Вы находитесь на странице: 1из 23

CHAPTER ONE

1.0 INTRODUCTION

Malaria remains a disease of public health concern in tropical countries (WHO, 2014). Malaria is

one of the major killer diseases affecting most tropical countries especially in Africa. It affects

over 500 million people worldwide and over one million children die annually from malaria

(Nandwania, 2018). Of all the human malaria parasite, Plasmodium, is the most pathogenic and

is frequently fatal if untreated in good time. In Nigeria, according to Nandwania (2019), total of

1.82 million cases of malaria and 0.89 million cases of Plasmodium falciparum cases with 902

death were reported in the year 2018. The diagnosis of malaria in tropical countries largely

depends on clinical assessment, microscopy, and recently, by rapid diagnostic tests (RDTs)

(Wongsrichanalai et al., 20l3)

Microscopy is considered the gold standard for diagnosis of malaria (WHO, 2010).

Affordability, low-threshold sensitivity, identification of species, and parasite density gives it an

edge over RDT (Makler, 1999). However, long turnaround time, erratic power supply, and need

for the technical expertise are some of the major challenges limiting its use in resource limited

countries where the burden of malaria is greatest (Makler, 1999). Rapid diagnostic tests on the

other hand, was developed to improve the sensitivity and objectivity of malaria diagnosis

through less reliance on microscopy. It is an immune-chromatographic capture procedure which

targets antigens abundant in all asexual and sexual stages of the parasite. Current RDTs detect

histidin rich protein 2 (Pfl-IRP2) from Plasmodiun falciparum and parasite-specific lactate

dehydrogenase (pLDH) or Plasmodium aldolase from the parasite glycolytic pathway found in

all species (Lon et al., 2015).

Traditional practice for outpatients has been to treat presumptively for malaria based on a history

of fever but, a significant proportion of those treated may not have parasites (over 50% in many

1
settings) and hence waste a considerable amount of drugs. This old clinical based practice is still

relevant today especially, in infants where time spent on getting a confirmatory laboratory

diagnosis could lead to increased fatality (Lon et al., 2015).

World Health Organization currently makes the tentative recommendation that parasite-based

diagnosis should be used in all cases of suspected malaria with the possible exception of children

in high-prevalence areas and certain other situations. For this recommendation to be adhered to,

rapid and accurate laboratory finding or demonstration of malaria parasite should be established

(Mouatcho and Goldring, 2013).

The traditional method of microscopic identification of parasite, however, is not only daunting in

poor power setting, but also time consuming and requiring a lot of expertise/training. Thus,

microscopy in Africa is generally, limited to larger clinics/tertiary centres. This conventional

staining of peripheral blood smears/microscopy, however, still remains the gold standard in

laboratory diagnosis of malaria. Rapid diagnostic tests (RDTs) for malaria could be considered

for most patients in endemic regions, especially in poor power settings where there is shortage of

qualified manpower in Africa. However, there is very little evidence, especially from malaria

endemic areas to guide decision-makers on the sensitivity and specificity of these RDTs

(Mouatcho and Goldring, 2013).

Rapid iagnosicfest performance can be affected by factors which include short shelf life because

of temperature instability, especially during transportation from the place of manufacturing or

supply to final place of use, suboptimal sensitivity at low.

Parasite densities and these RDTs Kits are species-specific or quantify parasite density.

Manufacturers recommend an optimal temperature range of 4-30°C. The pLDH-detecting RDTs

produced a consistent level of parasite lactate when maintained at 4°C, but activity is reduced

2
after cumulative exposure to temperatures likely to be encountered over a few months in a

malaria-endemic area (Lon et al., 2005).

1.1 Statement of the Research Problem

There have been cases of misdiagnosis of malaria in some healthcare facilities and it results. in

treatment failure in patience. For effective management of disease like malaria, accurate

diagnosis is imperative.

1.2 Justification for the Study

Malaria is an overwhelming problem in tropical developing countries, It is currently the most

important human parasitic disease in the world and continues to be a serious public health

problem. Effective diagnostic techniques permit early and appropriate diagnosis of malaria since

this prevents the infection from progressing to a severe stage or even death

It is very important to make a definitive diagnosis and confirm malaria in a

population. Although microscopy remains the gold standard for malaria diagnosis and

is faster than molecular techniques, it has major limitations such as low sensitivity,

difficulties associated with quality control and standardization and the need for ongoing training

and evaluation.

1.3 Aim of the Study

The aim of the study was to determine the prevalence of malaria in people residing in New

Kuchingoro Internaly Displaced Persons Camp Abuja, Nigeria.

3
1.4 Specific Objectives

The specific objectives of this study were to:

1. Determine the prevalence of malaria in IDP Camp using rapid diagnosis test kits

2. Determine the prevalence of malaria among people living in IDP camps in Kuchingoro in

relation to some socio-demographic factors, namely, age and sex.

4
CHAPTER TWO

LITERATURE REVIEW

2.1 Malaria

Malaria is a mosquito-borne infectious disease that affects humans and other animals.”.4alaria

causes symptoms that typically include fever, tiredness, vomiting, and headaches. In severe cases

it can cause yellow skin, seizures, coma, and death. symptoms usually begin 10 to 15 days after

an individua bitten by an infected mosquito, if not properly treated, people may have recurrences

of the disease months later\In those who have recently survived an infection, reinfection usually

causes milder symptoms. This partial resistance disappears over months to years if the person

has no continuing exposure to malaria (Silas, 2016).

Malaria is caused by single-cell called parasites of the Plasmodium group. The disease is most

commonly spread by an infected female Anopheles mosquito. The mosquito bite introduces the

parasites from the mosquito’s saliva into a person’s blood (John, 2014).

The parasites travel to the liver where they mature and reproduce. Five species of Plasmodium

can infect and be spread by humans. Most deaths are caused by P. falciparum, because P. vivax,

P. ovale, and P. malariae generally cause a milder form of malaria.

The species P. knowlesi rarely causes disease in humans but in animals notably,

primates. Malaria is typically diagnosed by the microscopic examination of blood

using blood films, or with antigen-based rapid diagnostic Kits. Methods that use the polymerase

chain reaction to detect the parasite’s DNA have been developed, but are not widely used in

areas where malaria is common due to their cost and complexity (Tijani, 2014).

5
2.2 Diagnoses of malaria

Malaria is an overwhelming problem in tropical developing countries, It is currently the most

important human parasitic disease in the world and continues to be a serious public health

problem. More than 85% of malaria cases and 90% of malaria deaths occur in sub-Saharan

Africa, mainly in young children (especially those under 5 years old) (John, 20l6) Effective

diagnostic techniques permit early and appropriate diagnosis of malaria since this prevents the

infection from progressing to a severe stage or even death. The World Health Organization

(WHO) recommends confirming all clinically suspected malaria cases before treatment using a

malaria-specific rapid diagnostic test (RDT) or by direct visualization of the parasites using

standard microscopy (Makler, 1999).

Malaria must be recognized promptly in order to treat the patient in time and to

prevent further spread of infection in the community via local rnosquitoes (Silas,

2016).

Malaria should be considered a potential medical emergency and should be

treated accordingly. Delay in diagnosis and treatment is a leading cause of death in malaria

patients in the United States. Malaria can be suspected based on the patient’s travel history,

symptoms, and the physical findings at examination. However, for a definitive diagnosis to be

made, laboratory tests must demonstrate the presence of malaria parasites or their component

(Silas, 2016).

Prompt and accurate diagnosis is critical to the effective management of malaria. The global

impact of malaria has spurred interest in developing effective diagnostic strategies not only for

6
resource-limited areas where malaria is a substantial burden on society, but also in developed

countries, where malaria diagnostic expertise is often lacking. Malaria diagnosis involves

identifying malaria parasites or antigens/products in patient blood. Although this may seem

simple, the diagnostic efficacy is subject to many factors. The different forms of the 5 malaria

species; the different stages of erythrocytichizogony, the endemicity of different species, the

interrelation between levels of tranmission, population movement, parasitemia, immunity, and

signs and symptoms; drug resistance, the problems of recurrent malaria, persisting viable or non-

viable parasitemia, and sequestration of the parasites in the deeper tissues, and the use of

chemoprophylaxis or even presumptive treatment on the basis of clinical diagnosis, can all

influence the identification and interpretation of malaria parasitemia in a diagnostic test (John,

2016).

Malaria is a potential medical emergency and should be treated accordingly. Delays in diagnosis

and treatment are leading causes of death in many countries. Diagnosis can

be difficult where malaria is no longer endemic for healthcare providers unfamiliar with the

disease. Clinicians may forget to consider malaria among the potential diagnoses for some

patients and not order the necessary diagnostic tests. Technicians

may be unfamiliar with, or lack experience with, malaria, and fail to detect parasites when

examining blood smears under a microscope. In some areas, malaria

transmission is so intense that a large proportion of the population is infected, but remains

asymptomatic.For example, in Africa, such carriers have developed sufficient immunity to

protect them from illness, but not infection. In such situations, finding malaria parasites in an ill

person does not necessarily mean that the illness is caused by the parasites. In many malaria-

endemic countries, the lack of resources is a major barrier to reliable and timely diagnosis.

Health personnel are undertrained, underequipped, and underpaid. They often face excessive

7
patient loads, and must divide their attention between malaria and other equally severe infectious

diseases, such as tuberculosis or HIV/AIDS (John, 2016).

2.3 Rapid Diagnostic Tests (RDTS)

Rapid diagnostic tests based(RDTs) malaria parasite antigen detection are now a key tool in the

case management of clinical malaria, especially at lower level peripheral health facilities where

routine microscopy is absent or of poor quality. The RDTs are also shown to be more cost-

effective in improving health outcomes than expert microscopy in most sub-Saharan African

settings. In addition to their clinical use, RDTs are increasingly being used in epidemiologic

surveys of Plasmodium spp. infection as part of national monitoring and evaluation efforts. For

example, of the 27 recent national malaria indicator surveys conducted in sub-Saharan Africa

since 2006, RDTs were used in 19 surveys, and in 3 of these surveys Plasmodium species

infection prevalence was estimated on the basis of RDTs alone (Phoebe, 2012) The use of RDTs

in large-scale surveys is preferable for therapeutic reasons because they

provide point-of-contact diagnosis and, if required, immediate treatment. Moreover, RDTs

overcome the human and technical capacity constraints faced by large-scale surveys in the use of

expert microscopy in terms o quality staining and reading of thousands of blood slides and the

logistics and costs associated with slide transportation, preparation, duplicate reading, and

quality assurance (Makler, 1999).

A well-recognized limitation of RDTs, especially those tests that detect the parasite antigen

histidine-rich protein 2 (HRP-2) specific to Plasmodium falciparum, is the occurrence of false-

positive results caused by persistent antigenemia even after effective anti-malarial

treatment.Although such false-positives results for RDTs may have limited relevance for clinical

8
case management, they will overestimate the true parasite prevalence compared with expert

microscopy or molecular parasite detection techniques (Peter, 2015).

2.4 Clinical Diagnosis of Malaria

A clinical diagnosis of malaria is traditional among medical doctors. This method is

least expensive and most widely practiced. Clinical diagnosis is based on the patients’ signs and

symptoms, and on physical findings at examination. The earliest symptoms

of malaria are very nonspecific and variable, and include fever, headache, weakness, myalgia,

chills, dizziness, abdominal pain, diarrhea, nausea, vomiting, anorexia, and pruritus (John, 2017).

A clinical diagnosis of malaria is still challenging because of the non-specific nature of the signs

and symptoms, which overlap considerably with other common, as well as potentially life-

threatening diseases, e.g. common viral or bacterial infections, and other febrile illnesses. The

overlapping of malaria symptoms with other tropical diseases impairs diagnostic specificity,

which can promote the indiscriminate use of antimalarials and compromise the quality of care for

patients with non-malarial fevers in endemic areas. The Integrated Management of Children

Illness (IMCI) has provided clinical algorithms for managing and diagnosing common childhood

illnesses by minimally trained healthcare providers in the developing world having inappropriate

equipment for laboratory diagnosis. A widely utilized clinical algorithm for malaria diagnosis,

compared with a fully trained pediatrician with access to laboratory support, showed very low

specificity (0-9%) but 100% sensitivity in African scttingspruritus (John, 2017). This lack of

specificity reveals the perils of distinguishing malaria from other causes of fever in children on

clinical grounds alone. Recently, another study showed that use of the IMCI clinical algorithm

resulted in 30% over-diagnosis of malaria. Therefore, the accuracy of malaria diagnosis can be

greatly enhanced by combining clinical-and parasite-based findings (Makler, 1999).

9
2.5 Laboratory Diagnosis of Malaria

Rapid and effective malaria diagnosis not only alleviates suffering, but also decreases

community transmission. The nonspecific nature of the clinical signs and symptoms of malaria

may result in over-treatment of malaria or non-treatment of other diseases in malaria-endemic

areas, and misdiagnosis in non-endemic areas. In the laboratory, malaria is diagnosed using

different techniques, e.g. conventional microscopic diagnosis by staining thin and thick

peripheral blood smears, other concentration techniques, e.g. quantitative buffy coat (QBC)

method,rapid diagnostic tests e.g., OptiMAL ICT, Para-HIT-f , ParaScreen, SD Bioline,

Paracheck, and molecular diagnostic methods, such as polymerase chain reaction (PCR). Some

advantages and shortcomings of these methods have also been described, related to sensitivity,

specificity, accuracy, precision, time consumed, cost-effectiveness, labor intensiveness, the need

for skilled microscopists, and the problem of inexperienced technicians (Phoebe, 2016)

2.5.1 Microscopic Diagnosis using stained thin and thick peripheral blood smears

Malaria is conventionally diagnosed by microscopic examination of stained blood films using

Giemsa, Wright’s or Field’s stains. This method has changed very little since Laverrans original

discovery of the malaria parasite, and improvements in staining techniques by Romanowsky in

the late 1980s. More than a century later, microscopic detection and identification of

Plasrnodium species in Giemsa-stained thick blood films (for screening the presenting malaria

parasite), and thin blood films (for species’ confirmation) remains the gold standard for

laboratory diagnosis. Malaria is diagnosed microscopically by staining thick and thin blood films

on a glass slide, to visualize malaria parasites. Briefly, the patient’s finger is cleaned with 70%

ethyl alcohol, allowed to dry and then the side of fingertip is picked with a sharp sterile

lancet and two drops of blood are placed on a glass slide. To prepare a thick blood

10
film, a blood spot is stirred in a circular motion with the corner of the slide, taking care not make

the preparation too thick, and allowed to dry without fixative (Silas,

20l6). After drying, the spot is stained with diluted Giemsa (1: 20, vol/vol) for 20 mm, and

washed by placing the film in buffered water for 3 mm. The slide is allowed to air-dry in a

vertical position and examination using a light microscope. As they are unfixed, the red cells lyse

when a water-based stain is applied. A thin blood film is prepared by immediately placing the

smooth edge of a spreader slide in a drop of blood, adjusting the angle between slide and

spreader to 45° and then smearing the blood with a swift and steady sweep along the surface.

The film is then allowed to air- dry and is fixed with absolute methanol. After drying, the

sample is stained with diluted Giemsa (1: 20, vol/vol) for 20 mm and washed by briefly dipping

the slide in and out of a jar of buffered water (excessive washing will decolorize the film). The

slide is then allowed to air-dry in a vertical position and examined under a light microscope

(Silas, 201 6). The wide acceptance of this technique by laboratories all around the world can be

attributed to its simplicity, low cost, its ability to identify the presence of parasites, the infecting

species, and assess parasite density-all parameters useful for the management of malaria.

Recently, a study showed that conventional malaria microscopic diagnosis at primary healthcare

facilities in Tanzania could reduce the prescription of antimalarial drugs, and also appeared to

improve the appropriate management of non-malarial, fevers (Lon et al,, 2005). However, the

staining and interpretation processes are labor intensive, time consuming, and require

considerable expertise and trained healthcare workers, particularly for identifying

species accurately at low parasitemia or in mixed malarial infections. The most important

shortcoming of microscopic examination is its relatively low sensitivity, particularly at low

parasite levels. Although the expert microscopist can detect up to 5 parasites, the average

microscopist detects only 50-100 parasites. This has probably resulted in underestimating

11
malaria infection rates, especially cases with low parasitemia and asymptomatic malaria. The

ability to maintain required levels of in malaria diagnostics expertise is problematic, especially in

remote medical centers in countries where the disease is rarely seen (Makler, 1999). Microscopy

is laborious and ill-suited for high-throughput use, and species determination at low parasite

density is still challenging. Therefore, in remote rural settings, e.g. peripheral medical clinics

with no electricity and no health-facility resources, microscopy is often

unavailable (Bon et al., 2015).

2.5.2 Serological tests

Diagnosis of malaria using serological methods is usually based on the detection of antibodies

against asexual blood stage malaria parasites. mmunofluorescence antibody testing (IFA) has

been a reliable serologic test for malaria in recent decades. Although IFA is time-consuming and

subjective, it is highly sensitive and specific (Bon et al., 2015).

- Literature clearly illustrates the reliability of IFA, so that it was usually regarded as the gold

standard for malarial serology testing. IFA is useful in epidemiological surveys, for screening

potential blood donors, and occasionally for providing evidence of recent infection in non-

immunes. Until recently, it was a validated method for detecting Plasmodium-specific antibodies

in various blood bank units, which was useful for screening prospective blood donors, so

avoiding transfusion-transmitted malaria. In France, for example, IFA is used as a part of a

targeted screening strategy, combined with a donor questionnaire. The principle of IFA is that,

following infection with any Plasmodium species, specific antibodies are produced within 2

weeks of initial infection, and persist for 3-6 months after parasite clearance. IFA uses specific

antigen or crude antigen prepared on a slide, coated and kept at -30 until used, and quantifies

both lgG and 1gM antibodies in patient serum samples. Titers>1 20 are usually deemed positive,

12
and < I : 20 unconfirmed. Titers>l: 200 can be classified as recent infections. In conclusion, IFA

is simple and sensitive, but time-consuming, It cannot be automated, which limits the number of

sera that can be studied daily. It also requires fluorescence microscopy and trained technicians;

readings can be influenced by the level of training of the technician, particularly for serum

samples with low antibody titers. Moreover, the lack of IFA reagent standardization makes it

impractical for routine use in blood-transfusion centers, and for harmonizing inter- laboratory

results (Phoebe, 2013).

2.5.3 Quality base channel (QBC) technique

The QBC technique was designed to enhance microscopic detection of parasites and simplify

malaria diagnosis. This method involves staining parasite deoxyribonucleic acid (DNA) in

micro-hematocrit tubes with fluorescent dyes, e.g. acridine orange, and its subsequent detection

by epi-fluorescent microscopy. Briefly, finger-prick blood is collected in a hematocrit tube

containing acridine orange and anticoagulant. The tube is centrifuged at 12,000g for 5 mm and

immediately examined using an epifluorescent microscopë Paraite nuclei fluoresces bright green,

while cytoplasm appears yellow-orange. The QBC technique has been shown to be a rapid and

sensitive test for diagnosing malaria in, numerous laborato settings. While it enhances sensitivity

for P. falciparum, it reduces sensitivity for non-falciparum species and decreases specificity due

to staining of leukocyte DNA. Recently, it has been shown that acridine orange is the preferred

diagnostic method (over light microscopy and immunochromatographic tests) in the context of

epidemiologic studies in asymptornatic populations in endemic areas, probably because of

increased sensitivity at low parasitemia. Nowadays, portable fluorescent microscopes using light

emitting diode (LED) technology, and pre-prepared glass slides with fluorescent reagent to label

parasites, are available commercially. Although the QBC technique is simple, reliable, and user-

13
friendly, it requires specialized instrumentation, is more costly than conventional light

microscopy, and is poor at determining species and numbers of parasites (Phoebe, 2013).

2.6 Molecular Diagnostic Methods

As mentioned in the preceding pages, traditional malaria diagnostic methods remain problematic.

New laboratory diagnostic techniques that display high sensitivity and high specificity, without

subjective variation, are urgently needed in various laboratories. Recent developments in

molecular biological technologies, e.g. PCR, loop-mediated isothermal amplification (LAMP),

microarray, mass spectrometry (MS), and flow cytometric (FCM) assay techniques, have

permitted extensive characterization of the malaria parasite and are generating new strategies for

malaria diagnosis (Silas, 2016).

2.6.1 PCR technique

PCR-based techniques are a recent development in the molecular diagnosis of malaria, and have

proven to be one of the most specific and sensitive diagnostic methods, particularly for malaria

cases with lowparasitemia or mixed infection. The PCR technique continues to be used

extensively to confirm malaria infection, followup therapeutic response, and identify drug

resistance. It was found to be more sensitive than QBC and some RDT(Lon et al., 2005).

Concerning with the gold standard method for malaria diagnosis, PCR has shown higher

sensitivity and specificity than conventional microscopic examination of stained peripheral blood

smears, and now seems the best method for malaria diagnosis. PCR can detect as few as 1-5

parasites/il of blood (0.0001% of infected red blood cells) compared with around 50-100

parasites/il of blood by microscopy or RDT. Moreover, PCR can help

14
detect drug-resistant parasites, mixed infections, and may be automated to process

large numbers of samples. Some modified PCR methods are proving reliable, e.g.,

nested PCR, real-time PCR, and reverse transcription PCR, and appear to be useful

second-line techniques when results of traditional diagnostic methods are unclear for

patients presenting with signs and symptoms of malaria; they also allow accurate

species determination. Recently, the PCR method has become widely accepted for

identifying P. knowlesi infections. Although PCR appears to have overcome the two

major problems of malaria diagnosis-sensitivity and specificity- the utility of PCR is

-frlimited by complex methodologies, high cost, and the need for specially trained technicians.

PCR, therefore, is not routinely implemented in developing countries because of the complexity

of the testing and the latk of resources to perform these tests adequately and routinely. Quality

control and equipment maintenance are also essential for the PCR technique, so that it may not

be suitable for malaria diagnosis in remote rural areas or even in routine clinical diagnostic

setting (Makler, 1999).

2.6.2 LAMP technique

The LAMP technique is claimed to be asimple and inexpensive molecular malariadiagnostic test

that detects the conserved 18 S.RNA gene of P. faiciparum. Other studies have shown high

sensitivity and specificity, not only for P. falciparuni, but also P. vivax, P. ovale and P.

malariae. These observations suggest that LAMP is more reliable and useful for routine

screening for malaria parasites in regions where vector-borne diseases, such as malaria, are

endemic. LAMP appears to be easy, sensitive, quick and lower in cost than PCR. However,

reagents require cold storage, and further clinical trials are needed to validate the feasibility and

clinical utility of LAMP (Lon et al., 2005).

15
2.6.3 Microarrays

Publication of the Plasmodium genome offers many maleria-diagnostic opportunities.

Microarrays may play an important role in the future diagnosis of infectious diseases. The

principle of the microarrays technique parallels traditional Southern hybridization. Hybridization

of labeled targets divided from nucleic acids in the test sample to probes on the array enables the

probing of multiple gene targets in a single experiment. ideally, this technique would be

miniaturized and automated for point-of- care diagnostics. A pan-microbial oligonucleotide

microarray has been developed for infectious disease diagnosis and has identified P. falciparum

accurately in clinical specimens. This diagnostic technique, however, .s still in the early stages of

development (Lon et al., 2005).

2.6.4 Automated blood cell counters (ACC)

An ACC is a practical tool for malaria diagnosis, with 3 reported approaches. The first used a

Cell-Dyn® 3500 apparatus to detect malaria pigment (hemozoin) in monocytes, and showed a

sensitivity of 95% and specificity of 88%, compared with the gold-standard blood smear. The

second method also used a Cell-Dyn® 3500, and analyzed depolarized laser light (DLL) to

detect malaria infection, with an overall sensitivity of 72% and specificity of 96% . The third

technique used a Beckman (Lon et al., 2005).

16
CHAPTER THREE

3.0 MATERIALS AND METHODS

3.1 Materials

The materials used in this research work were:

Microscope, rapid test kit, needle, Giemsa so1ution, ethylene diaminetetracetic acid

(EDTA sample bottles, Plainkhan tubes, 5mL syringes, Giemsa Stains, microscopic

Iides, RDT kits.

3.2 Sample Collection

Malaria surveys were undertaken in New Kuchingoro IDP Camp, 25

males and 25 females were randomly selected. Individual consents from the people

was obtained before sample collection. Respondents were asked to provide a finger prick blood

sample, which was used to Plasmodium spp. infection in the peripheral blood using RDTs. The

RDTs used in the surveys, depending on availability.

3.3 Rapid Diagnostic Test

Rapid diagnostic test was performed on 5 mcI of blood using Care Start TM Malaria HRP2

(Access Bio, Inc., Somerset, New Jersey, USA) according to manufacturer’s instructions. The

test was considered positive when the antigen line was visible in the test window and negative

when only the control band was visible. It has a sensitivity of 98% and specificity of 97.5% as

reported in the test kit pamphlet. The test is only limited to the detection of antigens of

P.faiciparum. The entire test kits were within the shelf life and were stored in the laboratory

store which is air-conditioned though not on a 24 Li basis. Hence, the temperature was variable.

Recommended storage temperature by the manufacturer is between 1°C and 40°C.

17
CHAPTER FOUR

4.0 RESULT

From Table 1 below ,the total number of male are 25 while female are 25 and from Table 2, The

percentage of positive result was 52% while the percentage of negative were 36% and 12% of

the result was invalid. The Table  shows that 26 of the 50 participant blood samples were

reactive for Malaria with no difference between males and females.

Table 1: Age and Sex Distribution

Sex   Number percentage


Male 25 32%  
Female 25 18%  
 

Table 2: Positive/ Negative figure


Variable   Number percentage
Positive 26 52%  
Negative 18 36%  
Invalid 6 12%  

18
CHAPTER FIVE

5.0 DISCUSSION

5.1 Discussion

There are four principal methods for diagnosing malaria. These are symptomatic, microscopy,

antigen test and molecular methods. Symptomatic diagnosis is the most common, and people in

poorer countries often use symptoms alone to diagnose malaria. In other areas, too, symptomatic

diagnosis is often the initial one, followed by one of the other methods. However, it should be

noted that many other diseases present symptoms very similar to malaria, and diagnosis by

symptoms alone can be misleading and even harmful. Treating for malaria where other treatment

is called for leaves the actual disease uncured and the patient in critical condition. It is therefore

imperative to follow up symptomatic diagnosis with one of the other more accurate methods.

Onset of long periodic fevers, chills and bodily pain are often taken together to be symptoms of

malaria. However, this diagnosis is often wrong; so at times is parasitemia, which means the

concentration of parasites in the blood; both can be caused by other sorts of infections. It has

been shown that retinopathy, the study of changes occurring in the retina of the eye, can give

good indication of malaria, because the color and other aspects of retinas were changed as a

result of particular diseases. A percentage parasitaemia need therefore be adopted to correlate

with clinical presentation.

Malaria is common despite interventions directed to mosquito vector control and treatment of

symptomatic cases. In areas where malaria is hyperendemic, such as the study area, a high

percentage of the population may harbor the Plasmodium parasite in their blood. A majority of

the tested migrant laborers were males in the age range of 15–29 and Amhara in ethnicity. Half

19
of them were coming from lowland areas. Eastern Sudan has a similar eco-epidemiology with

study area, and Sudanese come to the study area seeking temporary employment on large-scale

farms.

High prevalence of malaria (18.4%) was detected in the study area (hard to access population

group). The overall Plasmodium species proportion was found to be 13% P. falciparum, 1.8% P.

vivax, and 3.6% mixed infections. Results from this study revealed that number of visits to the

study area, outdoor sleeping habit, bed net utilization, home area, and educational status of

migrant laborers were significantly associated with risk of malaria (P<0.05).

This finding is in line with a previous study conducted in two districts of the North Gondar zone

(Metema and West Armachiho districts) with an overall malaria infection prevalence rate of 12%

and the species proportion of 9.6% P. falciparum, 1.7% P. vivax, and 0.7% mixed

infections.1 Migrant workers coming from highland and highland fringe areas and those with low

access to bed nets are affected by malaria infection. This similarity may be due to similar

geography of study areas except that the study period is different.

5.2 Conclusion

In the study it can be concluded that the rate of malaria is more in the IDP camps as more of 52%

of the cases were confirmed malaria Positive. Although, other bodily fluids like saliva or urine

can also be used as less invasive methods, blood is preferred for higher concentration of the

parasite.

20
5.3 Recommendations

The antibody based method as anticipated showed good level of sensitivity but, very unspecific.

Nigeria is a malaria endemic area, so antibodies against HRP-2 may be a common finding. In our

present work, it was 100% and if this is extrapolated to our larger society, it means that virtually

everyone that is febrile will test positive to the antibody method.

21
REFERENCE

Aikawa,W “Human cerebral malaria,” The American Journal of Tropical Medicine and Hygiene,
vol. 39, no. 1, pp. 3–10, 1988.

Bon, W. A. Ali, and F. Enqusellassie, “Self-treatment of malaria in rural communities, Butajira,


southern Ethiopia,” Bulletin of the World Health Organization, vol. 81, no. 4, pp. 261–
268, 2003.
Bray R. S. and R. E. Sinden, “The sequestration of Plasmodium falciparum infected
erythrocytes in the placenta,” Transactions of the Royal Society of Tropical Medicine and
Hygiene, vol. 73, no. 6, pp. 716–719, 1979.

John J.and P. Malaney, “The economic and social burden of malaria,” Nature, vol. 415, no.
6872, pp. 680–685, 2002.

Lon D, Go R, Miguel C, Walker J, Cacal L, Saul A.(2015). Diagnosis of malaria in a remote area
of the Philippines: comparison of techniques and their acceptance by health workers and
the community. Bull World Health Organ. 2001;79:933–941. 

Markler S. and J. G. Breman, (1999)“GAPS in the childhood malaria burden in Africa: cerebral
malaria, neurological sequelae, anemia, respiratory distress, hypoglycemia, and
complications of pregnancy,” The American Journal of Tropical Medicine and Hygiene,
vol. 64, no. 1-2, pp. 57–67, 2001.

Nadwani RW, Guerra CA, Noor AM, Myint HY, Hay SI. (2018) The global distribution of
clinical episodes of Plasmodium falciparum malaria. Nature. ;434:214–217.

Nadwani RW, Jorgensen P, Christophel EM, Palmer KL (2019) Malaria risk: estimation of the
malaria burden. Nature. 2005;437:E3. 

Nandwani S, Mathur M, Rawat S. Evaluation of the polymerase chain reaction analysis for
diagnosis of falciparum malaria in Delhi India. Indian J Med Microbiol. 2005;23(3):176–
178. 

Phoebe TK, (2012). Self-treatment of malaria in a rural area of western Kenya. Bull World
Health Organ.  73:229–236.

Silas G, Barnish G, Bates I, Iboro J (2016) Newer drug combinations for


malaria. BMJ. 2004;328:1511–1512. 

Tijani RB, (2014). Incidence and management of malaria in two communities of different
socioeconomic level, in Accra, Ghana. Ann Trop Med Parasitol.  94:771–778.

22
Tulu, N. A. “Malaria,” in The Ecology of Health and Disease in Ethiopia, H. Kloos and A. Z.
Zein, Eds., pp. 341–352, Westview Press, Boulder, Colo, USA, 2nd edition, 1993.

Wongsrichanalai P. and L. Hall, “Malaria on the move: human population movement and
malaria transmission,” Emerging Infectious Diseases, vol. 6, no. 2, pp. 28–45, 2000.

Yahaya TW, (2016) Clinical algorithms for malaria diagnosis lack utility among people of
different age groups. Trop Med Int Health.  10:530–536. 

Zhou, N. Minakawa, A. K. Githeko, and G. Yan, “Association between climate variability and
malaria epidemics in the East Africian highlands,” Proceedings of the National Academy
of Sciences of the United States of America, vol. 101, no. 8, pp. 2375–2380, 2004.

23

Вам также может понравиться