preparation and reproducibility of the mission in the community.[59] Detecting SMM infections is there-
staining was difficult because of the required aging process of fore just as important as detecting infections that cause a high
the methylene blue.[53] After the proposal and testing of various parasitemia.
additional modifications by numerous contemporary scientists Although light microscopy of Giemsa-stained thick and thin
during that time, Bernhard Nocht and Gustav Giemsa strove to blood films remains the gold standard in the detection of Plas-
improve the Romanowsky staining.[53] Bernhard Nocht had dis- modium parasites, there is a high demand for more sensitive and
covered that aging of methylene blue leads to the formation of a automated microscopy techniques to provide the proper determi-
new dye (“red from methylene blue”) that could stain the chro- nation of the parasitemia of the parasite in the patient’s blood and
matin of Plasmodium parasites.[53] Gustav Giemsa was further the Plasmodium species that is causing the infection.
able to improve this new staining by identifying the newly dis- The world health technology (WHT) autoanalyzer is an auto-
covered dye as azure B and determining that both eosin and mated malaria slide scanning system and was one of the first de-
an excess of Azure B needed to be dissolved in a mixture of vices that was tested and performed at a level comparable to many
glycerol and methanol to achieve the desired stability as well as human slide readers in a study conducted in 2012.[61] Prescott
reproducibility.[54] Gustav Giemsa’s stain remains, 115 years after et al. acknowledged that minimal additional equipment is needed
its first publication in 1904, the method of choice for the staining to examine blood films that have been stained with standard pro-
of Plasmodium parasites. cedures and that the device is able to perform with an estimated
The gold standard for malaria diagnosis is the routinely used limit of detection of 140 parasites µL–1 .[61,62]
microscopic examination of thick and thin blood films stained A Global Good Fund prototype was tested in 2017 and showed
with Giemsa’s stain.[55,56] To create a thick blood film, a drop of an estimated limit of detection of 100 parasites µL–1 for P. falci-
peripheral blood is removed from the patient’s finger, applied on parum while scanning 0.2 µL of blood.[62] A more recent approach
a glass slide, and laked before or during the subsequent stain- uses a prototype digital microscope device, the Autoscope, that
ing with Giemsa’s stain, which causes the rupture of the erythro- employs an automated microscopy algorithm based on machine-
cytes and allows only the visualization of leukocytes, platelets, learning to simplify the Plasmodium species detection as well as
and parasites. For the thin blood film, the patient’s peripheral the parasitemia determination.[63] The performance of the de-
blood is spread on a larger area of the glass slide. The smear is vice was comparable to routine microscopy when the slides con-
then briefly fixed in methanol and, after a brief drying period, tained an adequate volume of blood to meet the preset design
stained in Giemsa’s stain. The additional fixation step performed assumptions.[63] However, Autoscope and trained personnel both
during the preparation of the thin blood film leaves the infected missed the same low-parasitemia slides that were determined
erythrocytes intact. While thick blood films are generated to de- positive via polymerase chain reaction (PCR).[63]
tect the presence of Plasmodium parasites and to determine the CellaVision DM96 is another system that digitally determines
parasitemia, thin blood smears are used to determine the Plas- the cell morphology and is able to automatically classify leuko-
modium species responsible for the infection and the develop- cytes, as well as different forms of erythrocytes such as para-
mental stages that are currently circulating within the blood of sitized erythrocytes, on blood films using its advanced red blood
the patient. Thick and thin blood films allow a comparatively fast cell application (ARBCA).[64] Florin et al. examined the speci-
preparation and examination of the patient’s blood. ficity of the CellaVision DM96 and compared it to standard light
However, besides their obvious advantages, both techniques microscopy.[64] The device generally displayed a low sensitivity,
also have distinct disadvantages. A trained expert is required for but showed a good correlation with microscopy paired with short
both techniques to perform the entire procedure from the finger turnaround times, which might be applicable in follow-up exam-
prick to the final microscopic analyses. An untrained examiner inations to determine the parasitemia after initial diagnosis and
might, for example, confuse the ring stage Plasmodium parasites treatment.[64]
with the ring stages of another protozoan parasite, Babesia, or Another commercialized system, the X-rapid system, has been
vice versa.[57] The correct identification of the Plasmodium species exclusively developed for smartphone microscopy and provides a
and the determination of the parasitemia are therefore crucial to lens attachment as well as a light-emitting diode (LED) attach-
define the appropriate treatment, to exclude the occurrence of ment to allow the examination of erythrocytes.[62] Future studies
complications due to a severe infection, and to ensure the sur- still need to be conducted to see whether this system can be used
vival of the patient. Thin blood films are further less sensitive for routine malaria diagnostic procedures.
than thick blood films if the parasitemia is low, but in vitro work
showed that even in the hands of a trained expert, thick blood
films prepared from malaria cultures at known parasitemia con- 2.3. Microscopic Examination of Fluorescently Stained
sistently underestimate parasite densities.[56] This might be due Plasmodium Parasites
to a large number of parasites being lost during staining, which
limits the sensitivity of the method and leads to wrong estimates Fluorescent labeling followed by microscopic assessment of Plas-
of parasite density.[56] Another problem is the detection of the low- modium parasites is another applied technique in diagnostic pro-
density asymptomatic submicroscopic malaria (SMM) infections cedures. Usually, the blood of the patients is incubated with
during the dry season in the endemic areas.[58–60] A lack of breed- acridine orange that immediately stains DNA and RNA of the
ing sites for the Anopheles mosquitoes can lead to less frequent different developmental stages of Plasmodium parasites.[65,66] The
transmission occurrences. However, many patients can still be fluorescent parasites are then imaged using either a conventional
infected without displaying any symptoms, and they can act as fluorescence microscope, a halogen lamp plus interference filter

BioEssays 2020, 42, 1900138 1900138 (5 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

system or a fluorescence microscope based on LED.[66,67] The lat- However, various quantitative PCR (qPCR) assays have been
ter approach has the advantage that it can be used in field ap- developed to reliably detect Plasmodium parasites in clinical pa-
plications because of decreased energy consumption, stronger tients and even in asymptomatic samples.[44] Recently evalu-
brightness, and lower costs.[66,67] A recent study by Kimura et al. ated diagnostic real-time qPCR, comprising a portable device
showed that this approach produces superior results relative to and the Q3-Plus silicone chip, has been developed to avoid the
the Giemsa method with a correctly performed revised acridine use of bulky thermocyclers, temperature-controlled transporta-
orange staining protocol.[66,67] Although this is a feasible method tion, and storage of the necessary reagents.[82] However, ab-
leading to fast diagnostic results, trained personnel are needed to solute quantification of parasites by qPCR is challenging be-
correctly label the blood sample of the patient and to perform the cause a standard curve must be established, which can vary
analysis properly with the fluorescent microscope. between laboratories.[44] Droplet digital PCR (ddPCR) allows
accurate and absolute quantification by counting DNA molecules
encapsulated in approximately 15 000 discrete, volumetrically de-
fined, water-in-oil droplet partitions that are subjected to end-
2.4. Molecular Approaches to Diagnose Malaria via point PCR.[44,83,84]
Plasmodium-Specific Genes Loop-mediated isothermal amplification (LAMP) might be an
alternative to the various PCR approaches, because the reaction
The PCR was developed in 1983 by Kary Mullis, who won the can simply be conducted in one tube at a constant temperature,
Nobel Prize for this work 10 years later (Figure 2).[68,69] The PCR and it does not need a thermal cycler, which is particularly im-
uses the properties of thermostable DNA polymerases of bacte- portant for a fast malaria diagnosis in remote areas.[85] LAMP is
rial origin to amplify even small DNA fragments of interest by also a sensitive method that is able to detect placental malaria in
using distinct temperature changes and exposure. the peripheral blood of patients.[85]
To detect different species of Plasmodium parasites replicat-
ing in the erythrocytes of the patients, multiple PCR approaches
such as nested PCR (nPCR) or semi-nested multiplex PCR 2.5. Rapid Diagnostic Tests: An Alternative for Fast Malaria
(SnM-PCR) can be applied.[70–73] nPCR and SnM-PCR are de- Diagnosis
fined by two consecutive amplification rounds with a second set
of primer and the previously amplified DNA fragment or frag- In areas where microscopy of thin and thick blood films or other
ments representing the new template. The primer selection de- approaches to detect Plasmodium parasites in the blood of pa-
pends on the copy number of the targeted gene, the amount of ex- tients cannot be provided, antigen-based rapid diagnostic tests
pected Plasmodium-infected erythrocytes, and the body fluid that (RDTs) can be an important alternative for an easy and fast di-
is being examined.[72,74,75] For example, the small-subunit 18S agnosis that should not take longer than 15 min.[67,86] In the
rRNA gene and the highly conserved pfk13, encoding the Kelch13 last decade, the amount of available RDTs and the frequency
protein, can be targeted when fresh or even dried patient blood of their use in epidemic investigations, as well as surveys, have
samples are examined.[71,76–78] A recent publication by Lloyd et al. rapidly increased (Figure 2).[86] RDTs can also be a useful tool of
probed for cox3 and varATS genes (encoding for the cytochrome malaria self-diagnosis when applied by tourists traveling in en-
c oxidase subunit 3 and the var gene acidic terminal sequence, re- demic areas.[87]
spectively) when looking for P. falciparum parasites in the saliva Currently available RDTs are, for example, able to detect
of the patients.[72] histidine-rich protein 2 (HRP2), Plasmodium lactate dehydro-
The PCR approach represents the most sensitive method for genase (pLDH), or aldolase and provide the patient with a
the diagnosis of Plasmodium parasites because it is able to detect qualitative, but not a quantitative, result.[33,88,89] It has previously
submicroscopic parasitemias that are usually missed by other di- been suggested that cytoadherent biomass sequestered in the
agnostic approaches.[79] vascular bed of the inner organs can be estimated from HRP2
The application of various PCR approaches is further of partic- concentrations in the blood plasma.[33,89] However, a high rate of
ular importance when epidemiological data are needed for mon- false positives was recently reported in high-transmission areas
itoring malaria control and transmission during different sea- on account of the persistent antigenicity of HRP2 even weeks
sons, general mass screening and elimination interventions.[79] after the infection.[33,67,90] Aldolase tests have been shown to be
Despite the clear advantages of the PCR approaches, the exper- less sensitive when the patient is not infected with P. falciparum
imental setup can be expensive because of the required material. but with another Plasmodium species.[33] pLDH test can also
Another factor is the time necessary for the preparation of the vary in performance or even fail, depending on the Plasmodium
samples, setup of the reaction (storage of the reagents), time un- species the patient is infected with.[91] Plasmodium glutamate
til the thermocycler has completed the reaction, and the subse- dehydrogenase (pGDH) is another potential RDT candidate,
quent analyses of the results. A trained expert is further needed because the homohexameric protein is being produced by the
to setup and to conduct the experiment, to interpret the results, parasites throughout the entire schizogony, while being absent
and to perform troubleshooting in case the chosen PCR approach from host erythrocytes. However, there is a high similarity
does not work. A PCR-based diagnosis can fail when parasites between the glutamate dehydrogenase produced by the different
have genetically diverse sequences at the target region of the Plasmodium species, which can cause cross reaction in areas
primers or when the copy number of the target gene is low, which with mixed Plasmodium species infections.[92,93] Some studies
results in a lower amplification efficiency that will reduce the further reported the patient serum concentration of the protein
sensitivity.[80,81] lies usually in nanomolar range.[93,94] Therefore, further tests are

BioEssays 2020, 42, 1900138 1900138 (6 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

needed to determine whether pGDH-based RDTs can be applied sive and might need to be operated and carefully maintained by
to detect Plasmodium parasites in endemic areas.[93] trained personnel to ensure an appropriate diagnosis.
Another problematic point is that RDTs might give positive re-
sults in febrile patients whose fevers are not caused by Plasmod-
ium parasites.[47] Quality-assured RDTs can be important to con-
2.8. Microfluidic Devices: Small but Powerful Tools for Malaria
firm malaria cases, while the cause of fever in the non-malaria
Diagnosis
patients should be followed up and managed appropriately.[46]
A newly developed, highly sensitive RDT (HS-RDT) that has re-
Microfluidic devices are portable, easy-to-use, self-contained, and
cently been tested for its ability to diagnose malaria in pregnancy
low-cost diagnostic devices that allow the precise manipulation
in a low-transmission setting might be a good strategy to better
of small sample volumes while reducing reagent consumption,
detect Plasmodium parasites in the future.[95]
which is crucial for malaria endemic areas with limited access to
healthcare.[106–109] One of the first microfluidic approaches in an
effort to diagnose malaria was conducted by Hou et al. in 2010,
who used distinct changes of erythrocyte plasma membrane stiff-
2.6. Serological Detection of Plasmodium Parasites Using Serum
ness to detect Plasmodium parasites in the erythrocytes of the hu-
from Malaria Patients
man host.[110] In 2016, Xu et al. used origami paper folding for
sequential steps of DNA extraction followed by LAMP and a flu-
The immunofluorescence antibody test (IFAT) has been a reliable
orescence readout to detect Plasmodium parasites in finger prick
serological test for the presence of Plasmodium parasites.[96] This
blood samples within 45 min.[111] A recent study conducted in
approach can be used to detect Plasmodium-specific antibodies in
Uganda by Reboud et al. again applied a paper-based microflu-
epidemiological surveys and in the screening procedures of po-
idic device that combined the vertical sample-processing steps of
tential blood donors.[96,97] Plasmodium antigen is prepared on a
the origami device with a microfluidic lateral flow LAMP ampli-
slide and stored at –30 °C until patient serum is applied, and the
fication and a simple visualization system for multiplexed DNA-
amount of immunoglobulin G and M antibodies can be quanti-
based malaria diagnosis from finger prick blood samples.[108]
fied using fluorescence microscopy.[96]
This approach showed a higher sensitivity than microscopy and
A similar method for the detection of Plasmodium-specific anti-
RDTs and is currently being evaluated by the Uganda Vector Con-
bodies in the blood of patients is the enzyme-linked immunosor-
trol Division, Ministry of Health, for potential usage in areas
bent assay (ELISA) that uses different antigens from different
without centralized facilities.[108]
Plasmodium species for antibody detection using a 96-well plate
and an appropriate plate reader.[97] Although both approaches are
relatively simple and moderately sensitive, they are very time con-
suming, and trained personnel are also needed to conduct the 2.9. Aptamer-Mediated Plasmodium-Specific Diagnosis of
experiments and analyze the results. Malaria

Aptamers or “chemical antibodies” are short, single-stranded


oligonucleotides such as DNA, RNA, or synthetic xeno nucleic
2.7. Flow Cytometric Approaches to Identify Infected Erythrocytes acids molecules, obtained by in vitro evolution techniques and
defined by high affinity as well as specificity to interact with
Flow cytometry was initially developed in 1950s and is today used any desired corresponding target by folding into distinct tertiary
for the laser-based sorting and measurement of various param- structures.[112–115] Since the invention of the process of aptamer
eters of several thousand cells per second in a rapidly flowing selection, called “systematic evolution of ligands by exponential
fluid stream.[98] Various studies reported the successful detection enrichment” (SELEX) in the early 1990s by Tuerk and Gold, great
of Plasmodium species in different flow cytometry assays using efforts have been made to make the application clinically rele-
different devices over the years.[99–104] The Sysmex hematology vant for various diseases such as macular degeneration, cancer,
analyzers (Sysmex Corporation, Kobe, Japan) are promising ad- thrombosis, or inflammatory diseases.[115] The diagnostic appli-
junctive diagnostic tools to facilitate a fast detection of malaria cation of aptamers can be advantageous because the oligonu-
cases in a fluorescence flow cytometry setup by counting and dif- cleotides are cheap and easy to synthetize, have a high stability
ferentiating between cell types and Plasmodium species.[100] An- in harsh environmental conditions, and can be stored without
other promising approach in the detection of Plasmodium par- functional degradation.[114] The selection for the ideal aptamer
asites is the noninvasive in vivo photoacoustic flow cytometry candidate usually begins with a randomized oligonucleotide li-
(PAFC) that irradiates circulating erythrocytes directly in periph- brary that is incubated with the target molecule followed by affin-
eral vessels through the skin by using focused linear laser beams ity selection rounds and PCR amplification steps to determine
as well as laser-induced photoacoustic waves or fluorescence light the sequence with the highest affinity.[115] Several aptamers have
that are detected with ultrasound transducers and photodetec- already been commercialized to date.[115] So far, various aptamer
tors, respectively.[105] PAFC shows greater sensitivity than con- target proteins of Plasmodium parasites such as pLDH, the high
ventional flow cytometry and allows the examination of an almost mobility group Box 1 protein (HMGB1), PfEMP1, and pGDH
entire blood volume.[105] have been tested.[112,116–122] The future will tell whether the chem-
Flow cytometry is a very sophisticated approach to accurately ical flexibility of aptamers can further be exploited for binding
detect Plasmodium parasites, but the equipment can be expen- optimization and stability in blood, plasma, or samples, but

BioEssays 2020, 42, 1900138 1900138 (7 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

Box 1
Vaccine Development and Treatment Strategies to Protect Malaria Patients

The development of an effective vaccine against Plasmodium Protection against P. vivax could be achieved by targeting
parasites is crucial to reduce the amount of annual malaria TRAP, the P. vivax circumsporozoite protein (PvCSP) or
cases and lift some of the burden off the heavily affected the blood-stage antigen Duffy binding protein.[126–130] Vivax
malaria endemic areas, especially with regard to the two most malaria vaccine development and testing is currently ongoing
prevalent of the human-pathogenic Plasmodium species P. vi- as well.[126–130]
vax and P. falciparum.[123] However, vaccine development is Until a potent vaccine is developed, people living in endemic
scientifically and technically challenging. From all of the can- areas have to rely on artemisinin-based combination therapies
didates that are being developed and examined at the mo- (ACTs), primaquine and chloroquine (in case of an infection
ment, RTS,S/AS01 or MosquirixTM, which targets the P. fal- with a chloroquine-sensitive P. vivax strain) for the treatment
ciparum circumsporozoite protein, appears to be the most of malaria.[131] The WHO further suggests the addition of a
promising, and it is currently being tested in a field study to single low dose of primaquine to the antimalarial treatment in
protect the immunized individuals against P. falciparum.[124] order to reduce transmission of the infection in low transmis-
The non-replicating viral vector candidate ChAd63 MVA ME- sion areas.[131] Severe malaria, in contrast, is usually treated
TRAP (chimpanzee adenovirus 63 modified vaccinia virus with injectable artesunate (intramuscular or intravenous) fol-
Ankara multiple epitope-thrombospondin-related adhesion lowed by several days of ACT treatment.[131]
protein) and the whole parasite/attenuated sporozoite vaccine Diagnostic approaches must be able to detect the Plasmodium
candidate PfSPZ (Plasmodium falciparum sprozoite) both tar- species that is causing the infection and replicating within the
get P. falciparum-infected hepatocytes and are currently being erythrocytes to ensure a fast and appropriate treatment to pro-
investigated in phase IIb clinical trials.[124,125] tect the life of the patient.

aptamers represent a very promising approach for malaria lows substance identification and the observation of molecular
diagnosis.[117] changes.[139–141] In malaria diagnosis, surface-enhanced Raman
spectroscopy (SERS) is, for example, applied to detect the malaria
pigment hemozoin, an iron-containing paramagnetic byproduct
of the hemoglobin digestion (see below) during asexual replica-
2.10. Bloodless Malaria Diagnosis: Examining Bodily Fluids and
tion, by using silver nanoparticles that enhance the hemozoin
Feces for Parasite Detection
Raman signal by 106 to 107 folds.[142] Hemozoin has already been
the selected molecule for Raman spectroscopy-based Plasmodium
Drawing blood for diagnostic purposes can be challenging in en-
detection for quite a few years.[143]
demic areas. Bodily fluids such as saliva and urine or fecal matter
Another previously published SERS approach has shown that
represent a good alternative because they can be noninvasively
different stages of Plasmodium-infected erythrocytes, in particu-
obtained and tested for the presence of Plasmodium parasites us-
lar the SERS spectra from ring stage-infected erythrocytes, differ
ing different markers and technical approaches such as PCR,
from those of uninfected erythrocytes and erythrocytes infected
nPCR, lateral flow immunoassay, microfluidics with DNA sen-
with later developmental stages of the parasite.[144]
sor substrate, and immunochromatography.[80,132–137] The suc-
Raman spectroscopy is a sophisticated method that has the po-
cessful detection of Plasmodium DNA or proteins depends on
tential for a very sensitive detection of Plasmodium infection in
their concentration in the bodily fluid chosen for detection. A re-
malaria patients. However, like standard medical instruments,
cent study conducted in 2017 by Oyibo et al. reported that the
the clinical Raman system should be small in size, easy to trans-
urine malaria test (UMT) developed by Fyodor Biotechnologies,
port with a robust calibration and a high sensitivity among
Inc. (Baltimore, MD, USA) is the only nonblood malaria test that
other crucial parameters to become a successful tool in malaria
has undergone a full-scale premarket evaluation trial.[138]
diagnosis.[145]

2.11. Raman Spectroscopy to Identify Plasmodium-Infected 2.12. Hemozoin-Based Detection of Plasmodium Parasites
Erythrocytes via Different Parasite and Host Erythrocyte
Parameters Plasmodium parasites are not able to digest the toxic heme that
remains from the digestion of hemoglobin.[157] Heme is stored
Raman spectroscopy measures the wavelength (or wavelength in a metabolically crystallized form called malaria pigment or
shift) as well as the intensity of inelastically scattered light hemozoin.[157] Hemozoin does not only serve as heme storage,
(Raman effect) that has been emitted from a molecule in a but also represents a potent inhibitor of immune cells.[146] There-
liquid, solid, or gas due to interactions of the incident light fore, hemozoin is an important molecule that can serve as a
with the molecule’s vibrational energies or signature, which al- marker for malaria diagnosis. Besides the above-mentioned Ra-

BioEssays 2020, 42, 1900138 1900138 (8 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

man spectroscopy, hemozoin is being investigated as a poten- want to express their gratitude to the partners and further thank the fol-
tial marker for several different malaria diagnosis approaches lowing organizations for their support: UN Women (MSFT Imagine cup),
such as magneto-optical detection, laser desorption-time of flight Resilient Africa Network, ISHOW (American Society of Mechanical En-
gineers), Villgro Kenya, Aspirin Social Award (Bayer Foundation), Royal
(LD-TOF) mass spectrometry, hemozoin-catalyzed precipitation,
Academy of Engineering’s Africa Prize, E4 Impact Challenge, Merck Accel-
magnetic purification, etc.[147–154] erator, Rolex, the Parasitology Unit of the Centre for Infectious Diseases,
Interestingly, hemozoin is also being produced by other par- Heidelberg University Hospital (Heidelberg, Germany), the Centre for In-
asites that feed on blood such as Schistosoma mansoni, Haemo- fectious Diseases, Parasitology at Heidelberg University Medical School
proteus columbae, and the vector of the Chagas parasite, Rhodnius (Heidelberg, Germany), and the Malaria Molecular Laboratory, Mulago
prolixus.[155] These hemozoin molecules also show a similar struc- National Referral Hospital (Kampala, Uganda).
tural identity, which is advantageous concerning treatment, but
might potentially be challenging for diagnosis in case of a co-
infection.[155] Hemozoin production is further a hallmark of older Conflict of Interest
developmental stages of Plasmodium parasites that, in case of P. Brian Gitta is the C.E.O. of Matibabu and discloses financial interest.
falciparum, are able to sequester in a specific organ and can cause Nicole Kilian declares no conflict of interest.
a local hemozoin accumulation there.[15,156] These sequestered
parasites do not circulate in the peripheral blood of the patients
and remain undetected while juvenile ring stages, that have not Keywords
yet produced a significant amount of hemozoin, are also not con-
sidered during a hemozoin-based diagnostic procedure.[157] host-parasite interaction, infectious diseases, malaria, malaria diagnosis,
medical devices, Plasmodium

3. Conclusions and Outlook Received: August 5, 2019


Revised: November 5, 2019
Malaria is a curable disease: If you know you are infected. Published online: December 12, 2019

Many efforts are being made to understand the biology of Plas-


modium parasites and Anopheles mosquitoes to fight and erad-
icate a disease that has strongly shaped human evolution and [1] WHO, World Malaria Report 2018, WHO, Geneva, Switzerland 2018.
history. Early, fast, and accurate diagnosis of Plasmodium para- [2] A. Bartoloni, L. Zammarchi, Mediterr. J. Hematol. Infect. Dis. 2012,
sites is crucial for disease management and to avoid suboptimal 4, e2012026.
care that can lead to presumptive antimalarial over- or undertreat- [3] Z. Luo, S. A. Sullivan, J. M. Carlton, Ann. N. Y. Acad. Sci. 2015, 1342,
ment, causing the emergence of parasite drug resistance.[46,79] 53.
All presented diagnostic methods have advantages and disadvan- [4] A. Calderaro, G. Piccolo, C. Gorrini, S. Rossi, S. Montecchini, M. L.
tages and it is hard to determine which method will be the most Dell’Anna, F. De Conto, M. C. Medici, C. Chezzi, M. C. Arcangeletti,
promising in the future. Malar. J. 2013, 12, 321.
[5] A. Lalremruata, S. Jeyaraj, T. Engleitner, F. Joanny, A. Lang, S. Bélard,
The question is whether there will ever be a “perfect” method
G. Mombo-Ngoma, M. Ramharter, P. G. Kremsner, B. Mordmüller,
that consists of a device that is cheap to produce and easy to han- J. Held, Malar. J. 2017, 16, 398.
dle while providing sufficient sensitivity. This ideal device should [6] R. Zhou, S. Li, Y. Zhao, C. Yang, Y. Liu, D. Qian, H. Wang, D. Lu, H.
also be affordable for patients or hospitals, noninvasive, portable, Zhang, Sci. Rep. 2019, 9, 2191.
and with a potential to be applied at the patient’s home, given that [7] R. Tuteja, FEBS J. 2007, 274, 4670.
people living in rural areas might be too sick or do not have time [8] N. Hodson, B. Invergo, J. C. Rayner, J. S. Choudhary, Biochem. Soc.
or money to travel to a hospital. The journey towards simplify- Trans. 2015, 43, 240.
ing malaria diagnosis continues, and for now we have to use the [9] M. Cyrklaff, C. P. Sanchez, N. Kilian, C. Bisseye, J. Simpore, F.
available methods. However, more innovative ideas are coming Frischknecht, M. Lanzer, Science 2011, 334, 1283.
to the forefront in a bid to improve malaria diagnosis, as we have [10] N. Kilian, J. Y. Choi, D. R. Voelker, C. Ben Mamoun, J. Biol. Chem.
2018, 293, 17308.
reviewed here.
[11] B. L. Pasloske, R. J. Howard, Annu. Rev. Med. 1994, 45, 283.
Another important point is the documentation of malaria [12] N. A. Counihan, S. A. Chisholm, H. E. Bullen, A. Srivastava, P. R.
cases worldwide. This could be achieved with a platform that al- Sanders, T. K. Jonsdottir, G. E. Weiss, S. Ghosh, B. S. Crabb, D. J.
lows tracking of the geographical distribution and evolution of Creek, P. R. Gilson, T. F. de Koning-Ward, eLife 2017, 6.
cases to improve general disease surveillance and facilitate result [13] B. O. Abdulkareem, A. O. Adam, A. O. Ahmed, A. A. Mariam, U. U.
exchange among researchers and physicians in endemic areas. Samuel, J. Parasit. Dis. 2017, 41, 1093.
Time will tell whether the ultimate malaria test can be de- [14] N. Kilian, M. Dittmer, M. Cyrklaff, D. Ouermi, C. Bisseye, J. Simpore,
veloped and whether the observation of clinical cases will help F. Frischknecht, C. P. Sanchez, M. Lanzer, Cell. Microbiol. 2013, 15,
physicians and researchers monitor the current situation better 1111.
and provide adequate help and care. [15] N. Kilian, S. Srismith, M. Dittmer, D. Ouermi, C. Bisseye, J. Simpore,
M. Cyrklaff, C. P. Sanchez, M. Lanzer, Biol. Open 2015, 4, 400.
[16] Y. Adams, P. Kuhnrae, M. K. Higgins, A. Ghumra, Jra. Rowe, Infect.
Acknowledgements Immun. 2014, 82, 949.
[17] C. P. Sanchez, C. Karathanasis, R. Sanchez, M. Cyrklaff, J. Jäger, B.
The authors would like to thank the members of the Matibabu team and Buchholz, U. S. Schwarz, M. Heilemann, M. Lanzer, Commun. Biol.
Patrick Pitoniak for critically proofreading this manuscript. The authors 2019, 2, 172.

BioEssays 2020, 42, 1900138 1900138 (9 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

[18] P. R. Totino, S. C. Lopes, Front. Immunol. 2017, 8, 1148. [49] P. Galen, U. Gurkan, A. Fraiwan, Muhammad N. Hasan, D. E.
[19] K. K. Asare, M. Sakaguchi, A. B. Lucky, M. Asada, S. Miyazaki, Y. Grupp, J. K. Hoyt, J. Thorne, B. T. Grimberg (HEMEX HEALTH, INC.)
Katakai, S. Kawai, C. Song, K. Murata, K. Yahata, O. Kaneko, Para- 10375909, 2019.
sitol. Int. 2018, 67, 481. [50] B. Srivastava, A. R. Anvikar, S. K. Ghosh, N. Mishra, N. Kumar, A.
[20] A. B. Lucky, M. Sakaguchi, Y. Katakai, S. Kawai, K. Yahata, T. J. Tem- Houri-Yafin, J. J. Pollak, S. J. Salpeter, N. Valecha, Malar. J. 2015, 14,
pleton, O. Kaneko, PLoS One 2016, 11, e0164272. 526.
[21] F. A. Fatih, A. Siner, A. Ahmed, L. Woon, A. G. Craig, B. Singh, S. [51] I. Buchwalow, W. Boecker, M. Tiemann, Virchows Arch. 2015, 466,
Krishna, J. Cox-Singh, Malar. J. 2012, 11, 33. 111.
[22] B. E. Barber, M. J. Grigg, K. A. Piera, T. William, D. J. Cooper, K. [52] D. Romanowsky, St. Petersburg Med. Wochenschr. 1891, 16, 297.
Plewes, A. M. Dondorp, T. W. Yeo, N. M. Anstey, Emerg. Microbes [53] B. Fleischer, Trop. Med. Int. Health 2004, 9, 755.
Infect. 2018, 7, 1. [54] G. Giemsa, Centralblatt für Bakteriologie I Abteilung 1904, 32, 307.
[23] S. Akinyi, E. Hanssen, E. V. S. Meyer, J. Jiang, C. C. Korir, B. Singh, S. [55] A. P. Norgan, H. E. Arguello, L. M. Sloan, E. C. Fernholz, B. S. Pritt,
Lapp, J. W. Barnwell, L. Tilley, M. R. Galinski, Mol. Microbiol. 2012, Malar. J. 2013, 12, 231.
84, 816. [56] P. Bejon, L. Andrews, A. Hunt-Cooke, F. Sanderson, S. C. Gilbert, A.
[24] J. M. Chavatte, S. B. Tan, G. Snounou, R. T. Lin, Malar. J. 2015, 14, V. Hill, Malar. J. 2006, 5, 104.
454. [57] E. M. Bloch, Z. Mrango, M. Kasubi, J. Weaver, A. Mihailovic, B.
[25] E. Mundwiler-Pachlatko, H. P. Beck, Proc. Natl. Acad. Sci. USA 2013, Munoz, A. Weimer, A. Levin, L. Tonnetti, J. M. Linnen, V. Brès, D. E.
110, 19987. Norris, G. Carpi, S. K. West, PLoS Negl. Trop. Dis. 2019, 13, e0007632.
[26] B. Autino, Y. Corbett, F. Castelli, D. Taramelli, Mediterr. J. Hematol. [58] A. Jiram, C. H. Ooi, J. M. Rubio, S. Hisam, G. Karnan, N. M. Sukor,
Infect. Dis. 2012, 4, e2012061. M. M. Artic, N. P. Ismail, N. W. Alias, Malar. J. 2019, 18, 156.
[27] R. Udomsanpetch, K. Thanikkul, S. Pukrittayakamee, N. J. White, [59] M. Niang, L. G. Thiam, R. Sane, N. Diagne, C. Talla, S. Doucoure,
Trans. R. Soc. Trop. Med. Hyg. 1995, 89, 635. J. Faye, F. Diop, A. Badiane, B. Diouf, D. Camara, F. Diene-
[28] K. Silamut, R. Udomsangpetch, K. Thanikkul, N. J. White, B. J. An- Sarr, C. Sokhna, V. Richard, A. Toure-Balde, PLoS One 2017, 12,
gus, Am. J. Trop. Med. Hyg. 1996, 55, 560. e0182189.
[29] M. Edison, J. B. Jeeva, M. Singh, Indian J. Exp. Biol. 2011, 49, 11. [60] K. M. Fornace, N. Nuin, M. Betson, M. J. Grigg, T. William, N. M.
[30] A. G. Craig, M. F. Khairul, P. R. Patil, Malays. J. Med. Sci. 2012, 19, 5. Anstey, T. W. Yeo, J. Cox, L. T. Ying, C. J. Drakeley, J. Infect. Dis. 2016,
[31] B. Rahimi, A. Thakkinstian, N. J. White, C. Sirivichayakul, A. M. Don- 213, 784.
dorp, W. Chokejindachai, Malar. J. 2014, 13, 481. [61] W. R. Prescott, R. G. Jordan, M. P. Grobusch, V. M. Chinchilli, I. Klein-
[32] G. S. Rajahram, D. J. Cooper, T. William, M. J. Grigg, N. M. Anstey, schmidt, J. Borovsky, M. Plaskow, M. Torrez, M. Mico, C. Schwabe,
B. E. Barber, Clin. Infect. Dis. 2019, 69, 1703. Malar. J. 2012, 11, 155.
[33] N. J. White, S. Pukrittayakamee, T. T. Hien, Mul. Faiz, O. A. Mokuolu, [62] J. J. Pollak, A. Houri-Yafin, S. J. Salpeter, Front. Public Health 2017,
A. M. Dondorp, Lancet 2014, 383, 723. 5, 219.
[34] Institute of Medicine (US) Committee on the Economics of Anti- [63] K. Torres, C. M. Bachman, C. B. Delahunt, J. Alarcon Baldeon, F.
malarial Drugs in Saving Lives, Buying Time: Economics of Malaria Alava, D. Gamboa Vilela, S. Proux, C. Mehanian, S. K. McGuire, C.
Drugs in an Age of Resistance, (Eds: K. J. Arrow, C. Panosian, H. Gel- M. Thompson, T. Ostbye, L. Hu, M. S. Jaiswal, V. M. Hunt, D. Bell,
band), National Academies Press, Washington, DC 2004. Malar. J. 2018, 17, 339.
[35] L. J. Bruce-Chwatt, J. R. Soc. Med. 1981, 74, 531. [64] L. Florin, K. Maelegheer, A. Muyldermans, M. Van Esbroeck, E. Nu-
[36] F. E. Cox, Parasit. Vectors 2010, 3, 5. lens, J. Emmerechts, Diagn. Microbiol. Infect. Dis. 2018, 90, 253.
[37] J. Achan, A. O. Talisuna, A. Erhart, A. Yeka, J. K. Tibenderana, F. [65] L. Rickman, R. Oberst, R. Sangalang, J. Chulay, G. Long, A. Caban-
N. Baliraine, P. J. Rosenthal, U. D’Alessandro, Malar. J. 2011, 10, ban, J. Smith, S. Hoffman, Lancet 1989, 333, 68.
144. [66] D. Lenz, P. G. Kremsner, B. Lell, B. Biallas, M. Boettcher, B. Mord-
[38] A. Laveran, Bull. Mem. Soc. Med. Hop. Paris. 1881, 17,158. müller, A. A. Adegnika, Malar. J. 2011, 10, 194.
[39] M. Olivier, K. Van Den Ham, M. T. Shio, F. A. Kassa, S. Fougeray, [67] M. Kimura, I. Teramoto, C. W. Chan, Z. M. Idris, J. Kongere, W. Ka-
Front. Immunol. 2014, 5, 25. gaya, F. Kawamoto, R. Asada, R. Isozumi, A. Kaneko, Malar. J. 2018,
[40] E. Capanna, Int. Microbiol. 2006, 9, 69. 17, 72.
[41] R. Richardson, Lancet 2011, 377, 545. [68] J. Fore, Jr., I. R. Wiechers, R. Cook-Deegan, J. Biomed. Discov. Collab.
[42] L. Dembele, J.-F. Franetich, A. Lorthiois, A. Gego, A.-M. Zeeman, 2006, 1, 7.
C. H. M. Kocken, R. Le Grand, N. Dereuddre-Bosquet, G.-J. van [69] K. Mullis, F. Faloona, S. Scharf, R. Saiki, G. Horn, H. Erlich, Cold
Gemert, R. Sauerwein, J.-C. Vaillant, L. Hannoun, M. J. Fuchter, T. T. Spring Harb. Symp. Quant. Biol. 1986, 51, 263.
Diagana, N. A. Malmquist, A. Scherf, G. Snounou, D. Mazier, Nat. [70] P. Berzosa, A. de Lucio, M. Romay-Barja, Z. Herrador, V. González,
Med. 2014, 20, 307. L. García, A. Fernández-Martínez, M. Santana-Morales, P. Ncogo, B.
[43] V. Joste, C. Kamaliddin, E. Kendjo, V. Hubert, N. Argy, S. Houzé, Sci. Valladares, M. Riloha, A. Benito, Malar. J. 2018, 17, 333.
Rep. 2018, 8, 300. [71] M. Rougemont, M. Van Saanen, R. Sahli, H. P. Hinrikson, J. Bille, K.
[44] C. Koepfli, W. Nguitragool, N. E. Hofmann, L. J. Robinson, M. Jaton, J. Clin. Microbiol. 2004, 42, 5636.
Ome-Kaius, J. Sattabongkot, I. Felger, I. Mueller, Sci. Rep. 2016, 6, [72] Y. M. Lloyd, L. F. Esemu, J. Antallan, B. Thomas, S. Tassi Yunga, B.
39183. Obase, N. Christine, R. G. F. Leke, R. Culleton, K. O. Mfuh, V. R.
[45] C. Koepfli, L. J. Robinson, P. Rarau, M. Salib, N. Sambale, R. Nerurkar, D. W. Taylor, Trop. Med. Health 2018, 46, 22.
Wampfler, I. Betuela, W. Nuitragool, A. E. Barry, P. Siba, I. Felger, [73] N. Van Hong, P. van den Eede, C. Van Overmeir, I. Vythilingham,
I. Mueller, PLoS One 2015, 10, e0126747. A. Rosanas-Urgell, P. Vinh Thanh, N. D. Thang, N. M. Hung, L. X.
[46] V. N. Orish, J. Y. Ansong, O. S. Onyeabor, A. O. Sanyaolu, W. A. Hung, U. D’Alessandro, A. Erhart, Am. J. Trop. Med. Hyg. 2013, 89,
Oyibo, N. C. Iriemenam, Trop. Doct. 2016, 46, 191. 721.
[47] Z. Bisoffi, F. Gobbi, D. Buonfrate, J. Van den Ende, Mediterr. J. Hema- [74] K. Komaki-Yasuda, J. P. Vincent, M. Nakatsu, Y. Kato, N. Ohmagari,
tol. Infect. Dis. 2012, 4, e2012036. S. Kano, PLoS One 2018, 13, e0191886.
[48] O. O. Oladosu, W. A. Oyibo, ISRN Infect. Dis. 2013, 2013, 1.

BioEssays 2020, 42, 1900138 1900138 (10 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

[75] H. Al-Shehri, Bne. Power, J. Archer, A. Cousins, A. Atuhaire, M. [98] J. P. Robinson, B. Rajwa, V. Patsekin, V. J. Davisson. Expert Opin.
Adriko, M. Arinaitwe, A. D. Alanazi, Ees. LaCourse, N. B. Kabatere- Drug Discov. 2012, 7, 679.
ine, Jll. Stothard, Malar. J. 2019, 18, 109. [99] B. Malleret, C. Claser, A. S. M. Ong, R. Suwanarusk, K. Sriprawat,
[76] WWARN K13 Genotype-Phenotype Study Group, BMC Med. 2019, S. W. Howland, B. Russell, F. Nosten, L. Rénia, Sci. Rep. 2011, 1,
17, 1. 118.
[77] N. Chea, M. Ken, P. Bosman, N. Khim, W. Etienne, D. Ménard, P. [100] E. Pillay, S. Khodaiji, B. C. Bezuidenhout, M. Litshie, T. L. Coetzer,
Pannus, L. Canier, C. Khean, S. Alipon, M. C. Char, S. Kim, K. Loch, Malar. J. 2019, 18, 15.
J. Stassijns, M. De Smet, R. Eam, F. Nackers, J.-M. Kindermans, Am. [101] A. Post, B. Kabore, I. J. Reuling, J. Bognini, W. van der Heijden, S.
J. Trop. Med. Hyg. 2015, 92, 573. Diallo, P. Lompo, B. Kam, N. Herssens, K. Lanke, T. Bousema, R. W.
[78] B. Wihokhoen, A. Dondorp, P. Turner, C. J. Woodrow, M. Imwong, Sauerwein, H. Tinto, J. Jacobs, Q. de Mast, A. J. van der Ven, BMC
Am. J. Trop. Med. Hyg. 2016, 94, 322. Med. 2019, 17, 103.
[79] K. O. Mfuh, O. A. Achonduh-Atijegbe, O. N. Bekindaka, L. F. Esemu, [102] W. Roobsoong, S. P. Maher, N. Rachaphaew, S. J. Barnes, K. C.
C. D. Mbakop, K. Gandhi, R. G. F. Leke, D. W. Taylor, V. R. Nerurkar, Williamson, J. Sattabongkot, J. H. Adams, Malar. J. 2014, 13, 55.
Malar. J. 2019, 18, 73. [103] D. Yang, G. Subramanian, J. Duan, S. Gao, L. Bai, R. Chandramo-
[80] S. Kasetsirikul, J. Buranapong, W. Srituravanich, M. Kaewthama- hanadas, Y. Ai, PLoS One 2017, 12, e0179161.
sorn, A. Pimpin, Malar. J. 2016, 15, 358. [104] P. H. van Vianen, A. van Engen, S. Thaithong, M. van der Keur, H. J.
[81] J. Simam, M. Rono, J. Ngoi, M. Nyonda, S. Mok, K. Marsh, Z. Tanke, H. J. van der Kaay, B. Mons, C. J. Janse, Cytometry 1993, 14,
Bozdech, M. Mackinnon, BMC Genomics 2018, 19, 372. 276.
[82] R. C. P. Rampazzo, A. C. Graziani, K. K. Leite, J. A. Surdi, C. A. [105] C. Cai, K. A. Carey, D. A. Nedosekin, Y. A. Menyaev, M. Sarimol-
Biondo, M. L. N. Costa, T. Jacomasso, M. Cereda, M. De Fazio, M. laoglu, E. I. Galanzha, J. S. Stumhofer, V. P. Zharov, Cytometry A
A. Bianchessi, O. C. Moreira, C. Britto, J. D. N. Costa, V. M. Góes, 2016, 89, 531.
A. J. da Silva, M. A. Krieger, A. D. T. Costa, J. Mol. Diagn. 2019, 21, [106] E. J. Fong, C. Huang, J. Hamilton, W. J. Benett, M. Bora, A. Burklund,
839. T. R. Metz, M. Shusteff, J. Vis. Exp. 2015.
[83] B. J. Hindson, K. D. Ness, D. A. Masquelier, P. Belgrader, N. J. Here- [107] H. T. Nguyen, H. Thach, E. Roy, K. Huynh, C. M.-T. Perrault, Micro-
dia, A. J. Makarewicz, I. J. Bright, M. Y. Lucero, A. L. Hiddessen, T. machines 2018, 9, 461.
C. Legler, T. K. Kitano, M. R. Hodel, J. F. Petersen, P. W. Wyatt, E. R. [108] J. Reboud, G. Xu, A. Garrett, M. Adriko, Z. Yang, E. M. Tukahebwa,
Steenblock, P. H. Shah, L. J. Bousse, C. B. Troup, J. C. Mellen, D. K. C. Rowell, J. M. Cooper, Proc. Natl. Acad. Sci. USA 2019, 116, 4834.
Wittmann, N. G. Erndt, T. H. Cauley, R. T. Koehler, A. P. So, S. Dube, [109] A. A. S. Bhagat, H. W. Hou, L. D. Li, C. T. Lim, J. Han, Lab Chip 2011,
K. A. Rose, L. Montesclaros, S. Wang, D. P. Stumbo, S. P. Hodges, 11, 1870.
S. Romine, F. P. Milanovich, H. E. White, J. F. Regan, G. A. Karlin- [110] H. W. Hou, A. A. S. Bhagat, A. G. Lin Chong, P. Mao, K. S. Wei Tan,
Neumann, C. M. Hindson, S. Saxonov, B. W. Colston, Anal. Chem. J. Han, C. T. Lim, Lab Chip 2010, 10, 2605.
2011, 83, 8604. [111] G. Xu, D. Nolder, J. Reboud, M. C. Oguike, D. A. van Schalkwyk, C.
[84] L. B. Pinheiro, V. A. Coleman, C. M. Hindson, J. Herrmann, B. J. J. Sutherland, J. M. Cooper, Angew. Chem. Int. Ed. 2016, 55, 15250.
Hindson, S. Bhat, K. R. Emslie, Anal. Chem. 2012, 84, 1003. [112] Y.-W. Cheung, J. Kwok, A. W. L. Law, R. M. Watt, M. Kotaka, J. A.
[85] A. M. Vasquez, L. Zuluaga, A. Tobon, M. Posada, G. Vélez, I. J. Tanner, Proc. Natl. Acad. Sci. USA 2013, 110, 15967.
González, A. Campillo, X. Ding, Malar. J. 2018, 17, 262. [113] Y. Zhang, B. S. Lai, M. Juhas. Molecules 2019, 24, 941.
[86] WHO, FIND, CDC, Malaria Rapid Diagnostic Test Performance [114] G. Figueroa-Miranda, L. Feng, S. C.-C. Shiu, R. M. Dirkzwager, Y.-W.
Summary results of WHO product testing of malaria RDTs: round Cheung, J. A. Tanner, M. J. Schöning, A. Offenhäusser, D. Mayer,
1-8 (2008–2018), WHO, Geneva, Switzerland 2018. Sens. Actuators B 2018, 255, 235.
[87] D. Berthod, J. Rochat, R. Voumard, L. Rochat, B. Genton, V. [115] H. Kaur, J. G. Bruno, A. Kumar, T. K. Sharma, Theranostics 2018, 8,
D’Acremont, Malar. J. 2017, 16, 436. 4016.
[88] M. R. Hoque, M. M. A. Elfaki, M. A. Ahmed, S.-K. Lee, F. Muh, [116] K.-A. Frith, R. Fogel, J. Dean. Goldring, R. G. E. Krause, M. Khati, H.
M. M. Ali Albsheer, M. M. A. Hamid, E.-T. Han, Malar. J. 2018, 17, Hoppe, M. E. Cromhout, M. Jiwaji, J. L. Limson, Malar. J. 2018, 17,
297. 191.
[89] R. Megnekou, J. C. Djontu, B. C. Nana, J. D. Bigoga, M. Fotso, B. [117] D. F. Joseph, J. A. Nakamoto, O. A. Garcia Ruiz, K. Penaranda,
Fogang, R. F. G. Leke, Malar. J. 2018, 17, 450. A. E. Sanchez-Castro, P. S. Castillo, P. Milón, PLoS One 2019, 14,
[90] S. Samadoulougou, F. Kirakoya-Samadoulougou, S. Sarrassat, H. e0211756.
Tinto, F. Bakiono, I. Nebié, A. Robert, Malar. J. 2014, 13, 101. [118] A. Barfod, T. Persson, J. Lindh, Parasitol. Res. 2009, 105, 1557.
[91] J. Tang, F. Tang, H. Zhu, F. Lu, S. Xu, Y. Cao, Y. Gu, X. He, H. Zhou, [119] P. Jain, B. Chakma, N. K. Singh, S. Patra, P. Goswami, Mol. Biotech-
G. Zhu, J. Cao, PLoS Negl. Trop. Dis. 2019, 13, e0007254. nol. 2016, 58, 497.
[92] S. Krishna, P. K. Bharti, H. S. Chandel, A. Ahmad, R. Kumar, P. P. [120] P. Jain, S. Das, B. Chakma, P. Goswami. Anal. Biochem. 2016, 514,
Singh, M. P. Singh, N. Singh, Emerg. Infect. Dis. 2015, 21, 1853. 32.
[93] A. Ahmad, A. K. Verma, S. Krishna, A. Sharma, N. Singh, P. K. Bharti, [121] J. Ospina-Villa, C. López-Camarillo, C. A. Castañón-Sánchez, J. Soto-
PLoS One 2019, 14, e0218210. Sánchez, E. Ramírez-Moreno, L. Marchat, Genes 2018, 9, 584.
[94] P. Simpalipan, S. Pattaradilokrat, P. Harnyuttanakorn, Malar. J. 2018, [122] N. K. Singh, P. D. Thungon, P. Estrela, P. Goswami, Biosens. Bioelec-
17, 16. tron. 2019, 123, 30.
[95] A. M. Vasquez, A. C. Medina, A. Tobon-Castano, M. Posada, G. [123] WHO, 2018. World Malaria Report 2018.
J. Vélez, A. Campillo, I. J. González, X. Ding, PLoS One 2018, 13, [124] S. J. Draper, B. K. Sack, C. R. King, C. M. Nielsen, J. C. Rayner, M. K.
e0201769. Higgins, C. A. Long, R. A. Seder, Cell Host Microbe 2018, 24, 43.
[96] N. Tangpukdee, C. Duangdee, P. Wilairatana, S. Krudsood, Korean J. [125] A. X. Mo, G. McGugan, The American Journal of Tropical Medicine
Parasitol. 2009, 47, 93. and Hygiene 2018, 99, 827.
[97] C. Doderer, A. Heschung, P. Guntz, J.-P. Cazenave, Y. Hansmann, A. [126] J. W. Bennett, A. Yadava, D. Tosh, J. Sattabongkot, J. Komisar, L.
Senegas, A. W. Pfaff, T. Abdelrahman, E. Candolfi, Malar. J. 2007, 6, A. Ware, W. F. McCarthy, J. J. Cowden, J. Regules, M. D. Spring, K.
19. Paolino, J. D. Hartzell, J. F. Cummings, T. L. Richie, J. Lumsden, E.

BioEssays 2020, 42, 1900138 1900138 (11 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc
www.advancedsciencenews.com www.bioessays-journal.com

Kamau, J. Murphy, C. Lee, F. Parekh, A. Birkett, J. Cohen, W. R. Ballou, [138] W. A. Oyibo, N. Ezeigwe, G. Ntadom, O. O. Oladosu, K. Rainwater-
M. E. Polhemus, Y. F. Vanloubbeeck, J. Vekemans, C. F. Ockenhouse, Loveth, W. O’Meara, E. Okpokoro, W. Brieger, J. Clin. Microbiol.
PLoS Negl. Trop. Dis. 2016, 10, e0004423. 2017, 55, 253.
[127] K. Bauza, T. Malinauskas, C. Pfander, B. Anar, E. Y. Jones, O. [139] B. C. Windom, D. W. Hahn, in Encyclopedia of Tribology (Eds: Q. J.
Billker, A. V. Hill, A. Reyes-Sandoval, Infect. Immun. 2014, 82, Wang, Y.-W. Chung), Springer, Boston, MA 2013, pp. 2742-2747.
1277. [140] B. R. Masters, Opt. Photonics News, 20, 2009.
[128] A. M. Salman, E. Montoya-Diaz, H. West, A. Lall, E. Atcheson, C. [141] K. Eberhardt, C. Stiebing, C. Matthaus, M. Schmitt, J. Popp, Expert
Lopez-Camacho, J. Ramesar, K. Bauza, K. A. Collins, F. Brod, F. Reis, Rev. Mol. Diagn. 2015, 15, 773.
L. Pappas, L. González-Cerón, C. J. Janse, A. V. S. Hill, S. M. Khan, [142] K. Chen, C. Yuen, Y. Aniweh, P. Preiser, Q. Liu, Sci. Rep. 2017, 7,
A. Reyes-Sandoval, Sci. Rep. 2017, 7, 46482. 44511.
[129] E. Atcheson, K. Bauza, A. M. Salman, E. Alves, J. Blight, M. E. [143] L. Frame, J. Brewer, R. Lee, K. Faulds, D. Graham, Analyst 2017, 143,
Viveros-Sandoval, C. J. Janse, S. M. Khan, A. V. S. Hill, A. Reyes- 157.
Sandoval, Infect. Immun. 2018, 86. [144] F. Chen, B. R. Flaherty, C. E. Cohen, D. S. Peterson, Y. Zhao,
[130] K. Singh, P. Mukherjee, A. R. Shakri, A. Singh, G. Pandey, M. Bakshi, Nanomedicine 2016, 12, 1445.
G. Uppal, R. Jena, A. Rawat, P. Kumar, R. Bhardwaj, S. S. Yazdani, [145] I. Pence, A. Mahadevan-Jansen, Chem. Soc. Rev. 2016, 45, 1958.
D. Hans, S. Mehta, A. Srinivasan, K. Anil, R. L. Madhusudhan, J. [146] D. C. Gowda, X. Wu, Front. Immunol. 2018, 9, 3006.
Patel, A. Singh, R. Rao, S. Gangireddy, R. Patil, S. Kaviraj, S. Singh, [147] O. Rifaie-Graham, J. Pollard, S. Raccio, S. Balog, S. Rusch, M. A.
D. Carter, S. Reed, D. C. Kaslow, A. Birkett, V. S. Chauhan, C. E. Hernández-Castañeda, P.-Y. Mantel, H.-P. Beck, N. Bruns, Nat. Com-
Chitnis, npj Vaccines 2018, 3, 48. mun. 2019, 10, 1369.
[131] WHO, 2018. Overview of malaria treatment. [148] J. L. Burnett, J. L. Carns, R. Richards-Kortum, Malar. J. 2017, 16, 447.
[132] K. O. Mfuh, S. Tassi Yunga, L. F. Esemu, O. N. Bekindaka, J. Yonga, J. [149] M. Christner, H. Frickmann, E. Klupp, H. Rohde, M. Kono, E. Tan-
C. Djontu, C. D. Mbakop, D. W. Taylor, V. R. Nerurkar, R. G. F. Leke, nich, S. Poppert, J. Microbiol. Methods 2019, 160, 104.
Malar. J. 2017, 16, 434. [150] B. T. Grimberg, K. O. Grimberg, Expert Rev. Anti Infect. Ther. 2016,
[133] D. Tao, B. McGill, T. Hamerly, T. Kobayashi, P. Khare, A. Dziedzic, 14, 879.
T. Leski, A. Holtz, B. Shull, A. E. Jedlicka, A. Walzer, P. D. Slowey, [151] R. J. Maude, W. Buapetch, K. Silamut, Am. J. Trop. Med. Hyg. 2009,
C. C. Slowey, S. E. Nsango, D. A. Stenger, M. Chaponda, M. Mu- 81, 782.
lenga, K. H. Jacobsen, D. J. Sullivan, S. J. Ryan, R. Ansumana, W. [152] P. A. Demirev, Expert Rev. Mol. Diagn. 2004, 4, 821.
J. Moss, I. Morlais, R. R. Dinglasan, Sci. Transl. Med. 2019, 11, [153] P. A. Zimmerman, J. M. Thomson, H. Fujioka, W. E. Collins, W. E.
pii: eaan4479. Collins, Am. J. Trop. Med. Hyg. 2006, 74, 568.
[134] M. S. Hede, S. Fjelstrup, F. Lotsch, R. Zoleko, A. Klicpera, M. [154] A. Butykai, A. Orbán, V. Kocsis, D. Szaller, S. Bordács, E. Tátrai-
Groger, J. Mischlinger, L. Endame, L. Veletzky, R. Neher, A. K. W. Szekeres, L. F. Kiss, A. Bóta, B. G. Vértessy, T. Zelles, I. Kézsmárki,
Simonsen, E. Petersen, G. Mombo-Ngoma, M. Stougaard, Y.-P. Sci. Rep. 2013, 3, 1431.
Ho, R. Labouriau, M. Ramharter, B. R. Knudsen, Sci. Rep. 2018, 8, [155] M. F. Oliveira, S. W. Kycia, A. Gomez, A. J. Kosar, D. Scott Bohle, E.
4122. Hempelmann, D. Menezes, M. A. Vannier-Santos, P. L. Oliveira, S.
[135] M. Jirku, K. Pomajbíková, K. J. Petrželková, Z. Huzova, D. Modrý, J. T. Ferreira, FEBS Lett. 2005, 579, 6010.
Lukešet, Emerg. Infect. Dis. 2012, 18, 634. [156] R. McGready, A. Brockman, T. Cho, M. A. Levesque, A. N. Tkachuk,
[136] C. Putaporntip, P. Buppan, S. Jongwutiwes, Clin. Microbiol. Infect. S. R. Meshnick, F. Nosten, Trans. R. Soc. Trop. Med. Hyg. 2002, 96,
2011, 17, 1484. 644.
[137] A. Tobon-Castano, S. Barrera Escobar, C. Giraldo Castro, J. Trop. [157] L. M. Coronado, C. T. Nadovich, C. Spadafora, Biochim. Biophys. Acta
Med. 2017, 2017, 1. Gen. Subj. 2014, 1840, 2032.

BioEssays 2020, 42, 1900138 1900138 (12 of 12) © 2019 The Authors. BioEssays Published by Wiley Periodicals, Inc

You might also like